Vous êtes sur la page 1sur 188

haematologica

Journal of the European Hematology Association


Published by the Ferrata Storti Foundation

Ancient Greek The origin of a name that reflects Europe’s cultural roots.

aÂma [haima] = blood


a·matow [haimatos] = of blood
Scientific Latin
lÒgow [logos]= reasoning

Scientific Latin haematologicus (adjective) = related to blood

haematologica (adjective, plural and neuter,


Modern English used as a noun) = hematological subjects

The oldest hematology journal,


publishing the newest research results.
2016 JCR impact factor = 7.702

Haematologica, as the journal of the European Hematology Association (EHA), aims


not only to serve the scientific community, but also to promote European cultural
identify.
haematologica
Journal of the European Hematology Association
Published by the Ferrata Storti Foundation

Editor-in-Chief
Luca Malcovati (Pavia)

Managing Director
Antonio Majocchi (Pavia)

Associate Editors
Omar I. Abdel-Wahab (New York), Hélène Cavé (Paris), Simon Mendez-Ferrer (Cambridge), Pavan Reddy (Ann
Arbor), Andreas Rosenwald (Wuerzburg), Monika Engelhardt (Freiburg), Davide Rossi (Bellinzona), Jacob Rowe
(Haifa, Jerusalem), Wyndham Wilson (Bethesda), Paul Kyrle (Vienna), Swee Lay Thein (Bethesda), Pieter Sonneveld
(Rotterdam)

Assistant Editors
Anne Freckleton (English Editor), Cristiana Pascutto (Statistical Consultant), Rachel Stenner (English Editor),
Kate O’Donohoe (English Editor), Ziggy Kennell (English Editor)

Editorial Board
Jeremy Abramson (Boston); Paolo Arosio (Brescia); Raphael Bejar (San Diego); Erik Berntorp (Malmö); Dominique
Bonnet (London); Jean-Pierre Bourquin (Zurich); Suzanne Cannegieter (Leiden); Francisco Cervantes (Barcelona);
Nicholas Chiorazzi (Manhasset); Oliver Cornely (Köln); Michel Delforge (Leuven); Ruud Delwel (Rotterdam);
Meletios A. Dimopoulos (Athens); Inderjeet Dokal (London); Hervé Dombret (Paris); Peter Dreger (Hamburg); Martin
Dreyling (München); Kieron Dunleavy (Bethesda); Dimitar Efremov (Rome); Sabine Eichinger (Vienna); Jean Feuillard
(Limoges); Carlo Gambacorti-Passerini (Monza); Guillermo Garcia Manero (Houston); Christian Geisler
(Copenhagen); Piero Giordano (Leiden); Christian Gisselbrecht (Paris); Andreas Greinacher (Greifswals); Hildegard
Greinix (Vienna); Paolo Gresele (Perugia); Thomas M. Habermann (Rochester); Claudia Haferlach (München); Oliver
Hantschel (Lausanne); Christine Harrison (Southampton); Brian Huntly (Cambridge); Ulrich Jaeger (Vienna); Elaine
Jaffe (Bethesda); Arnon Kater (Amsterdam); Gregory Kato (Pittsburg); Christoph Klein (Munich); Steven Knapper
(Cardiff); Seiji Kojima (Nagoya); John Koreth (Boston); Robert Kralovics (Vienna); Ralf Küppers (Essen); Ola Landgren
(New York); Peter Lenting (Le Kremlin-Bicetre); Per Ljungman (Stockholm); Francesco Lo Coco (Rome); Henk M.
Lokhorst (Utrecht); John Mascarenhas (New York); Maria-Victoria Mateos (Salamanca); Giampaolo Merlini (Pavia);
Anna Rita Migliaccio (New York); Mohamad Mohty (Nantes); Martina Muckenthaler (Heidelberg); Ann Mullally
(Boston); Stephen Mulligan (Sydney); German Ott (Stuttgart); Jakob Passweg (Basel); Melanie Percy (Ireland); Rob
Pieters (Utrecht); Stefano Pileri (Milan); Miguel Piris (Madrid); Andreas Reiter (Mannheim); Jose-Maria Ribera
(Barcelona); Stefano Rivella (New York); Francesco Rodeghiero (Vicenza); Richard Rosenquist (Uppsala); Simon Rule
(Plymouth); Claudia Scholl (Heidelberg); Martin Schrappe (Kiel); Radek C. Skoda (Basel); Gérard Socié (Paris); Kostas
Stamatopoulos (Thessaloniki); David P. Steensma (Rochester); Martin H. Steinberg (Boston); Ali Taher (Beirut);
Evangelos Terpos (Athens); Takanori Teshima (Sapporo); Pieter Van Vlierberghe (Gent); Alessandro M. Vannucchi
(Firenze); George Vassiliou (Cambridge); Edo Vellenga (Groningen); Umberto Vitolo (Torino); Guenter Weiss
(Innsbruck).

Editorial Office
Simona Giri (Production & Marketing Manager), Lorella Ripari (Peer Review Manager), Paola Cariati (Senior Graphic
Designer), Igor Ebuli Poletti (Senior Graphic Designer), Marta Fossati (Peer Review), Diana Serena Ravera (Peer Review)

Affiliated Scientific Societies


SIE (Italian Society of Hematology, www.siematologia.it)
SIES (Italian Society of Experimental Hematology, www.siesonline.it)
haematologica
Journal of the European Hematology Association
Published by the Ferrata Storti Foundation

Information for readers, authors and subscribers


Haematologica (print edition, pISSN 0390-6078, eISSN 1592-8721) publishes peer-reviewed papers on all areas of experi-
mental and clinical hematology. The journal is owned by a non-profit organization, the Ferrata Storti Foundation, and
serves the scientific community following the recommendations of the World Association of Medical Editors
(www.wame.org) and the International Committee of Medical Journal Editors (www.icmje.org).

Haematologica publishes editorials, research articles, review articles, guideline articles and letters. Manuscripts should be
prepared according to our guidelines (www.haematologica.org/information-for-authors), and the Uniform Requirements
for Manuscripts Submitted to Biomedical Journals, prepared by the International Committee of Medical Journal Editors
(www.icmje.org).

Manuscripts should be submitted online at http://www.haematologica.org/.

Conflict of interests. According to the International Committee of Medical Journal Editors (http://www.icmje.org/#conflicts),
“Public trust in the peer review process and the credibility of published articles depend in part on how well conflict of
interest is handled during writing, peer review, and editorial decision making”. The ad hoc journal’s policy is reported in
detail online (www.haematologica.org/content/policies).

Transfer of Copyright and Permission to Reproduce Parts of Published Papers. Authors will grant copyright of their articles to the
Ferrata Storti Foundation. No formal permission will be required to reproduce parts (tables or illustrations) of published
papers, provided the source is quoted appropriately and reproduction has no commercial intent. Reproductions with com-
mercial intent will require written permission and payment of royalties.

Detailed information about subscriptions is available online at www.haematologica.org. Haematologica is an open access
journal. Access to the online journal is free. Use of the Haematologica App (available on the App Store and on Google
Play) is free.
For subscriptions to the printed issue of the journal, please contact: Haematologica Office, via Giuseppe Belli 4, 27100
Pavia, Italy (phone +39.0382.27129, fax +39.0382.394705, E-mail: info@haematologica.org).

Rates of the International edition for the year 2018 are as following:
Institutional Personal
Print edition Euro 600 Euro 150

Advertisements. Contact the Advertising Manager, Haematologica Office, via Giuseppe Belli 4, 27100 Pavia, Italy (phone
+39.0382.27129, fax +39.0382.394705, e-mail: marketing@haematologica.org).

Disclaimer. Whilst every effort is made by the publishers and the editorial board to see that no inaccurate or misleading
data, opinion or statement appears in this journal, they wish to make it clear that the data and opinions appearing in the
articles or advertisements herein are the responsibility of the contributor or advisor concerned. Accordingly, the publisher,
the editorial board and their respective employees, officers and agents accept no liability whatsoever for the consequences
of any inaccurate or misleading data, opinion or statement. Whilst all due care is taken to ensure that drug doses and other
quantities are presented accurately, readers are advised that new methods and techniques involving drug usage, and
described within this journal, should only be followed in conjunction with the drug manufacturer’s own published liter-
ature.

Direttore responsabile: Prof. Edoardo Ascari; Autorizzazione del Tribunale di Pavia n. 63 del 5 marzo 1955.
Printing: Press Up, zona Via Cassia Km 36, 300 Zona Ind.le Settevene - 01036 Nepi (VT)
haematologica
Journal of the European Hematology Association
Published by the Ferrata Storti Foundation

The 4th World Congress on Controversies in Multiple 23rd Congress of EHA


calendar of events

Myeloma June 14-17, 2018


Chairs: M Mohty, A Nagler, T Facon Stockholm, Sweden
May 3-5, 2018
Paris, France
EHA-SAH Hematology Tutorial on lymphoid Malignancies
and Plasma Cell Dyscrasias
EHA Hematology Tutorial on Thalassemia September 14-15, 2018
May 10-11, 2018 Buenos Aires, Argentina
Shiraz, Iran

EHA-SWG Scientific Meeting on Aging and Hematology


JACIE Inspector Training Course Chair: D Bron
EBMT / JACIE October 12-14, 2018
Chair: E McGrath Warsaw, Poland
May 24-25, 2018
Barcelona, Spain

35th International Congress of the ISBT


The International Society of Blood Transfusion (ISBT)
Chairs: K Pavenski, E van der Schoot, E Wood
June 2-6, 2018
Toronto, Canada

20th Congress of the European Society for


Haemapheresis
European Society for Haemapheresis
June 13-14, 2018
Valencia, Spain Calendar of Events updated on April 5, 2018
haematologica
Journal of the European Hematology Association
Table of Contents Published by the Ferrata Storti Foundation
Volume 103, Issue 5: May 2018
Cover Figure
Cluster of osteoblasts with eccentric round to oval nuclei and abundant blue, mottled cytoplasm. Courtesy of Prof. Rosangela Invernizzi.

Editorials
747 (Auto-)immune signature in aplastic anemia
Antonio M. Risitano

749 Hematopoietic stem cell mobilization with plerixafor in sickle cell disease
Matthew M. Hsieh and John F. Tisdale

751 Age-related clonal hematopoiesis and monoclonal B-cell lymphocytosis / chronic lymphocytic leukemia: a new association?
Adalgisa Condoluci and Davide Rossi

Perspectives
753 Denosumab for bone lesions in multiple myeloma – what is its value?
Daniel A. Goldstein

755 Osteoprotective medication in the era of novel agents: a European perspective on values, risks and future solutions
Monika Engelhardt

Articles
Bone Marrow Failure
759 Deep sequencing and flow cytometric characterization of expanded effector memory CD8+CD57+ T cells frequently reveals T-cell receptor
Vβ oligoclonality and CDR3 homology in acquired aplastic anemia
Valentina Giudice et al.

Red Cell Biology & its Disorders


770 Safety and efficacy of plerixafor dose escalation for the mobilization of CD34+ hematopoietic progenitor cells in patients with sickle cell
disease: interim results
Farid Boulad et al.

778 Plerixafor enables safe, rapid, efficient mobilization of hematopoietic stem cells in sickle cell disease patients after exchange transfusion
Chantal Lagresle-Peyrou et al.

787 RON kinase inhibition reduces renal endothelial injury in sickle cell disease mice
Alfia Khaibullina et al.

Myeloproliferative Disorders
798 The KIT and PDGFRA switch-control inhibitor DCC-2618 blocks growth and survival of multiple neoplastic cell types in advanced
mastocytosis
Mathias Schneeweiss et al.

Acute Myeloid Leukemia


810 Distinct protein signatures of acute myeloid leukemia bone marrow-derived stromal cells are prognostic for patient survival
Steven M. Kornblau et al.

822 Clinical relevance of IDH1/2 mutant allele burden during follow-up in acute myeloid leukemia. A study by the French ALFA group
Yann Ferret et al.

Haematologica 2018; vol. 103 no. 5 - May 2018


http://www.haematologica.org/
haematologica
Journal of the European Hematology Association
Published by the Ferrata Storti Foundation

Acute Lymphoblastic Leukemia


830 MSH6 haploinsufficiency at relapse contributes to the development of thiopurine resistance in pediatric B-lymphoblastic leukemia
Nikki A. Evensen et al.

Hodgkin Lymphoma
840 A phase II study of the oral JAK1/JAK2 inhibitor ruxolitinib in advanced relapsed/refractory Hodgkin lymphoma
Eric Van Den Neste et al.

Non-Hodgkin Lymphoma
849 A B-cell receptor-related gene signature predicts survival in mantle cell lymphoma: results from the Fondazione Italiana Linfomi
MCL-0208 trial
Riccardo Bomben et al.

857 Incidence and risk factors for relapses in HIV-associated non-Hodgkin lymphoma as observed in the German HIV-related lymphoma
cohort study
Philipp Schommer et al.

Chronic Lymphocytic Leukemia


865 Highly similar genomic landscapes in monoclonal B-cell lymphocytosis and ultra-stable chronic lymphocytic leukemia with low
frequency of driver mutations
Andreas Agathangelidis et al.

874 Toxicities and outcomes of 616 ibrutinib-treated patients in the United States: a real-world analysis
Anthony R. Mato et al.

Plasma Cell Disorders


880 A novel nano-immunoassay method for quantification of proteins from CD138-purified myeloma cells: biological and clinical utility
Irena Misiewicz-Krzeminska et al.

890 Impact of extramedullary disease in patients with newly diagnosed multiple myeloma undergoing autologous stem cell transplantation:
a study from the Chronic Malignancies Working Party of the EBMT
Nico Gagelmann et al.

Hemostasis
898 Immobilized fibrinogen activates human platelets through glycoprotein VI
Pierre H Mangin et al.

Coagulation & its Disorders


908 Circulating microRNAs as biomarkers of disease and typification of the atherothrombotic status in antiphospholipid syndrome
Carlos Pérez-Sánchez et al.

Letters to the Editor


Letters are available online only at www.haematologica.org/content/103/5.toc

e184 Iron overload in transfusion-dependent survivors of hemoglobin Bart’s hydrops fetalis.


Ali Amid et al.
http://www.haematologica.org/content/103/5/e184

Haematologica 2018; vol. 103 no. 5 - May 2018


http://www.haematologica.org/
haematologica
Journal of the European Hematology Association
Published by the Ferrata Storti Foundation

e188 A defined culture method enabling the establishment of ring sideroblasts from induced pluripotent cells of X-linked sideroblastic anemia
Shunsuke Hatta et al.
http://www.haematologica.org/content/103/5/e188

e192 The mutational landscape of 18 investigated genes clearly separates four subtypes of myelodysplastic/myeloproliferative neoplasms
Manja Meggendorfer et al.
http://www.haematologica.org/content/103/5/e192

e196 Clonal evolution in the transition from cutaneous disease to acute leukemia suggested by liquid biopsy in blastic plasmacytoid dendritic
cell neoplasm
Eleni Ladikou et al.
http://www.haematologica.org/content/103/5/e196

e200 Heterozygous carriers of germline c.657_661del5 founder mutation in NBN gene are at risk of central nervous system relapse of B-cell
precursor acute lymphoblastic leukemia
Bartłomiej Tomasik et al.
http://www.haematologica.org/content/103/5/e200

e204 Ibrutinib for chronic lymphocytic leukemia: international experience from a named patient program
Peter Hillmen et al.
http://www.haematologica.org/content/103/5/e204

e207 IGHV segment utilization in immunoglobulin gene rearrangement differentiates patients with anti-myelin-associated glycoprotein neu-
ropathy from others immunoglobulin M-gammopathies
Jean-Sebastien Allain et al.
http://www.haematologica.org/content/103/5/e207

e211 Outcomes of patients with relapsed aggressive adult T-cell leukemia-lymphoma: clinical effectiveness of anti-CCR4 antibody and allogene-
ic hematopoietic stem cell transplantation
Shigeo Fuji et al.
http://www.haematologica.org/content/103/5/e211

e215 Sequential loss of tumor surface antigens following chimeric antigen receptor T-cell therapies in diffuse large B-cell lymphoma
Haneen Shalabi et al.
http://www.haematologica.org/content/103/5/e215

Case Reports
Case Reports are available online only at www.haematologica.org/content/103/5.toc

e219 Novel hereditary spherocytosis-associated splice site mutation in the ANK1 gene caused by parental gonosomal mosaicism
Xiong Wang, et al.
http://www.haematologica.org/content/103/5/e219

e223 Severe hemolysis and transfusion reactions after treatment with BGB-3111 and PD-1 antibody for Waldenström macroglobulinemia
Jad Othman et al.
http://www.haematologica.org/content/103/5/e223

e226 Usefulness of initial plasma dabigatran concentration to predict rebound after reversal
Nicolas Gendron et al.
http://www.haematologica.org/content/103/5/e226

Haematologica 2018; vol. 103 no. 5 - May 2018


http://www.haematologica.org/
EDITORIALS
(Auto-)immune signature in aplastic anemia
Antonio M. Risitano
Hematology, Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy

E-mail: amrisita@unina.it
doi:10.3324/haematol.2018.190884

A
cquired idiopathic aplastic anemia (IAA) is a rare only limited data are available about the characterization of
hematologic disorder characterized by the failure of specific functional CD8+ T-cell subsets. On the other hand,
hematopoiesis secondary to an immune-mediated novel techniques of deep DNA sequencing have become
damage of the bone marrow. IAA is bona fide considered an available, and their application in IAA has led to the descrip-
auto-immune disease with a T-cell-mediated pathophysiolo- tion of somatic mutations in myeloid cells,15 with the subse-
gy.1 In this issue of the Journal, Giudice et al.2 describe the quent ongoing debate about their actual meaning,16 but also
oligoclonal pattern of effector memory CD8+ CD57+ T cells in to high throughput TCR analysis.17 In their study, Giudice et
IAA patients by using a combined deep sequencing and flow al. specifically investigated the compartment of effector
cytometry approach. Indeed, Giudice et al. show that clonally memory T cells (TEM) in IAA patients, looking for possible
expanded T-cell populations are frequently detectable even clonality as assessed by flow cytometry analysis of Vβ usage
within the effector memory compartment, and that they tend and sequencing of the hypervariable complementary deter-
to correlate with disease activity. Thus, the characterization of mining region 3 (CDR3) of the TCR.
the T-cell receptor (TCR) repertoire by high-resolution tech- In agreement with previous reports,1,10 Giudice et al. con-
niques may play a role in confirming the diagnosis of firm that AA patients often exhibit a skewed usage of Vβ
immune-mediated IAA and to monitor affected patients dur- families, usually within the CD8+ T-cell compartment; these
ing their disease course. oligoclonal expansions are more frequent in patients with
There is widespread clinical and experimental evidence to increased percentage of TEM (as defined by co-expression of
support the autoimmune pathophysiology of IAA.1 The CD8 and CD57), which are found in approximately 70% of
most striking is that patients with IAA may respond to T- AA patients. These gross abnormalities of the TCR Vβ usage
cell-targeted immunosuppressive therapies (IST), with rates were dissected at the clonal level by deep sequencing of the
of hematologic responses ranging between 50% and 70%.3 TCR, which allows the comparison of more than 107 reads
Almost four decades of investigations have provided us with corresponding to TCRs harbored by individual T cells.
a plethora of experimental data corroborating the hypothesis Indeed, clonal expansions were identified by repetitive use
of an immune-mediated pathophysiology. Increased circu- of TCR β variable (TRBV) and joint (TRJV) genes (i.e. redun-
lating activated T cells were described in IAA patients in the dant TRBV/TRBJ combinations), as well as by CDR3 size
‘80s.4 These T cells may suppress hematopoiesis through and DJ length profiles. Immunodominant clones within dif-
the secretion of different inflammatory cytokines5,6 and/or ferent T-cell subsets were invariably detected in all AA
via cell-mediated direct killing. Among the different inhibito- patients with increased CD8+ CD57+ TEM; however, the actu-
ry cytokines, interferon-γ (IFN-γ) plays a major role in sup- al magnitude of these clonal expansions was extremely het-
pressing human hematopoietic stem cells (HSC) in vivo, as erogeneous in the different T-cell subsets. Indeed, these
suggested by in vitro inhibition of cell cycle progression and clones remain minimally expanded (approx. 3%) within the
induction of apoptosis of hematopoietic progenitors.7 More CD4+ compartment, while they become largely dominant
recently, it has been suggested that IFN-γ may also exert its (approx. 18%) in the CD8+ compartment; the expansion
inhibitory effect on HSC impairing the homeostatic survival appears even larger when the CD8+ CD57+ TEM is analyzed.
signal delivered by thrombopoietin through its cognate This difference in behavior of TCR heterogeneity in differ-
receptor c-MPL.8 This inhibitory milieu is generated by ent T-cell subsets of AA patients was further confirmed by
immune cells, and mostly by T cells that become activated analysis of Simpson’s diversity score, which shows how
and proliferate in response to an antigen-driven stimulation. TCR diversity decreased from CD4+ to CD8+ T cells, and
While the search for these putative antigens has remained even more from both CD4+ and CD8+ T cells to CD8+ CD57+
unsuccessful, the demonstration of clonal expansion of T-cell TEM. In summary, the work performed by Giudice et al. con-
populations identified by their TCR has been considered firms that clonal CD8+ T-cell expansions are common in AA
robust proof of a T-cell-mediated pathophysiology in IAA.9,10 patients, in agreement with the well-established T-cell-medi-
Our growing understanding of the immune system and ated immune pathophysiology of AA.1 But for the first time,
the availability of powerful novel techniques has nurtured here the Authors provide evidence that the clonal expan-
continuous research in the field of IAA. On the one hand, sions are not limited to the CD8+ effector T cells, since they
investigators have tried to further dissect the abnormalities can be found even in the TEM compartment. Very interesting-
of the immune system in patients suffering from IAA. ly, with the caveat of a limited sample size, Giudice et al.
Indeed, looking at specific functional T-cell subsets, recur- show that abnormalities of the TEM compartment (i.e.
rent immune derangements have been found, such as increased TEM, with possible clonal expansions) may be asso-
increased T-helper type 17 cells (Th17)11 and reduced regula- ciated with a dismal outcome in AA, due to refractory or
tory T cells (Treg).12,13 However, irrespective of the deep phe- relapsed disease. This is further supported by the observa-
notyping of CD4+ T cells [i.e. by multiparameter mass tion of concordance between the expansion of the immun-
cytometry, termed cytometry by time-of-flight (CyTOF)],14 odominant CD8+ CD57+ TEM clone and disease activity (sim-

haematologica | 2018; 103(5) 747


Editorials

ilar to what has already been shown for expansion within are increased in circulating CD8+ T cells of AA patients.20
the bulk CD8+ compartment10). However, while these observations depict the broad
Memory is the hallmark of adaptive immunity; indeed, immune derangement which is expected in an autoim-
antigen-driven clonal expansion of effector T cells may mune disease such as AA, they do not provide any clues
generate antigen-specific lymphocytes that may persist about the pathogenic role of these cells. In contrast, the
life long (reviewed by Sallusto et al.18). These cells, which demonstration of clonality within the TEM compartment
are known as memory cells, confer immediate and effec- shown by Giudice et al. seems pathogenically more rele-
tive protection against pathogens upon antigen rechal- vant, according to the scenario depicted in Figure 1.
lenging; indeed, memory immune cells are selected for Indeed, a clonal immune response specific for (or cross-
their higher affinity for cognate antigens, and rapidly acti- reactive with) HSC can be elicited by unknown antigens
vate from their resting state after antigen stimulation.18 or triggers, eventually causing some impairment of
Among T cells, three subtypes of memory cells have been hematopoiesis. If these clonal TEFF do not undergo apop-
described:18 i) TEM, that carry the protective memory tosis (as usually occurrs in a physiological immune
because of their ability to deliver effector functions; ii) response), they may lead to overt AA.1 Given the plas-
central memory T cells (TCM), that home to secondary ticity of T cells, some of these clonal, antigen-specific
lymphoid organs and exert reactive memory through cells may eventually acquire a TEM or a TCM functional
their cross-differentiation toward TEM and effector T cells phenotype, generating some skewing within the broad
(TEFF); and iii) memory stem T cells (TSCM), which have TEM repertoire, as found by Giudice et al.2 The presence of
been described as a less differentiated subset that has bet- these clonal (possibly HSC-specific) TEM would account
ter self-renewal and the ability to differentiate into dis- for continuous damage to the hematopoiesis, since in the
tinct subsets of memory T cells. Only limited data are presence of persistent antigen spread they may serve as
available about memory T cells in AA. In 2009, Hu et al. a reservoir for newly-generated TEFF. Thus, irrespective of
reported that TEM and TEMRA (a further subset of terminally the regulatory role postulated by Giudice et al.,2 the pres-
differentiated TEM characterized by CD45RA expression ence of clonal TEM would represent the signature of a
and by better effector function) are increased in CD4+ and deeper immune derangement, possibly associated with a
CD8+ T-cell subsets in AA patients, while naïve T cells dismal clinical outcome.
are decreased.19 More recently, the US National In conclusion, the availability of high-throughput tech-
Institutes of Health group has described that even TSCM nologies is providing biomarkers which anticipate future

Figure 1. T-cell clonality in aplastic anemia. Unknown antigens and triggers may elicit a clonal immune response specific for (or cross-reactive with) hematopoietic
stem cells (HSC). These clonal, activated T cells tend to expand delivering their immune damage over hematopoiesis. At the same time, some of these clonally
expanded activated T cells may acquire an effector memory T cell (TEM) functional phenotype, leading to skewing of the TEM cell repertoire. While activated effector T
cells (TEFF) may undergo anergy or apoptosis (even as a consequence of immunosuppressive therapies), TEM represent a continuous reservoir for HSC-specific T cells,
which may exert their effector function upon rechallenging with the antigen. (This is very likely, given the typical antigen spread seen in autoimmune diseases.) Thus,
the presence of clonal TEM, which likely share the same antigen-specificity with large TEFF clones found in bulk CD8+ populations, represent a biomarker of a deep-root-
ed immune derangement, possibly associated with a dismal disease course.

748 haematologica | 2018; 103(5)


Editorials

applications in the management of AA patients. Indeed, Intracellular interferon-gamma in circulating and marrow T cells
detected by flow cytometry and the response to immunosuppressive
deep whole exome sequencing,15 CyTOF14 and deep TCR therapy in patients with aplastic anemia. Blood. 2002;100(4):1185-
analysis2 all help to better describe the pathogenic events 1191.
underlying bone marrow failure syndromes. Even if none 7. Selleri C, Maciejewski JP, Sato T, Young NS. Interferon-gamma con-
of them translates into immediate therapeutic decisions, stitutively expressed in the stromal microenvironment of human
marrow cultures mediates potent hematopoietic inhibition. Blood:
they are all useful to confirm the diagnosis, to determine 1996;87(10):4149-4157.
the prognosis and possibly to monitor the clinical course 8. Alvarado LJ, Andreoni A, Huntsman HD, et al. Heterodimerization
of AA patients. Indeed, this latter application may be use- of TPO and IFNγ Impairs Human Hematopoietic Stem/Progenitor
Cell Signaling and Survival in Chronic Inflammation Blood.
ful for early identification of refractory or relapsing 2017;130(Suppl 1):4.
patients, paving the way for pre-emptive therapeutic 9. Zeng W, Nakao S, Takamatsu H, et al. Characterization of T-cell
interventions. Moreover, the deep dissection at the clonal repertoire of the bone marrow in immune-mediated aplastic anemia:
evidence for the involvement of antigen-driven T-cell response in
and at the functional levels of the immune T-cell compart- cyclosporine-dependent aplastic anemia. Blood. 1999;93(9):3008-
ment (e.g. combining CyTOF and TCR analysis) may also 3016.
answer some open questions in the field. For example, 10. Risitano AM, Maciejewski JP, Green S, Plasilova M, Zeng W, Young
the differential depletion of some specific T-cell subsets NS. In-vivo dominant immune responses in aplastic anaemia: molec-
ular tracking of putatively pathogenetic T-cell clones by TCR beta-
might explain the different outcome seen with different CDR3 sequencing. Lancet. 2004;364(9431):355-364.
ATG preparations.3 These novel technologies may help 11. de Latour RP, Visconte V, Takaku T, et al. Th17 immune responses
identify the specific T-cell subsets which are crucial to the contribute to the pathophysiology of aplastic anemia. Blood.
2010;116(20):4175-4184.
pathophysiology of AA (and possibly differentially 12. Solomou EE, Rezvani K, Mielke S, et al. Deficient CD4+ CD25+
depleted by distinct ATG brands), possibly driving the FOXP3+ T regulatory cells in acquired aplastic anemia. Blood.
development of future targeted therapies. 2007;110(5):1603-1606.
13. Kordasti S, Marsh J, Al-Khan S, et al. Functional characterization of
CD4+ T cells in aplastic anemia. Blood. 2012;119(9):2033-2043.
14. Kordasti S, Costantini B, Seidl T, et al. Deep phenotyping of Tregs
References identifies an immune signature for idiopathic aplastic anemia and
predicts response to treatment. Blood. 2016;128(9):1193-1205.
1. Young NS. Current concepts in the pathophysiology and treatment 15. Yoshizato T, Dumitriu B, Hosokawa K, et al. Somatic Mutations and
of aplastic anemia. Hematology Am Soc Hematol Educ Program. Clonal Hematopoiesis in Aplastic Anemia. N Engl J Med.
2013;2013:76-81. 2015;373(1):35-47.
2. Giudice V, Feng X, Lin Z, et al. Deep sequencing and flow cytometric 16. Cooper JN, Young NS. Clonality in context: hematopoietic clones in
characterization of expanded effector memory CD8+CD57+ T cells their marrow environment. Blood. 2017;130(22):2363-2372.
frequently reveals T-cell receptor Vbeta oligoclonality and CDR3 17. Calis JJ, Rosenberg BR. Characterizing immune repertoires by high
homology in acquired aplastic anemia. Haematologica. 2018;103(5): throughput sequencing: strategies and applications. Trends
759-769. Immunol. 2014;35(12):581-590.
3. Scheinberg P, Nunez O, Weinstein B, et al. Horse versus rabbit 18. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector
antithymocyte globulin in acquired aplastic anemia. N Engl J Med. memory T cell subsets: function, generation, and maintenance. Annu
2011;365(5):430-438. Rev Immunol. 2004;22:745-763.
4. Zoumbos NC, Gascón P, Djeu JY, Trost SR, Young NS. Circulating 19. Hu X, Gu Y, Wang Y, Cong Y, Qu X, Xu C. Increased CD4+ and
activated suppressor T lymphocytes in aplastic anemia. N Engl J CD8+ effector memory T cells in patients with aplastic anemia.
Med. 1985;312(5):257-265. Haematologica. 2009;94(3):428-429.
5. Zoumbos NC, Gascón P, Djeu JY, Young NS. Interferon is a mediator 20. Hosokawa K, Muranski P, Fenx X, et al. Memory Stem T Cells in
of hematopoietic suppression in aplastic anemia in vitro and possi- Autoimmune Disease: High Frequency of Circulating CD8+
bly in vivo. Proc Natl Acad Sci USA. 1985;82(1):188-192. Memory Stem Cells in Acquired Aplastic Anemia. J Immunol.
6. Sloand E, Kim S, Maciejewski JP, Tisdale J, Follmann D, Young NS. 2016;196(4):1568-1578.

Hematopoietic stem cell mobilization with plerixafor in sickle cell disease


Matthew M. Hsieh and John F. Tisdale
Cellular and Molecular Therapeutics Section, Sickle Cell Branch, National Heart, Lung, and Blood Institute (NHLBI), National
Institutes of Health (NIH), Bethesda, Maryland

E-mail: matthewhs@nhlbi.nih.gov
doi:10.3324/haematol.2018.190876

A
After more than a half-century since the molecular apheresis is the standard method for HSC collection in
basis for sickle cell disease (SCD) was described by healthy adult donors, yet this approach is associated with
Linus Pauling and colleagues, we now possess the high rates of adverse events requiring hospitalization in
molecular tools to contemplate a one-time cure through SCD, including vaso-occlusive crises, multi-organ failure,
genetic modification of autologous hematopoietic stem and even death, prompting our call for a moratorium on its
cells (HSC). For these promising gene transfer and gene use for HSC mobilization in SCD.1 Thus, bone marrow
editing strategies to become a reality, a sufficient number harvesting is the default approach, with evidence support-
of HSC of high purity must be obtained. Filgrastim, or ing its utility in both animal models and in vitro studies uti-
granulocyte colony-stimulating factor, mobilization and lizing patients’ material.2-4 However, bone marrow harvest-

haematologica | 2018; 103(5) 749


Editorials

ing employed in an ongoing HSC gene therapy trial was lier reports. We will need more patients to ascertain the
recently recognized to result in suboptimal yields of high contribution of leukocytosis alone and/or the duration of
purity HSC at the end of collection and processing, along leukocytosis in developing sickle-related complications.
with substantial pain after each harvest, and most subjects Thirdly, only three patients underwent leukapheresis and
required two or three harvests to yield sufficient cell doses though adverse events appear acceptable, expanded
for manufacturing.5,6 accrual could capture additional side effects. Furthermore,
In this issue of the Journal, two groups of investigators if patients with SCD do not meet the goal and need addi-
report their results using a third approach to HSC collec- tional mobilization and collection, there could be cumu-
tion in SCD through mobilization with an inhibitor of the lative side effects. Finally, the peak of mobilization of
CXCR4 chemokine receptor, plerixafor. Boulad et al. per- CD34+ cells appeared to be much earlier, at 3-6 hours.
formed a dose escalation study of plerixafor among a This observation is distinct from that in healthy donors,
total of 15 SCD patients at steady state.7 Ten of the in whom the peak is observed at 6-12 hours.9 Perhaps the
patients were receiving concomitant treatment with chronically hyperproliferative marrow in SCD partly
hydroxyurea. Only a minority of patients in each cohort explains this early release of HSC; there could be other
achieved the target of ≥30 CD34+ cells/μL at 12 h after the factors at play. Regardless, this observation suggests that
plerixafor injection: three out six at a dose of 80 μg/kg, for optimal collection, apheresis should be started within
one out of three at a dose of 160 μg/kg, and two out of six 4-6 hours of dosing.
at a dose of 240 μg/kg. Two patients (15%) experienced a As clinical applications of gene transfer and gene editing
vaso-occlusive crisis during the study period – one each at strategies are being implemented in SCD, obtaining ade-
80 and 240 μg/kg. None of the patients underwent leuka- quate numbers of HSC safely from patients could be the
pheresis, thus attribution of these adverse events could be ‘bottleneck’, preventing broad dissemination of these
narrowed to plerixafor. On the other hand, Lagresle- exciting approaches. The early results provide optimism
Peyrou et al. reported the outcomes of three patients who that mobilization with plerixafor could be a safer and more
received plerixafor at a dose of 240 μg/kg.8 All three efficacious alternative for HSC collection to either filgras-
patients received at least 2 months of red cell exchange tim mobilization or bone marrow harvesting, and provide
transfusion to target a sickle hemoglobin (HbS) near 30% general confidence for the further development of these
while hydroxyurea was discontinued. The peak CD34+ promising approaches to a one-time cure for SCD.
cell count reached >75 cells/μL at as early as 3 h after the
injection. All three patients also underwent leukapheresis ©2017 NIH (National Institutes of Health)
of 15 to 21 L, with a resulting total CD34+ cell yield of 4.5
to 5.8 x 106 cells/kg and a purity of 80% to 95%. No pain,
vaso-occlusive crises, or sickle-related events were References
observed in these three patients.
1. Fitzhugh CD, Hsieh MM, Bolan CD, Saenz C, Tisdale JF. Granulocyte
While the number of patients is relatively small in both colony-stimulating factor (G-CSF) administration in individuals with
studies, important lessons relevant to autologous HSC sickle cell disease: time for a moratorium? Cytotherapy.
mobilization and collection in SCD with plerixafor can be 2009;11(4):464-471.
2. Hematti P, Tuchman S, Larochelle A, Metzger ME, Donahue RE,
gleaned. The first lesson regards preparation of the Tisdale JF. Comparison of retroviral transduction efficiency in CD34+
patients. Specifically, stopping hydroxyurea and utilizing cells derived from bone marrow versus G-CSF-mobilized or G-CSF
red cell transfusions, simple or exchange, to target a HbS plus stem cell factor-mobilized peripheral blood in nonhuman pri-
of 30% were likely key factors in the successful mobiliza- mates. Stem Cells. 2004;22(6):1062-1069.
3. Uchida N, Fujita A, Hsieh MM, et al. Bone marrow as a hematopoietic
tion of the series reported by Lagresle-Peyrou et al. stem cell source for gene therapy in sickle cell disease: evidence from
Conversely, the absence of these measures in the study Rhesus and SCD patients. Hum Gene Ther Clin Dev. 2017;28(3):136-
by Boulad et al. may explain why the majority of their 144.
4. Uchida N, Bonifacino A, Krouse AE, et al. Accelerated lymphocyte
patients failed to reach the target CD34+ concentration. reconstitution and long-term recovery after transplantation of lentivi-
This is consistent with prior work demonstrating a lower ral-transduced rhesus CD34+ cells mobilized by G-CSF and plerixafor.
CD34+ cell content in the marrow of SCD patients on Exp Hematol. 2011;39(7):795-805.
hydroxyurea when compared to those not on the drug.3 5. Kanter J, Walters MC, Hsieh MM, et al. Interim results from a phase I/II
clinical study of lenitoglobin gene therapy for severe sickle cell disease.
Discontinuation of hydroxyurea combined with sched- Blood. 2016;128(22):1176.
uled red cell transfusion to keep the HbS near 30% may 6. Leonard A, Bonifacino A, Dominical VM, et al. Bone marrow charac-
also have improved purity, which was 80% to 95% in the terization in sickle cell disease: inflammation and stress erythropoiesis
lead to suboptimal CD34 recovery compared to normal volunteer bone
study by Lagresle-Peyrou et al., while helping to mini- marrow. Blood. 2017;130(Suppl 1):966.
mize the risk of sickle cell-related adverse events while 7. Boulad F, Shore T, van Besien K, et al. Safety and efficacy of plerixafor
hydroxyurea treatment was interrupted. Secondly, leuko- dose escalation for the mobilization of CD34+ hematopoietic progen-
itor cells in patients with sickle cell disease: interim results.
cyte and neutrophil counts increased 2- to 3-fold just Haematologica. 2018;103(5):770-777.
hours after a single injection of plerixafor, even at the
lowest dose of 80 μg/kg tested. Although increases of a
8. Lagresle-Peyrou C, Lefrere F, Magrin E, et al. Plerixafor enables the safe,
rapid, efficient mobilization of haematopoietic stem cells in sickle cell
similar magnitude also occurred with filgrastim, the disease patients after exchange transfusion. Haematologica.
2018;103(5):778-786.
adverse events seen with filgrastim may have been relat- 9. Pantin J, Purev E, Tian X, et al. Effect of high-dose plerixafor on
ed to the prolonged duration of 5 to 6 days from filgras- CD34(+) cell mobilization in healthy stem cell donors: results of a ran-
tim that led to the high rates of adverse events in the ear- domized crossover trial. Haematologica. 2017;102(3):600-609.

750 haematologica | 2018; 103(5)


Editorials

Age-related clonal hematopoiesis and monoclonal B-cell lymphocytosis / chronic


lymphocytic leukemia: a new association?
Adalgisa Condoluci and Davide Rossi
Hematology, Institute of Oncology Research, and Oncology Institute of Southern Switzerland, Bellinzona, Switzerland

E-mail: davide.rossi@eoc.ch
doi:10.3324/haematol.2018.191098

A
gathangelidis et al.1 compared the mutational land- hematopoietic precursor that was able to participate in
scape of low-count monoclonal B-cell lymphocyto- both lymphoid and myeloid differentiation. By docu-
sis (MBL), high-count MBL and highly stable chron- menting that the DNA from PMN was free from contam-
ic lymphocytic leukemia (CLL) with confirmed lack of pro- ination by MBL/CLL DNA, for example, by using molec-
gression after a very long follow up (>10 years). The ular minimal residual disease methods relying on the
Authors also studied the polymorphonuclear (PMN) frac- individual patient’s specific immunoglobulin gene
tion and germline DNA from buccal swabs of the same rearrangements, the Authors have provided further evi-
individuals. Whole genome sequencing was complement- dence of this important finding which, although previ-
ed with deep sequencing of targeted genes. ously reported,5,6 had remained a subject of debate.
The sample size, along with the low coverage imposed Several hematologic malignancies, including CLL, mul-
by whole genome sequencing, are both limitations in tiple myeloma (MM) and acute myeloid leukemia (AML),
efforts to discover yet unknown variants that might actu- have well-defined precursor states that precede the devel-
ally be recurrent in these conditions. While opment of overt cancer. CLL is always preceded by a high
Agathangelidis et al. acknowledge these limitations, three MBL count,7 MM is almost always preceded by mono-
major findings characterize their manuscript.1 First, low- clonal gammopathy of undetermined significance
count MBL, high-count MBL and highly stable CLL share (MGUS),8 and at least a quarter of all patients with
a similar genetic landscape and mutational signatures, myelodysplastic syndromes (MDS) have disease that
which include the presence of mutations in known driv- evolves into AML.9 Deep genomic sequencing of normal
ers associated with poor outcome, such as NOTCH1, subjects revealed that during human aging, the expansion
SF3B1, POT1,2-4 indicating that these mutations are not of 1 or more hematopoietic stem and progenitor cells
sufficient to drive the aggressiveness of the disease by (HSPC) will result in clones that will sustainably con-
themselves. Second, the mutational landscape of paired tribute more than others to the production of mature
PMN suggests that most of these patients carry a clonal blood cells. Accordingly, age-related clonal hematopoiesis
hematopoiesis that could possibly be age-related. Third, (ARCH) is defined as the expansion of HSPC clones, har-
a number of somatic mutations were found in both the boring specific, disruptive, and recurrent genetic variants,
MBL/CLL cells and PMN, supporting the idea that the in individuals without clear diagnosis of hematologic
MBL/CLL clone stemmed from a common ancestral malignancies.10 MDS are frequently preceded by ARCH.11

Figure 1. Hypothetical model of evolution from age-related clonal hematopoiesis to monoclonal B-cell lymphocytosis/chronic lymphocytic leukemia.

haematologica | 2018; 103(5) 751


Editorials

Some ARCH-related mutations can increase the risk for mutations. Haematologica. 2018;103(5):865-873.
2. Rossi D, Rasi S, Fabbri G, et al. Mutations of NOTCH1 are an inde-
leukemia,12 while others possibly increase the risk for pendent predictor of survival in chronic lymphocytic leukemia.
heart disease and diabetes.13 From a pathogenetic stand- Blood. 2012;119(2):521-529.
point, the study of Agathangelidis et al.1 provides the 3. Wang L, Lawrence MS, Wan Y, et al. SF3B1 and other novel cancer
genes in chronic lymphocytic leukemia. N Engl J Med.
proof of principle that ARCH may also associate with 2011;365(26):2497-2506.
expansion of B-cell clones with CLL phenotype, and con- 4. Herling CD, Klaumünzer M, Rocha CK, et al. Complex karyotypes
nects ARCH with MBL and CLL in a continuum of evolu- and KRAS and POT1 mutations impact outcome in CLL after chlo-
tion from HSCP clones to mature B-cell clones (Figure 1), rambucil-based chemotherapy or chemoimmunotherapy. Blood.
2016;128(3):395-404.
thus validating in vivo in patients the notion initially 5. Damm F, Mylonas E, Cosson A, et al. Acquired initiating mutations
reported from mice studies that the propensity to gener- in early hematopoietic cells of CLL patients. Cancer Discov.
ate clonal B cells has already been acquired at the HSCP 2014;4(9):1088-1101.
6. Quijada-Álamo M, Hernández-Sánchez M, Robledo C, et al. Next-
stage.14 To robustly establish this association and to gain generation sequencing and FISH studies reveal the appearance of
greater insight into the pathogenetics, larger cohorts of gene mutations and chromosomal abnormalities in hematopoietic
MBL and CLL patients should be investigated with the progenitors in chronic lymphocytic leukemia. J Hematol Oncol.
2017;10(1):83.
rigorous approach utilized by Agathangelidis et al.1 7. Landgren O, Albitar M, Ma W, et al. B-cell clones as early markers
One of the long-term complications of chemoim- for chronic lymphocytic leukemia. N Engl J Med. 2009;360(7):659-
munotherapy in CLL is the development of treatment- 667.
related MDS/AML.15 Chemoimmunotherapy poses a 8. Landgren O, Kyle RA, Pfeiffer RM, et al. Monoclonal gammopathy
of undetermined significance (MGUS) consistently precedes multiple
strong selection bottleneck to HSCPs, and thus only the myeloma: a prospective study. Blood. 2009;113(22):5412-5417.
fittest HSCPs survive and repopulate after the stress of 9. Pfeilstöcker M, Tuechler H, Sanz G, et al. Time-dependent changes
chemoimmunotherapy.16 HSCP fitness may be sustained in mortality and transformation risk in MDS. Blood.
2016;128(7):902-910.
by somatic mutations in the context of a preceding 10. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal
ARCH, and it is increasingly recognized as a risk factor hematopoiesis associated with adverse outcomes. N Engl J Med.
for therapy-related MDS/AML.17 Among elderly patients 2014;371(26):2488-2498.
who receive chemotherapy and develop therapy-related 11. Malcovati L, Gallì A, Travaglino E, et al. Clinical significance of
somatic mutation in unexplained blood cytopenia. Blood.
MDS/AML, most have ARCH before chemotherapy. 2017;129(25):3371-3378.
Consistently, ARCH associates with an increased rate of 12. Genovese G, K¨ahler AK, Handsaker RE, et al. Clonal hematopoiesis
therapy-related AML/MDS.17 Following this line of evi- and blood-cancer risk inferred from blood DNA sequence. N Engl J
Med. 2014;371(26):2477-2487.
dence, the study by Agathangelidis et al.1 prompts inves- 13. Jaiswal S, Natarajan P, Silver AJ, et al. Clonal hematopoiesis and risk
tigation into whether the finding of an ARCH in CLL of atherosclerotic cardiovascular disease. N Engl J Med.
patients who receive chemoimmunotherapy is a risk fac- 2017;377(2):111-121.
14. Kikushige Y, Ishikawa F, Miyamoto T, et al. Self-renewing
tor for the development of therapy-related MDS/AML. If hematopoietic stem cell is the primary target in pathogenesis of
this is proved to be the case, given the availability of human chronic lymphocytic leukemia. Cancer Cell. 2011;20(2):246-
novel agents for the treatment of CLL that are not stress- 259.
ful for HSCP, ARCH might become a new biomarker for 15. Benjamini O, Jain P, Trinh L, et al. Second cancers in patients with
chronic lymphocytic leukemia who received frontline fludarabine,
tailoring treatment in CLL. cyclophosphamide and rituximab therapy: distribution and clinical
outcomes. Leuk Lymphoma. 2015;56(6):1643-1650.
16. Wong TN, Ramsingh G, Young AL, et al. Role of TP53 mutations in
the origin and evolution of therapy-related acute myeloid leukaemia.
References Nature. 2015;518(7540):552-555.
17. Takahashi K, Wang F, Kantarjian H, et al. Preleukaemic clonal
1. Agathangelidis A, Ljungström V, Scarfò L, et al. Highly similar
haemopoiesis and risk of therapy-related myeloid neoplasms: a case-
genomic landscapes in monoclonal B-cell lymphocytosis and ultra-
control study. Lancet Oncol. 2017;18(1):100-111.
stable chronic lymphocytic leukemia with low frequency of driver

752 haematologica | 2018; 103(5)


PERSPECTIVES

Denosumab for bone lesions in multiple myeloma – what is its value?


Daniel A. Goldstein
Davidoff Cancer Center, Rabin Medical Center, Petach Tikvah, Israel

E-mail: danielagoldstein@gmail.com
doi:10.3324/haematol.2017.185264

I
n June 2017 the Food and Drug Administration (FDA) zoledronic acid in delaying or preventing first on-study
accepted a supplemental biologics license application skeletal-related event [hazard ratio (HR)=0.82; 95% con-
seeking to expand the currently approved indication of fidence interval (95% CI): 0.71- 0.95; P= 0.01).4 Likewise,
denosumab to patients with bone lesions from multiple denosumab was superior in terms of time to first skeletal-
myeloma. The FDA set a prescription drug user act related event in patients with bone metastases from
(PDUFA) action date of February 3, 2018. Denosumab is an prostate cancer. The median time to first on-study skele-
inhibitor of receptor activator of nuclear factor κ-B ligand tal-related event was 20.7 months (95% CI: 18.8-24.9)
(RANKL) and was previously approved for post- with denosumab compared to 17.1 months (95% CI:
menopausal women at risk of osteoporosis in addition to 15.0-19.4) with zoledronic acid (HR=0.82, 95% CI: 0.71-
patients at risk of skeletal-related events due to bone 0.95; P=0.008 for superiority).5 Despite the reduction in
metastases from solid tumors and giant cell tumors of the skeletal-related events with denosumab, there was no
bone. The application for use in patients with myeloma is associated improvement in overall survival in patients
based on the findings of the recently presented ‘482 trial.1 with either breast or prostate cancer.4,5 In patients with
This commentary seeks to understand the value of this giant cell tumors of the bone, an open label study with
therapy for patients with multiple myeloma. denosumab demonstrated a high level of efficacy: 96% of
Denosumab is a monoclonal antibody and uses a novel patients with surgically unsalvageable giant cell tumors of
mechanism to decrease bone resorption. RANKL is a pro- the bone did not have disease progression after a median
tein expressed on osteoblastic stromal cells. It binds to follow-up of 13 months.6
receptor activator of nuclear factor-κB (RANK) and thus The ‘482 trial was an international phase 3, random-
mediates osteoclastic differentiation, activation, and sur- ized, double-blind trial comparing the safety and efficacy
vival. RANKL therefore controls osteoclast-mediated of monthly denosumab to monthly zoledronic acid in
bone resorption. Osteoprotegerin is a soluble RANKL patients with multiple myeloma.1 The trial enrolled 1718
decoy receptor which binds RANKL and is the key regu- patients and the primary endpoint was the time to first
lator of the RANKL–RANK pathway. Denosumab binds on-study skeletal-related event, and was powered to
to RANKL thus blocking the interaction of RANKL with demonstrate non-inferiority. Secondary endpoints were
RANK, mimicking the endogenous effects of osteoprote- time to first skeletal-related event (powered to superiori-
gerin. This agent has been shown to lead to a decrease in ty), time to first and subsequent skeletal-related events
bone resorption, based on changes in serum and urinary (powered to superiority), and overall survival. The study
N-telopeptide, which are markers of osteoclastic bone met the primary endpoint and demonstrated that deno-
resorption.2 sumab was non-inferior to zoledronic acid in terms of
Until recently bisphosphonates had been the standard skeletal-related events (HR=0.98; 95% CI: 0.85-1.14;
therapy for strengthening bone in a variety of conditions P=0.01). The trial failed to meet the secondary endpoints
such as osteoporosis and cancer. Bisphosphonates essen- of demonstrating superiority in terms of time to first
tially bind to bone mineral and inhibit the activity of skeletal-related event or overall survival. The authors per-
mature osteoclasts. Non-nitrogen containing bisphospho- formed an unplanned exploratory analysis to evaluate
nates achieve this goal by being metabolized to ATP progression-free survival as an endpoint and found a pro-
analogs that block osteoclast function and induce osteo- longed progression-free survival in the denosumab group
clast apoptosis. Nitrogen-containing bisphosphonates (HR=0.82; 95% CI: 0.68-0.99; P=0.036). Although the
inhibit farnesyl pyrophosphate synthase, thus preventing trial was well conducted with double-blind randomiza-
the post-translational modification of guanosine triphos- tion, this finding should be considered only as hypothe-
phate binding proteins which are essential for osteoclast sis-generating, as it was an unplanned endpoint analysis
function and survival.3 The essential difference between and such analyses are known to have a lack of statistical
bisphosphonates and denosumab is that bisphosphonates reliability.7
inhibit mature osteoclasts while denosumab inhibits There were no significant differences between the two
osteoclastic precursors. groups in terms of adverse events apart from hypocal-
Denosumab has already gained FDA approval for mul- cemia and renal toxicity. In patients with baseline creati-
tiple indications based on advanced phase clinical trials. nine clearance ≤60 mL/minute, 13% of patients in the
In postmenopausal women with low bone mineral densi- denosumab group developed renal toxicity, compared to
ty, it was found to lead to a 3.0% to 6.7% increase in 26% of patients in the zoledronic acid group (P<0.01).
bone mineral density of the lumbar spine.2 Multiple trials The rate of creatinine doubling from baseline in the zole-
have compared zoledronic acid and denosumab in dronic acid group was nearly twice as high as in the deno-
patients with solid tumors. In patients with bone metas- sumab group (6.5 versus 3.3%). Conversely, there were
tases from breast cancer, denosumab was superior to higher rates of hypocalcemia in patients receiving deno-

haematologica | 2018; 103(5) 753


Perspectives

sumab (17%) compared to those receiving zoledronic survival or skeletal events. In addition, the safety profile
acid (12%) (P=0.009). is very similar. There appears to be slightly more renal
In the USA we can calculate the cost of the drugs to toxicity with zoledronic acid; however, this is balanced
Medicare by using the average sales price (www.cms.gov). by the higher rates of hypocalcemia with denosumab.
This accounts for discounts and rebates and is a close esti- Although there was a demonstration of benefit in terms
mate of the cost to Medicare. The patent for zoledronic of progression-free survival, this finding should be treated
acid expired in 2013, at which point the reimbursement with caution, as it emerged from a post hoc exploratory
cost decreased. The average sales price for 4 mg of zole- analysis. There are, however, significant differences in
dronic acid is $48 and that for 120 mg of denosumab is costs – both to society and to patients. Denosumab costs
$2044. The annual cost is therefore $576 for zoledronic approximately $24,000 more per patient per year in the
acid, and $24,528 for denosumab – a difference of almost USA. Zoledronic acid is also considerably cheaper than
$24,000. In addition to this cost there is a mark-up of denosumab in Europe as well. Perhaps the most appropri-
4.3% that Medicare reimburses to providers. It should be ate management would be for all patients to receive zole-
noted that this mark-up may provide a financial incentive dronic acid, except those with a contraindication due to a
to the physician to prescribe the more expensive medica- low creatinine clearance. The reason for the high cost of
tion, despite the higher cost to the patient and insurer. new cancer drugs is complex. Without doubt, one of the
Finally, treatment centers also charge an infusion cost of many reasons is that the cost of drug development is
approximately $140, billed with code 96413 high, partially related to the many regulatory require-
(www.cms.gov). While these are the costs to Medicare, ments. However, while cancer is still often an incurable
we must also recognize that the patient often shares a sig- disease, we must strive towards bringing forward new
nificant proportion of the cost. In 2015, the average annu- therapies that provide clinically meaningful benefits to
al Medicare beneficiary cost share was $527 for deno- our patients.9 In an era of medical bancruptcies and
sumab and $68 for zoledronic acid (www.cms.gov - 2015 increasing healthcare costs, we owe it to both our
Medicare drug spending data). The price of drugs is dif- patients and society to incorporate costs into clinical deci-
ferent in other countries around the world; however, it is sion-making.
clear that everywhere in the world zoledronic acid is sig-
nificantly cheaper than denosumab. This commentary is
not intended to assess what was the most appropriate References
launch price of these drugs at the very different times of
1. Raje NS, Roodman GD, Willenbacher W, et al. Impact of denosumab
their being launched. The purpose is to discuss the most (DMB) compared with zoledronic acid (ZA) on renal function in the
appropriate choice of therapy in 2018, when the prices treatment of myeloma bone disease. J Clin Oncol.
are significantly different, due to one of the options being 2017;35(15_suppl):8005.
2. McClung MR, Lewiecki EM, Cohen SB, et al. Denosumab in post-
available in the significantly cheaper, generic form. menopausal women with low bone mineral density. N Engl J Med.
There is some additional convenience from using deno- 2006;354(8):821-831.
sumab. Firstly, denosumab can be given subcutaneously 3. Baron R, Ferrari S, Russell RG. Denosumab and bisphosphonates:
which may be preferable to the intravenous administra- different mechanisms of action and effects. Bone. 2011;48(4):677-
692.
tion of zoledronic acid. Secondly, denosumab is dosed the 4. Stopeck AT, Lipton A, Body J-J, et al. Denosumab compared with
same for all patients, and no adjustment is needed accord- zoledronic acid for the treatment of bone metastases in patients with
ing to renal function, whereas dose adjustments are nec- advanced breast cancer: a randomized, double-blind study. J Clin
Oncol. 2010;28(35):5132-5139.
essary for zoledronic acid. It is doubtful however, that 5. Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic
this additional convenience justifies the additional annual acid for treatment of bone metastases in men with castration-resis-
cost in the USA of $24,000 per patient. tant prostate cancer: a randomised, double-blind study. Lancet.
Recent data for zoledronic acid demonstrate equivalent 2011;377(9768):813-822.
6. Chawla S, Henshaw R, Seeger L, et al. Safety and efficacy of deno-
efficacy in patients with bone metastases secondary to sumab for adults and skeletally mature adolescents with giant cell
breast cancer, irrespective of whether the drug is given tumour of bone: interim analysis of an open-label, parallel-group,
monthly or every 3 months.8 Could these data perhaps be phase 2 study. Lancet Oncol. 2013;14(9):901-908.
7. Raghav KP, Mahajan S, Yao JC, et al. From protocols to publications:
extrapolated to patients with multiple myeloma? There a study in selective reporting of outcomes in randomized trials in
are currently no good quality data regarding the use of oncology. J Clin Oncol. 2015;33(31):3583-3590.
denosumab every 3 months in patients with neoplastic 8. Hortobagyi GN, Van Poznak C, Harker WG, et al. Continued treat-
ment effect of zoledronic acid dosing every 12 vs 4 weeks in women
bone disease. with breast cancer metastatic to bone: the OPTIMIZE-2 randomized
In an era of financial challenges for healthcare, we, as clinical trial. JAMA Oncol. 2017;3(7):906-912.
physicians, must be careful stewards of finite healthcare 9. Ellis LM, Bernstein DS, Voest EE, et al. American Society of Clinical
resources. There appears to be no benefit from using Oncology perspective: raising the bar for clinical trials by defining
clinically meaningful outcomes. J Clin Oncol. 2014;32(12):1277-
denosumab instead of zoledronic acid in terms of overall 1280.

754 haematologica | 2018; 103(5)


Perspectives

Osteoprotective medication in the era of novel agents: a European perspective on values,


risks and future solutions
Monika Engelhardt,1,2 Georg W. Herget,3 Giulia Graziani,1,2 Gabriele Ihorst,4 Heike Reinhardt,1,2 Stefanie Ajayi,1,2
Stefan Knop5 and Ralph Wasch1,2
1
Department of Medicine I, Hematology, Oncology & Stem Cell Transplantation, Medical Center, University of Freiburg;
2
Comprehensive Cancer Center Freiburg (CCCF), Medical Center, University of Freiburg; 3Department of Orthopedics and
Trauma Surgery, Medical Center, University of Freiburg; 4Clinical Trials Unit, Medical Center, University of Freiburg and
5
Hematology, Oncology, Gastroenterology, University of Würzburg, Germany
E-mail: monika.engelhardt@uniklinik-freiburg.de
doi:10.3324/haematol.2018.188516

O
steolytic bone disease is one of the most promi- order to prevent one skeletal-related event, 6-15 MM
nent features of multiple myeloma (MM) and is patients need to be treated.15
present in up to 80% of patients at diagnosis.1 The European Myeloma Network (EMN) and
Bone destruction leads to skeletal-related events (i.e. verte- International Myeloma Working Group (IMWG) have
bral and other pathological fractures) and/or spinal cord therefore recommended that all MM patients with ade-
compression. MM is mainly due to an increase in osteo- quate renal function (creatinine clearance >30 mL/min)
clastic activity which is accompanied by low osteoblastic and osteolytic disease at diagnosis should be treated with
function.1 Bisphosphonates and other bone-targeting zoledronic acid [4 mg i.v. infusion, over at least 15 min,
agents (such as denosumab which inhibits RANKL and every 4 weeks (Q4W) or pamidronate (90 mg, in a 3-hour
osteoclast function and is not renally cleared), effective infusion, Q4W], in addition to specific anti-myeloma
anti-myeloma treatment, radiotherapy and surgery are the therapy (grade 1A; definition of evidence levels: Online
main therapies used for the management of bone disease Supplementary Table S1). Symptomatic patients, without
in MM.1–3 Regarding the definition of MM-defining events, bone disease assessed by conventional radiography, can
there are important studies which suggest that asympto- be treated with zoledronic acid (grade 1B). The advantage
matic patients with more than one focal lesion detectable is not clear for patients without detectable bone involve-
by magnetic resonance imaging have a higher risk of pro- ment on magnetic resonance imaging or positron emis-
gression to symptomatic MM (>70% within 2 years).1,4–6 sion tomography/computed tomography.
These patients have been described by international Bisphosphonates are not routinely recommended in
myeloma experts as having symptomatic disease.5,6 smoldering MM (grade 1A); but in cases of osteoporosis
Based on phase 3 studies, the bisphosphonates, or vertebral fractures that are not due to the MM, bispho-
pamidronate and zoledronic acid, have been found to sphonates should be given at the doses given for osteo-
reduce skeletal-related events compared to placebo.7–9 porosis (5 mg zoledronic acid/year). For high-risk smol-
Three randomized studies have compared the effect of dering MM, the treating physician should consider using
different bisphosphonates or different dosages of the the bisphosphonate doses and schedules typically used to
same bisphosphonate. In the first study, zoledronic acid treat symptomatic MM (grade 1B). Zoledronic acid
was as effective as pamidronate in reducing skeletal-relat- should be given continuously (grade 1B). It is debatable
ed events in the era of conventional chemotherapy.9,10 In whether patients who achieve a very good partial
the second, two doses of intravenous pamidronate (30 response or better have benefits from the continuous use
versus 90 mg) showed comparable results regarding time of zoledronic acid. Regarding pamidronate, there are no
to skeletal-related events and survival time free of such data to support its continuous use; thus it should be given
events.11 The limitation of this study was that it was pow- for 2 years and then at the physician’s discretion (grade
ered to show differences in quality of life and not in 2C).1,2 Of note, bisphosphonates are now available as
skeletal-related events.11 The third study compared intra- generic drugs, whereas denosumab has just been
venous (i.v.) zoledronic acid with oral clodronate and approved by the Food and Drug Administration for use in
showed that zoledronic acid reduced the risk of skeletal- MM (January 2018; likewise anticipated in Europe) and is
related events compared to clodronate in all MM patients, patent-protected. This approval was based on the results
irrespective of the presence of lytic lesions at diagnosis, of a large phase III study comparing denosumab with
and improved overall survival by 10 months in patients zoledronic acid, in which the efficacy and safety of the
with lytic lesions at diagnosis.12,13 These effects continued drugs were assessed in newly diagnosed MM. Eligible
in patients who received zoledronic acid for >2 years.14 patients were randomized 1:1 to denosumab 120 mg sub-
There was no sub-analysis according to the response sta- cutaneously Q4W or zoledronic acid 4 mg (with dose
tus of the patients, thus it is not clear whether the contin- adjustments according to renal function) i.v. Q4W along
uous use of zoledronic acid produces similar results in with anti-myeloma therapy. The primary objective was
patients who have achieved excellent responses (≥very non-inferiority of denosumab to zoledronic acid with
good partial response). A meta-analysis was unable to respect to time to first on-study skeletal-related event.
confirm superiority of zoledronic acid over pamidronate, Overall survival was a secondary endpoint; progression-
but revealed a survival advantage from zoledronic acid free survival was an exploratory endpoint. The 1718
versus placebo.15 This analysis also determined that in patients enrolled were randomized into two arms, each

haematologica | 2018; 103(5) 755


Perspectives

with 859 participants. With regards to delaying time to than in the zoledronic acid group (n=129; 15.0%).
first on-study skeletal-related event, denosumab was not Therefore, denosumab showed non-inferiority to zole-
inferior to zoledronic acid [P=0.01; hazard ratio dronic acid in delaying time to first on-study skeletal-
(HR)=0.98; 95% confidence interval (95% CI): 0.85-1.14]. related event. Patients on denosumab had a significantly
Fewer adverse events potentially related to renal impair- lower rate of renal adverse events compared to those on
ment were reported with denosumab than with zole- zoledronic acid. The bone-specific benefits in combina-
dronic acid (10.0% versus 17.1%, P<0.001). The HR for tion with the renal function results and possible prolonga-
progression-free survival was 0.82 (95% CI: 0.68-0.99; tion of progression-free survival with denosumab were
P=0.036). The overall survival HR between denosumab promising and have led to invigorating discussions about
and zoledronic acid was 0.9 (95% CI: 0.70-1.16; P=0.41), why progression-free survival data were more favorable
with fewer deaths in the denosumab arm (n=121; 14.1%) with denosumab. The observations definitely need deep-

Table 1. Cost comparison of osteoprotective medications for MM: Germany versus USA.
Costs Germany Costs USA
[Monthly costs in Euro (€)] [Monthly costs in Euro (€)]
Drug (original) Dose & mode of administration Original price Generic price Original price Generic price
Denosumab (Xgeva ) ®
120 mg s.c. bolus 440 - 1890 -
Zoledronic acid (Zometa®) 4 mg i.v. over 15 min 368 279 814 49
Pamidronate 90 mg i.v. over 3 hours out of trade 251 out of trade 47
German prices: ATaxx® (Dr. Heni Software GmbH & Co.KG, Freiburg); USA prices: https://www.drugs.com/price-guide/. In Germany, the AMNOG is limiting the cost of new phar-
maceutical products; In the USA, a deflation of generic prices has been reported, due to an increasing number of competing companies entering the market
(https://www.nytimes.com/2017/08/08/health/generic-drugs-prices-falling.html).

A B

C D

Figure 1. Features of the diagnosis of multiple myeloma. (A) Time from onset of symptoms to the diagnosis of MM: patients (%) diagnosed within <3, 3-6, 6-11 or
>12 months in the retrospective versus prospective analysis. (B) First suspicion of MM: frequency (%) of patients whose MM was first suspected by different types
of physicians (n=176 patients; prospective cohort). (C-D) Prospective analysis: patients‘ satisfaction (n=176).

756 haematologica | 2018; 103(5)


Perspectives

er understanding and statistical evaluation: the relation- seeking agents are currently needed. Since skeletal-related
ship between the occurrence of skeletal-related events events continue to occur in the first months of treatment
and progression-free survival-defining events needs to be and with relapse (despite the use of novel agents and
defined. Furthermore, an assessment of cumulative inci- osteoprotection1–3), effective prevention and reduction of
dence rates of skeletal-related events with death as a destructive skeletal-related events remain fundamental.1,20
competing event will be helpful, as the slight overall sur- Recent data from the national registry, Hospital Episode
vival disadvantage in the zoledronic acid arm might have Statistics determined fracture rates and the effect on over-
led to fewer skeletal-related events. Nevertheless, with all survival in MM patients between 2001 and 2015:
full publication of the results16 and with EMA approval, expectedly, fracture rates were 18 times higher with MM
denosumab will be used in Europe in MM patients.1–3 in the first year after admission than in the general popu-
Given these insights and major advances in the under- lation, and remained elevated for up to 10 years. In line
standing of the disease, early MM diagnosis, especially of with the data on early diagnosis in MM,2,3,17 the increased
symptomatic patients, has been advocated. However, fracture risk preceded the first admission with MM and
since MM is an insidiously developing malignancy and conversely the incidence of MM increased after admis-
may appear with non-specific symptoms, e.g. bone pain, sion with one or more fractures. Fractures were associat-
the diagnosis and therapeutic decisions can be complicat- ed with poorer outcome (HR for overall survival: 1.2),
ed. A German study group (DSMM) and EMN project indicating the need for regular use of bone supportive
addressed this aspect with the aim of optimizing the drugs despite novel agent-based treatment.21 In addition,
prompt diagnosis and further improving the quality of cost analyses in 1028 MM patients (596 with ≥1 skeletal-
MM care.17 An initial retrospective analysis of 101 MM related events and 432 without skeletal-related events)
patients was followed by a prospective study of 176 demonstrated that a higher frequency of skeletal-related
patients using a structured MM-specific questionnaire. events was associated with greater utilization of health-
The median time from the patients' first symptoms to the care resources, suggesting that bone supportive drugs
final MM diagnosis was 4 months (range, 0.5-120) in the need to be used diligently to avoid higher healthcare costs
retrospectively studied cohort and very similar to the 6 due to skeletal complications and patients’ discontent.22
months (range, 0.5-60) in the prospective cohort. Of inter- Since bisphosphonates in symptomatic MM have been
est, the time from onset of symptoms to diagnosis of MM suggested, but beyond 2 years and with stable MM are
was ≥12 months in 20% of the patients in the retrospec- left to the discretion of the treating physician, a random-
tive analysis and 35% in the prospective study (Figure ized trial assessed 170 untreated, symptomatic patients
1A). The frequencies of MM-related bone fractures, renal using zolendronic acid for 4 versus 2 years.23 All patients
complications and infections occurring before the diagno- were treated with the same induction therapy and stem-
sis of MM was made were 41%, 35% and 16%, respec- cell transplantation. The group treated for 4 years had
tively. Moreover, 43% had one, 20% had two and 3% substantially fewer skeletal-related events than the group
had three of these complications. The most frequent treated for 2 years (21 versus 43%, respectively; P<0.001).
symptom was bone pain, which occurred in 73% of MM Actuarial curves at 5 years showed that progression-free
patients before the final MM diagnosis was made. In 6% survival was 75% (95% CI: 64%-82%) and overall sur-
of patients, MM was first suspected by orthopedists, vival 68% (95% CI, 60%-76%) in the group treated for 4
whereas the clinical suspicion was raised by nephrolo- years; these rates were not significantly different from
gists in 16% of cases, even though renal impairment was those of the control group treated for 2 years with zole-
less frequent (Figure 1B). Of interest, 61% of patients dronic acid (P=0.67); but this trial was underpowered to
were completely or fairly satisfied with the diagnostic show differences in survival. The trial did, however, con-
process, whereas 39% were less satisfied (Figure 1C). firm that the continued use of zoledronic acid was useful
Fifty-eight percent of the patients believed that their dis- to reduce skeletal-related events and to preserve a better
ease could have been diagnosed more expeditiously quality of life.23
(Figure 1D). Patients, who criticized the slow diagnostic With bisphosphonates and denosumab being potent
process had a much longer median time interval from options in MM, Goldstein, in this issue of the Journal,
symptom onset to their final MM diagnosis compared to comments on both costs and the fact that novel patent-
those who were less critical (9 versus 3 months, respec- protected drugs will induce greater expenditure than
tively). These results demonstrate that there is still con- generically available alternatives.24 While there is an
siderable latency in the diagnosis of MM. However, even unequivocal need to thoroughly evaluate and measure
with early diagnosis and treatment with novel agents, "real" advances with new drugs, shortcomings of this
skeletal-related events continue to occur, in part due to commentary are the "generalization" regarding patent
MM responses ("melting-down MM") and relapses, versus generic medications, the understatement of pro-
reminding us that progress in MM involves understand- gression-free survival differences, convenience of subcu-
ing how best to avoid skeletal-related events before the taneous versus i.v. medication, and the decreased renal
diagnosis of the disease is made and with antimyeloma impairment and safer use of denosumab in patients with
treatment, because this substantially influences patients' renal impairment. Moreover, Goldstein’s conclusions
coping and their approval of our MM care.2,3,17 The notion only apply to the health system in the USA, whereas
that treatment based on novel agents promotes bone- reimbursement of medication providers and financial
healing - apart from osteoprotective supportive agents incentives to physicians to prescribe more expensive
such as bisphosphonates and denosumab - has recently drugs are different in Europe (Table 1).24 The rising cost of
led to the demanding discussion18,19 of whether bone- patented cancer medicines in the USA is a known phe-

haematologica | 2018; 103(5) 757


Perspectives

scape of smoldering multiple myeloma: a European perspective.


nomenon: from 2000 until now, there have been 5- to 10- Oncologist. 2016;21(3):333-342.
7. Berenson JR, Lichtenstein A, Porter L, et al. Efficacy of pamidronate
fold increases in the cost of new drugs.25 Price differences in reducing skeletal events in patients with advanced multiple
in Europe can, therefore, be substantially different from myeloma. Myeloma Aredia Study Group. N Engl J Med.
those in the USA (Table 1). In Germany, a benefit assess- 1996;334(8):488-493.
ment of pharmaceuticals in accordance with the Act on 8. Berenson JR, Rosen LS, Howell A, et al. Zoledronic acid reduces
skeletal-related events in patients with osteolytic metastases.
the Reform of the Market for Medicinal Products Cancer. 2001;91(7):1191-1200.
(AMNOG) is limiting the cost of new pharmaceutical 9. Rosen LS, Gordon D, Kaminski M, et al. Zoledronic acid versus
products. In the USA, a deflation of generic prices has pamidronate in the treatment of skeletal metastases in patients with
breast cancer or osteolytic lesions of multiple myeloma: a phase III,
been reported, due to an increasing number of competing double-blind, comparative trial. Cancer J. 2001;7(5): 377-387.
companies entering the market. As clinicians and 10. Rosen LS, Gordon D, Kaminski M, et al. Long-term efficacy and safe-
researchers we know that medical advances are needed, ty of zoledronic acid compared with pamidronate disodium in the
and developmental costs for new drugs have steadily treatment of skeletal complications in patients with advanced multi-
ple myeloma or breast carcinoma: a randomized, double-blind, mul-
increased with regulatory requirements. Goldstein ticenter, comparative trial. Cancer. 2003;98(8):1735-1744.
reminds us of the financial burden associated with new 11. Gimsing P, Carlson K, Turesson I, et al. Effect of pamidronate 30 mg
cancer agents, which we offer to our patients with the versus 90 mg on physical function in patients with newly diagnosed
multiple myeloma (Nordic Myeloma Study Group): a double-blind,
aim to 'never harm but always aid': nevertheless, the judge- randomised controlled trial. Lancet Oncol. 2010;11(10):973-982.
ment regarding bisphosphonate generics versus denosum- 12. Morgan GJ, Davies FE, Gregory WM, et al. First-line treatment with
ab is biased and the comparison is cumbersome. His con- zoledronic acid as compared with clodronic acid in multiple myelo-
ma (MRC Myeloma IX): a randomised controlled trial. Lancet.
clusions that generic bisphosphonates have a novel coun- 2010;376(9757):1989-1999.
terpart and that the financial burden with denosumab is 13. Morgan GJ, Child JA, Gregory WM, et al. Effects of zoledronic acid
higher are, however, worth noting.24 Thus, the good news versus clodronic acid on skeletal morbidity in patients with newly
prevails that treatment options for the prevention of bone diagnosed multiple myeloma (MRC Myeloma IX): secondary out-
comes from a randomised controlled trial. Lancet Oncol.
complications have increased with the introduction of 2011;12(8):743-752.
denosumab, providing a new choice for patients and 14. Morgan GJ, Davies FE, Gregory WM, et al. Effects of induction and
physicians. Once initiated, bisphosphonates or denosum- maintenance plus long-term bisphosphonates on bone disease in
patients with multiple myeloma: the Medical Research Council
ab should be continued for at least 2 years, after which a Myeloma IX Trial. Blood. 2012;119(23):5374-5383.
suspension of bisphosphonate treatment may be consid- 15. Mhaskar R, Redzepovic J, Wheatley K, et al. Bisphosphonates in
ered in very responsive patients, although this may be multiple myeloma: a network meta-analysis. Cochrane Database
associated with skeletal risks, especially in those with Syst Rev. 2012;(5):CD003188.
16. Raje N, Terpos E, Willenbacher W, et al. An international, ran-
prior skeletal complications. For patients in whom bis- domised, double-blind study of denosumab compared to zoledronic
phosphonates were stopped after 2 years, the drug should acid in bone disease treatment of newly diagnosed multiple myelo-
be resumed on a monthly basis if the MM recurs and/or ma. Lancet Hematol. 2018;19(3):370-381.
17. Graziani G, Herget G, Ihorst G, et al. Time from first symptom onset
new skeletal-related events occur, independently of the to the final diagnosis of multiple myeloma - possible risks and future
use of novel agent-based therapies.1–3 solutions: large retrospective and conformatory prospective
“Deutsche Studiengruppe Multiples Myelom” (DSMM) analysis.
Blood. 2017;130(Suppl 1):4710.
Acknowledgment 18. Delforge M, Terpos E, Richardson PG, et al. Fewer bone disease
We are deeply indebted to esteemed experts of the Deutsche events, improvement in bone remodeling, and evidence of bone
Studiengruppe Multiples Myelom, German Multiple Myeloma healing with bortezomib plus melphalan-prednisone vs. melphalan-
Study Group, European Myeloma Network Group and prednisone in the phase III VISTA trial in multiple myeloma. Eur J
Haematol. 2011;86(5):372-384.
International Myeloma Working Group for their valuable dis- 19. Mohty M, Malard F, Mohty B, Savani B, Moreau P, Terpos E. The
cussion. effects of bortezomib on bone disease in patients with multiple
myeloma. Cancer. 2014;120(5):618-623.
20. Engelhardt M, Terpos E, Kleber M, et al. European Myeloma
Network recommendations on the evaluation and treatment of
References newly diagnosed patients with multiple myeloma. Haematologica.
2014;99(2):232-242.
1. Terpos E, Kleber M, Engelhardt M, et al. European Myeloma 21. McIlroy G, Mytton J, Evison F, et al. Increased fracture risk in plasma
Network Guidelines for the management of multiple myeloma-relat- cell dyscrasias is associated with poorer overall survival. Br J
ed complications. Haematologica. 2015;100(10):1254-1266. Haematol. 2017;179(1):61-65.
2. Terpos E, Christoulas D, Gavriatopoulou M. Biology and treatment 22. Nash Smyth E, Conti I, Wooldridge JE, et al. Frequency of skeletal-
of myeloma related bone disease. Metabolism. 2018;80:80-90. related events and associated healthcare resource use and costs in US
3. Yee AJ, Raje NS. Denosumab for the treatment of bone disease in patients with multiple myeloma. J Med Econ. 2016;19(5):477-486.
solid tumors and multiple myeloma. Future Oncol. 2018;14(3):195- 23. Avilès A, Nambo M-J, Huerta-Guzmàn J, Cleto S, Neri N. Prolonged
203. use of zoledronic acid (4 Years) did not improve outcome in multiple
4. Hillengass J, Fechtner K, Weber M-A, et al. Prognostic significance of myeloma patients. Clin Lymphoma Myeloma Leuk. 2017;17(4):207-
focal lesions in whole-body magnetic resonance imaging in patients 210.
with asymptomatic multiple myeloma. J Clin Oncol. 24. Goldstein D. Denosumab for bone lesions in multiple myeloma -
2010;28(9):1606-1610. what is its value? Haematologica. 2018(5):753-754.
5. Rajkumar SV, Dimopoulos MA, Palumbo A, et al. International 25. Kantarjian H, Steensma D, Rius Sanjuan J, Elshaug A, Light D. High
Myeloma Working Group updated criteria for the diagnosis of mul- cancer drug prices in the United States: reasons and proposed solu-
tiple myeloma. Lancet Oncol. 2014;15(12):e538-548. tions. J Oncol Pract. 2014;10(4):e208-211.
6. Caers J, Fernández de Larrea C, Leleu X, et al. The changing land-

758 haematologica | 2018; 103(5)


Bone Marrow Failure ARTICLE

Deep sequencing and flow cytometric


characterization of expanded effector memory Ferrata Storti
Foundation
CD8+CD57+ T cells frequently reveals T-cell
receptor Vβ oligoclonality and CDR3 homology
in acquired aplastic anemia
Valentina Giudice,1 Xingmin Feng,1 Zenghua Lin,1,2 Wei Hu,3 Fanmao Zhang,3
Wangmin Qiao,3 Maria del Pilar Fernandez Ibanez,1 Olga Rios,1
and Neal S. Young1

1
Hematology Branch, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda,
MD, USA; 2Department of Hematology, Affiliated Hospital of Nantong University, Jiangsu,
Haematologica 2018
China and 3BGI Genomics, BGI-Shenzhen, China
Volume 103(5):759-769

ABSTRACT

O
ligoclonal expansion of CD8+CD28– lymphocytes has been con-
sidered indirect evidence for a pathogenic immune response in
acquired aplastic anemia. A subset of CD8+CD28– cells with
CD57 expression, termed effector memory cells, is expanded in several
immune-mediated diseases and may have a role in immune surveillance.
We hypothesized that effector memory CD8+CD28–CD57+ cells may
drive aberrant oligoclonal expansion in aplastic anemia. We found
CD8+CD57+ cells frequently expanded in the blood of aplastic anemia
patients, with oligoclonal characteristics by flow cytometric Vβ usage
analysis: skewing in 1-5 Vβ families and frequencies of immunodomi-
nant clones ranging from 1.98% to 66.5%. Oligoclonal characteristics
were also observed in total CD8+ cells from aplastic anemia patients
with CD8+CD57+ cell expansion by T-cell receptor deep sequencing, as Correspondence:
well as the presence of 1-3 immunodominant clones. Oligoclonality was
confirmed by T-cell receptor repertoire deep sequencing of enriched fengx2@nhlbi.nih.gov
CD8+CD57+ cells, which also showed decreased diversity compared to
total CD4+ and CD8+ cell pools. From analysis of complementarity-deter-
mining region 3 sequences in the CD8+ cell pool, a total of 29 sequences Received: July 21, 2017.
were shared between patients and controls, but these sequences were
highly expressed in aplastic anemia subjects and also present in their Accepted: December 30, 2017.
immunodominant clones. In summary, expansion of effector memory Pre-published: February 1, 2018.
CD8+ T cells is frequent in aplastic anemia and mirrors Vβ oligoclonal
expansion. Flow cytometric Vβ usage analysis combined with deep
sequencing technologies allows high resolution characterization of the doi:10.3324/haematol.2017.176701
T-cell receptor repertoire, and might represent a useful tool in the diag-
nosis and periodic evaluation of aplastic anemia patients. (Registered at Check the online version for the most updated
clinicaltrials.gov identifiers: 00001620, 01623167, 00001397, 00071045, information on this article, online supplements,
00081523, 00961064) and information on authorship & disclosures:
www.haematologica.org/content/103/5/759

Introduction ©2018 Ferrata Storti Foundation


Material published in Haematologica is covered by copyright.
Acquired aplastic anemia (AA) is a bone marrow (BM) failure syndrome charac- All rights are reserved to the Ferrata Storti Foundation. Use of
published material is allowed under the following terms and
terized by peripheral blood (PB) pancytopenia and BM hypocellularity due to conditions:
hematopoietic stem and progenitor cell destruction.1-7 There is indirect evidence to https://creativecommons.org/licenses/by-nc/4.0/legalcode.
support the hypothesis of autologous immune attack: clinical response to Copies of published material are allowed for personal or inter-
immunosuppressive therapies (IST),1,2,8-10 the predominant role of activated cyto- nal use. Sharing published material for non-commercial pur-
poses is subject to the following conditions:
toxic T cells (CTLs) in BM growth inhibition,1,10-12 identification of putative https://creativecommons.org/licenses/by-nc/4.0/legalcode,
autoantigens,13,14 and oligoclonal expansion of CD8+ lymphocytes.8,14-17 sect. 3. Reproducing and sharing published material for com-
Oligoclonality of T-cell populations has been defined by flow cytometry and mercial purposes is not allowed without permission in writing
deep sequencing of the T-cell receptor (TCR) Vβ repertoire and by spectratyping from the publisher.
of complementarity-determining region 3 (CDR3) length skewing.18-21 The TCR, an
aβ or γd heterodimer, is responsible for antigen recognition and T-cell activation.22-

Haematologica | 2018; 103(5) 759


V. Giudice et al.

26
Each chain is the result of a complex gene locus Table S2). DNA was also isolated from beads-sorted CD8+CD57+
rearrangement, known as VDJ recombination.23 On a cells from 2 of the 12 SAA patients with enough cells for further
rearranged VDJ segment, a terminal deoxynucleotidyl analysis (mean: 1.6 μg of DNA). TCR repertoire sequencing was
transferase enzyme increases TCR variability through performed with an Illumina HiSeq 2000 sequencer (Illumina Inc.,
insertions/deletions within a hypervariable region, the San Diego, CA, USA). Detailed information is provided in the
CDR3, generating a unique potential antigen-specific Online Supplementary Methods. Data have been deposited in the
sequence.23 Early in infection, CD28+ CTLs with different NCBI GEO database (accession n. GSE101660).
antigen affinity are selected and expanded (polyclonal
phase).27 In late stages, high antigen-affinity CD28– T cells Statistical analysis
are in resting state as memory T cells.28-29 The expression Data were analyzed using R (RStudio, Boston, MA, USA) and
of CD57 on CD8+CD28– T cells identifies a subset of Prism (v.7.02; GraphPad software Inc., La Jolla, CA, USA). Mann-
memory T cells termed effector memory because of their Whitney U test, Wilcoxon signed rank sum test, pair and un-
high antigen-affinity and ability to be rapidly activated paired t-tests, or χ2 test were used for data with abnormal distribu-
after antigen stimulation, as “tissue-guards”.29,30 Direct evi- tions. Bonferroni and Dunn’s corrections were used for multiple
dence of their high antigen-affinity is decreased diversity comparisons. P≤0.05 was considered statistically significant, after
in the CD8+CD57+ TCR repertoire (oligoclonality) due to adjustment with Bonferroni and false discovery rate (FDR).35
the presence of only a few selected clones of memory Linear regression was performed for correlations. Log-rank
cells.27 (Mantel-Cox) test was used for progression-free survival data
In this work, we investigated the frequency and oligo- analysis. Simpson's diversity index was calculated according to
clonal expansion of effector CD28–CD57+ memory cells in the following formula:
CD4+ and CD8+ T cells and the TCR Vβ repertoire in AA
patients by flow cytometry and deep sequencing tech-
nologies, to provide additional evidence for the immune
hypothesis in AA pathophysiology.

in which ni represents the clone size as the number of copies of


Methods each clonotype (i) or the total number of CDR3 sequences belong-
ing to each i clonotype, and n is the total number of different
Human samples clonotypes in the sample or the total number of sequences for
Heparinized whole PB was collected from patients and healthy each sample.
subjects after informed consent, in accordance with the
Declaration of Helsinki31 and protocols approved by the National
Heart, Lung, and Blood Institute Institutional Review Board Results
(National Institutes of Health, Bethesda, MD, USA) (see Online
Supplementary Table S1 for clinical characteristics). HLA haplotypes Effector memory CD8+CD57+ T cells frequently show
are reported in Online Supplementary Table S2. All patients received oligoclonal expansion of TCR Vβ repertoire by flow
a diagnosis of severe AA (SAA) according to the International cytometry
Study of Aplastic Anemia and Agranulocytosis32 and to the criteria Immunophenotyping and flow-cytometry Vβ usage
of Camitta.33 At the time of blood sampling, none of the patients were performed in 24 SAA patients. Clinical characteris-
had received therapy. Peripheral blood mononuclear cells (PBMCs) tics are reported in Online Supplementary Table S1. A group
were isolated by Ficoll-Paque density gradient centrifugation (MP of 34 healthy subjects was studied in order to define nor-
Biomedicals, LLC, Santa Ana, CA, USA) according to the manu- mal ranges of T-cell populations and Vβ family expression.
facturer’s instructions. SAA patients showed higher frequencies of CD8+CD57+
cells (25.6±17.3% vs. 13.3±12.6% in healthy individuals;
Flow cytometry P=0.003), and decreased frequency of CD8+CD28+ cells
A minimum of 4x106 PBMCs from each subject were stained for (56.8±25.7% vs. 68.8±19.1%; P=0.046). A negative corre-
TCR Vβ repertoire analysis (Online Supplementary Figure S1). The lation between CD57 and CD28 expression was also pres-
manufacturer’s instructions for the IOTest Beta Mark (Beckman ent (r2=0.601, P<0.0001). No differences were found for
Coulter, Miami, FL, USA) were optimized to avoid incorrect com- CD28+ and CD57+ cells within the CD4+ subset (P=0.974
pensation due to the existence of FITC/PE double positive anti- and P=0.250, respectively) (Figure 1A).
bodies in the kit, as described in Online Supplementary Methods. By Vβ usage, polyclonal expansion was observed in
Expansion of CD8+CD57+ cells was defined using as a threshold total CD4+, CD4+CD28+, CD4+CD57+ and CD8+CD28+
the mean frequency calculated in healthy subjects. Vβ skewing in cells in both healthy subjects and SAA patients (Figure 1B
SAA patients was described when the frequency of one Vβ family and Online Supplementary Figure S2). Oligoclonal expan-
was higher than the mean+3Standard Deviation (SD) in healthy sion of CD8+CD57+ cells was present in 92% of SAA
subjects.16 The term “immunodominant clone” was defined as the patients with 1-3 immunodominant clones, while in total
Vβ+ population by flow cytometry or the DNA sequence by deep CD8+ cells oligoclonality was reported only in 33% of
sequencing present in the highest abundance.17 cases (Online Supplementary Figure S3). Patients did not
show expansion of a shared Vβ family, as each subject car-
TCR repertoire deep sequencing ried a different TCR Vβ rearrangement in effector memo-
For VDJ combination and CDR3 sequence profiling,34 DNA was ry CD8+ T cells (Figure 2A). None of the 7 patients with-
isolated from FACS- or beads- (Miltenyi Biotec Inc., San Diego, out CD8+CD57+ cell expansion showed Vβ skewing in any
CA, USA) sorted CD4+ and CD8+ T cells from 12 SAA patients subgroup, with mean frequency of the immunodominant
with CD8+CD57+ cell expansion and 9 healthy subjects clone of 3.8% (range: 0.21-6.01%). Conversely, all 17
(mean: 3.2 μg of DNA; range: 0.2-27.4 μg) (Online Supplementary patients with effector memory CD8+ cell expansion

760 haematologica | 2018; 103(5)


TCR repertoire of effector memory T cells in AA

showed Vβ skewing in 1-5 Vβ subgroups, and frequencies frequency (n=7) experienced relapse (median survival not
of the immunodominant clones ranged from 2.1% to reached; median follow-up time: 13.6 months), while 7 of
66.5% (mean: 9.9%). the 17 SAA subjects with expanded effector memory T
Progression-free survival (PFS) analysis was performed cells relapsed or died (median survival 13.2 months; medi-
on all SAA patients, divided according to pre-treatment an follow up: 10.4 months). However, statistical signifi-
frequencies of effector memory CD8+ T cells (cut-off value cance between the two curves was not reached (P=0.089).
13.3%) (Figure 2B). No patients with low CD8+CD57+ cell Vβ usage was analyzed in CD4+ and CD8+ T-cell subsets

**

Figure 1. Immunophenotyping and flow cytometry analysis of Vβ usage in severe aplastic anemia (SAA) patients and healthy subjects. (A) Percentages of CD28+
and CD57+ cells were calculated in both CD4+ and CD8+ compartments for healthy controls and SAA patients. Data are shown as mean±Standard Deviation (SD).
Unpaired t-test was performed. *P<0.05; **P<0.01. (B) Vβ usage was studied in T-cell compartments (by row), and percentages of each Vβ family were reported as
total CD4+ or CD8+ cell percentage. For Vβ usage in healthy subjects, data are shown as mean+SD, combining the results from all 34 healthy donors. For SAA
patients, 2 representative cases are shown.

haematologica | 2018; 103(5) 761


V. Giudice et al.

in serial samples available for Patients 4, 22, and 34 in Supplementary Figure S4A). Vβ usage was also investigated
order to understand the correlation of Vβ usage with clin- at baseline in the BM of these 2 patients (Online
ical course (Figure 2C and Online Supplementary Figure Supplementary Figure S4B), and high concordance with Vβ
S4A). For Patient 22, at baseline, Vβ was the immunodom- usage of PB CD8+CD57+ cells was described.
inant clone and mostly enriched in CD8+CD57+ cell popu- Increased expansion of effector memory CD8+ T cells
lation, rather than in CD4+CD28+, CD4+CD57+, and with age has been reported;36 therefore, we used a pool of
CD8+CD28+ cells. After ten days of IST, the size of the age-matched healthy controls to assess the effect of age.
CD8+CD57+ clone was greatly reduced. The clone was There was no correlation between the size of the immun-
detected again at six months (3 months before clinical odominant clone in CD8+CD57+ cells and age in healthy
relapse) and further increased at relapse (Figure 2C), sug- subjects (r2=0.0003, P=0.919) or in SAA patients (r2=0.140,
gesting association of Vβ expansion with clinical status. P=0.079) (Online Supplementary Figure S5). However, a cor-
Patients 4 and 34 were non-responders at three months, relation was found between CD57 expression and age in
and non- and minimal partial-responders at the 6-month SAA patients (r2=0.552, P<0.0001).
time point. However, no significant changes in immun- We then assessed the effect of transfusion history on
odominant clone size were observed (Online oligoclonal expansion of effector memory T cells, as trans-

B C

Figure 2. Vβ usage at diagnosis and during treatment. (A) Percentages of Vβ family in CD8+CD57+ cells were calculated on total CD8+ cells, and Vβ skewing in severe
aplastic anemia (SAA) patients was defined using the mean+3Standard Deviation (SD) of a given Vβ group in healthy donors. Relative expansion of each Vβ subgroup
is shown in the bar graph. Patients were divided based on the absence or presence of expanded CD8+CD57+ cells, using the mean in healthy donors (13.3%). Skewing
of one Vβ family is reported as an orange bar. (B) Progression-free survival rate of SAA patients with CD8+CD57+ cells ≤13.3% (n=7) or >13.3% (n=17) prior to treat-
ment. Log-rank (Mantel-Cox) test was performed. (C) Vβ usage was performed in Patient 22 at diagnosis, at 10 days of treatment, and at 6 and 9 months (relapse).
Perturbations during treatment and relapse are reported as percentage of positive CD8+CD57+ and Vβ2+ cells (left), or absolute lymphocyte and Vβ2 clone count
(right).

762 haematologica | 2018; 103(5)


TCR repertoire of effector memory T cells in AA

fusions expose T cells to multiple foreign epitopes.37 A 13.3±12.6%, respectively; P=0.216) (Online Supplementary
group of 5 pure red cell aplasia (PRCA) patients, 10 sickle Figure S6A). Vβ skewing was described in 4 PRCA patients
cell disease (SCD) subjects, and 8 myelodysplastic (MDS) in 1-8 subgroups, in all 8 MDS subjects in 1-4 Vβ families,
patients who had been heavily transfused before sampling and in 7 SCD patients in 1-5 subgroups (Online
were studied for CD8+CD57+ cell expansion and Vβ usage Supplementary Figure S6B). Oligoclonal expansion was
(Online Supplementary Table S1). No variations were described in 91% of cases and subjects without skewing
observed in CD8+CD57+ cell frequencies when PRCA, did not present effector memory T-cell expansion.
MDS, and SCD patients were compared to healthy sub- Subsequently, we combined all subjects and divided them
jects (22.8±25.1% vs. 21.6±9.7% vs. 30.6±28.3% vs. according to their transfusion history and the presence of

Figure 3. Characterization of Vβ/Jβ plot, CDR3 size and DJ length profiles in healthy donors by deep sequencing. (A) T-cell receptor β variable (TRBV)/T-cell receptor
β joining (TRBJ) plots showed a “citylike” landscape for total CD4+ and CD8+ cell populations in healthy subjects (HC). (B) The size of the complementarity region 3
(CDR3) and DJ length profiles were also defined, showing similar features in CD4+ and CD8+ cells.

haematologica | 2018; 103(5) 763


V. Giudice et al.

CD8+CD57+ T-cell expansion. By χ2 test, transfusion histo- in CD4+ T cells, and 10,961,961±3,879,596 in CD8+ popula-
ry did not correlate to CD8+CD57+ expansion (P=0.255). tions. The mean frequency of the immunodominant clone
was 4.1±4.1% in CD4+ cells and 17.3±16.9% in CD8+ cells.
Total CD8+ cell TCR repertoires are polyclonal in By plotting the number of reads of each Vβ and Jβ
healthy subjects matching,17 a “citylike” landscape was described for total
Deep sequencing of TCR repertoire was performed in CD4+ and CD8+ T-cell populations (Figure 3A and Online
CD4+ and CD8+ populations sorted from 9 healthy donors. Supplementary Figure S7A), because of the presence of a
The average depth of sequencing was 10,790,646±4,050,138 more homogenous distribution of TCR rearrangement fre-

Figure 4. The TCR repertoire by deep sequencing analysis from total CD8+ cells in severe aplastic anemia (SAA) patients with CD8+CD57+ cell expansion. In contrast
to healthy CD8+ profiles, most SAA patients (AA) displayed oligoclonal features in a TRBV/TRBJ rearrangement plot (A) and CDR3 size and DJ length profiles (B). CD4+
and CD8+ profiles are shown for each SAA patient. In AA1 and AA6, only CD8+ cells were sufficient for deep sequencing.

764 haematologica | 2018; 103(5)


TCR repertoire of effector memory T cells in AA

quencies (“midtown”). The normal CDR3 size and DJ etry, and also in CD8+CD57+ cells from 2 of these patients.
length profiles were defined by comparing distributions The average depth of sequencing was
among healthy donors (Figure 3B and Online 13,861,048±4,992,677 in CD4+ T cells,
Supplementary Figure S7B). In CD4+ and CD8+ cells, profiles 13,873,207±5,029,195 in CD8+ T cells, and
were typically distributed in a Gaussian manner with 10- 21,030,616±1,238,660 in CD8+CD57+ cells. The mean fre-
12 different size classes of 30-65 nucleotides (nt) sizes at 3 quency of the immunodominant clone was 3.3±3.4%
nt intervals, and they completely overlapped. Similarly, DJ (range: 0.2-11.8%) in CD4+, 18.2±14.9% (range: 3.3-
length profiles assumed an asymmetric Gaussian distribu- 54.1%) in CD8+ cells, and 59±28.9% in CD8+CD57+cells.
tion with 2-5 predominant length classes without nt inter- By plotting TRBV/TRBJ rearrangements from CD4+ cells,
vals in CD4+ and CD8+ subsets. the “citylike” landscape was found in 11 out of 12 patients
(Figure 4A and Online Supplementary Figure S8A). In the
Deep sequencing allows detailed characterization of CD8+ cell pool, the “citylike” landscape was found in 3
oligoclonality in SAA patients (AA7, AA8, and AA11), whose clones showed
Deep sequencing of TCR repertoire was performed in very low frequencies (4.3%, 3.4%, and 3.3%, respective-
CD4+ and CD8+ cells from 12 SAA patients who had ly). The remaining patients displayed a “skyscraper” land-
demonstrated CD8+CD57+ cell expansion by flow cytom- scape due to the presence of 1-3 immunodominant clones.

Figure 5. The TCR repertoire by deep sequencing of enriched CD8+CD57+ cells in severe aplastic anemia (SAA) patients. (A) The enrichment of the clone in effector
memory CD8+ T cells, comparing TRBV/TRBJ rearrangement in total CD8+ cells (left) with those in CD8+CD57+ cells (right) from the same patients. (B) CDR3 size and
DJ length profiles from CD8+CD57+ cells also overlapped with those in CD4+ and CD8+ profiles.

haematologica | 2018; 103(5) 765


V. Giudice et al.

CDR3 size and DJ length profiles were defined in total remaining patients, CDR3 profiles showed different
CD4+ and CD8+ cells and compared within each patient shapes with 8-13 different classes and 1 or 2 predomi-
(Figure 4B and Online Supplementary Figure S8B). In CD4+ nant peaks of various nt sizes (36-57). For DJ length pro-
cells, all patients displayed CDR3 profiles with normal files in SAA patients, the asymmetric Gaussian distribu-
Gaussian distribution, as described in healthy subjects. In tion was described in all CD4+ cells, and in 6 (AA5-AA8
CD8+ cells, Patients AA7, AA8, and AA10 to AA12 had and AA11-AA12) CD8+ populations. Similarly,
CDR3 size profiles with Gaussian distributions. In the TRBV/TRBJ rearrangement plots from the CD8+CD57+

Figure 6. Homology assessment. (A) CDR3 sequence pools were analyzed among patients (AA) and healthy subjects (HC) for the presence of homology. Shared and
immunodominant sequences were reported as a heatmap based on their relative expression: in the same row, from lowest (gray; <0.01%) to highest (red; >5%) val-
ues. (B) Structural analysis was performed comparing the sequences for common pattern, using both alignments at the N-terminal of Vβ gene (left) or at the C-ter-
minal of Jβ gene (right).

766 haematologica | 2018; 103(5)


TCR repertoire of effector memory T cells in AA

cell pool of AA3 and AA4 showed the same “skyscraper” mean frequency of 0.1±0.2% and 4.7±9.4% in SAA
landscape, but more enriched (Figure 5A), as well as patients, and 0.0003±0.0001% and 0.01±0.002% in
CDR3 size and DJ length profiles that overlapped those healthy subjects. No matches were found in the enriched
in total CD8+ cells, although these were again more CD8+CD57+ T-cell pool (Online Supplementary Table S3).
enriched (Figure 5B).

Discussion
TCR repertoire diversity and shared CDR3 sequences
in SAA patients and healthy subjects The character of oligoclonal expansion of CD8+CD28–
Simpson’s index of diversity was calculated for each lymphocytes in AA, described by Risitano et al.,8,41 strong-
healthy and each SAA subject. This index is used in ecol- ly suggests an antigen driven mechanism of T-cell activa-
ogy for in-depth assessment of the degree of diversity of a tion, ultimately leading to destruction of hematopoietic
system, related to the richness (or number of species pres- stem and progenitor cells. In this study, we focused on a
ent) and evenness (or relative abundance of each).38 Thus, subset of CD8+CD28–CD57+ T lymphocytes, termed effec-
this index assesses the probability that 2 randomly select- tor memory cells because of their high antigen-affinity
ed individuals from a system belong to the same species. and their ability to undergo activation after antigen stimu-
For a TCR repertoire, the index measures the probability lation.27,36 Others have reported the expansion of effector
that 2 CDR3 sequences randomly selected from CD4+ or memory CD8+ T cells in the tumor microenvironment and
CD8+ pools of one subject could be identical.17,38 A value in PB from patients with solid tumors, hematologic malig-
close to 0 means infinite diversity, and a value close to 1 nancies, chronic infections and autoimmune disorders.36,41-
no diversity.36 In our cohort, Simpson’s indexes were sim- 44
In cancers, effector memory T cells appear to have an
ilar in healthy controls and SAA patients for CD4+ important role in immune surveillance; for example, their
(0.990±0.005 vs. 0.991±0.005, respectively; P=0.537) and increase after interferon (IFN)a treatment correlates to
CD8+ cells (0.983±0.013 vs. 0.927±0.084, respectively; better prognosis in melanoma patients,43 and expanded
P=0.060). By paired t-test, Simpson’s indexes in SAA sub- CD8+CD57+ T cells reach normal levels after removal of
jects were significantly different between CD4+ and CD8+ head and neck cancer.36 Higher frequencies of CD8+CD57+
cells (P=0.047) (Online Supplementary Figure S9A). When cells have also been described in SAA and MDS patients,
compared to those indices in CD8+CD57+ cells, decreased which decrease in responders after anti-thymocyte globu-
diversity in the CD8+ effector memory compartment was lin treatment.44 The expansion of effector memory cells
described (total CD4+ cells vs. CD8+CD57+ cells, P<0.0001; could also occur in older healthy subjects as a result of life-
total CD8+ cells vs. CD8+CD57+ cells, P=0.0003) (Online long exposure to common pathogens, but it is related to
Supplementary Figure S9B). reduced overall immune responsiveness to novel
In order to test the hypothesis that an autoreactive clone antigens.45,46 Consistent with previous studies, the SAA
is triggered by autoantigens, CDR3 amino acid sequences patients in our cohort had higher frequencies of
from healthy subjects and patients were screened for CD8+CD57+ cells at diagnosis (25.6% in SAA patients vs.
homology and then compared to public and private TCR 13.3% in healthy subjects). Moreover, patients with CD8+
repertoires reported in literature,39 as described in the effector memory T-cell expansion at diagnosis experi-
Online Supplementary Methods. From analysis of sequences enced shorter PFS. No age-effects were seen for clone size
in the CD8+ cell pool, a total of 29 CDR3 sequences were in either patients or healthy subjects.
shared between patients and controls, but these sequences There is indirect evidence of immune pathophysiology
were highly expressed in SAA patients and also present in of AA, including oligoclonal expansion of effector CD8+ T
their immunodominant clones (Figure 6A). When we lymphocytes.8,41 Clonality of T-cell subsets can be studied
searched for these common sequences in the whole CDR3 by flow cytometry analysis of Vβ usage, CDR3 size spec-
sequence repertoire from CD8+CD57+ cells in AA subjects, tratyping or deep sequencing of VDJ combinations, and
CD8+CD57+ cell pools from AA3 and AA4 showed enrich- CDR3 nucleotide and amino acid sequences.17-20,47 In SAA
ment in frequencies of the immunodominant clones (from patients, expansion of at least one Vβ family in both CD4+
53.61% to 79.12% for AA3 patient and from 23.45% to and CD8+ effector CD28dim cells by flow cytometry with
38.14% for AA4 patient). Structural analysis of shared and polyclonal features in CD4+ and oligoclonal characteristics
immunodominant sequences did not show a pattern of in CD8+ cells has been described.1,8,15,41,44 The CDR3 size
common charged residue among patients (Figure 6B and skewing by spectratyping in the CD8+ population but not
Online Supplementary Methods). Immunodominant and in CD4+ cells confirmed clonality in CD8+ cells.8,21,41 In our
shared CDR3 sequences were compared with CDR3 β current work, we demonstrate by flow cytometry and
repertoires reported in literature for infectious, autoim- deep sequencing that oligoclonal expansion occurs mainly
mune and malignant diseases.39 For confirmation, the in effector memory CD8+ cell compartment, as the
VDJdb database, a database of known antigen specific immunodominant clones were highly enriched in
sequences (https://vdjdb.cdr3.net), was also used. No match- CD8+CD57+ cells and had decreased diversity on deep
es were found for infectious diseases, while 2 sequences sequencing. Effector memory T cells are a circulating T-
present in AA9 were reported previously in AA and parox- cell population that can migrate to the BM under different
ysmal nocturnal hemoglobinuria (PNH).8,40 Lastly, we types of stimulation.30 Vβ usage of peripheral CD8+CD57+
sought to perform homology assessment for reported T cells may mirror that in the BM, as suggested by the
PNH-related clonotypes on the entire TCR repertoire high concordance between PB and BM in our AA patients.
without frequency restriction, as small PNH clones could In our cohort, 75% of SAA patients with effector memory
be present in healthy individuals.40 Two of the reported 12 cell expansion also showed oligoclonal features by deep
CDR3 sequences (CATSRTGGETQYF and CATSRV- sequencing of the total CD8+ cell compartment:
VAGETQYF) were found in our TCR repertoires with a TRBV/TRBJ rearrangement plots with a “skyscraper” pro-

haematologica | 2018; 103(5) 767


V. Giudice et al.

file due to the presence of 1-3 immunodominant clones, by the number of possible epitopes.41,48 For autoimmune dis-
and predominant classes in CDR3 size and DJ length pro- eases, including T-LGLL, clonotypes can be private to a spe-
files. These findings were confirmed by TCR repertoire cific disease because of the unlimited number of possible
sequencing of CD8+CD57+ enriched cells from SAA epitopes.17,41,49 Despite the large diversity of TCR CDR3
patients. Similar deep sequencing features have been sequences, only 29 shared CDR3 sequences were found in
described in whole blood in T-cell large granular lympho- our cohort; these were highly expressed in SAA patients
cyte leukemia (T-LGLL).17 T-LGLL, a chronic lymphopro- and enriched in the CD8+ CD57+ T-cell population. By using
liferation of TCRaβ+CD3+CD5dimCD8+CD57+CD16+ cells sensitive techniques such as deep sequencing, we detected
with monoclonal TCRγ-chain rearrangement and preva- sequences at very low frequencies. However, the finding
lence in the elderly, is frequently associated with autoim- that a group of clonotypes is shared between SAA patients
mune diseases.16,17 Thus, similarities between our SAA and healthy subjects suggests the existence of common epi-
cohort and T-LGLL patients suggest a common patho- topes driving activation of T-cell autologous clones (as also
physiology of expanded autoreactive T lymphocytes. described by Gargiulo et al. in PNH patients40). As chronic
Characterization of long-term Vβ usage has already been transfusion could be the source of antigen exposure, we
proposed as a biomarker of disease progression in T-LGLL investigated its effect on effector memory T-cell expansion
and AA,8,16 given that immunodominant clones can in other hematologic diseases. A comparison between
remerge during relapse.16 However, high heterogeneity in transfused patients and healthy subjects showed no signifi-
the TCR repertoire during immunosuppressive therapies cant variations in CD8+ CD57+ cell frequencies (P=0.216).
has been reported.8,16 In our study, immunodominant Oligoclonal expansion of effector memory CD8+ T cells
clones were longitudinally investigated in 3 AA patients. is frequent in AA and may correlate with prognosis, con-
In Patient 4, expanded clones slightly decreased during sistent with a role of effector memory T cells in BM
treatment but did not disappear, as no hematologic destruction during active disease. Deep sequencing tech-
improvements were observed. In Patient 34, clone 17 nologies allow in-depth characterization of the TCR reper-
remained stable during the course of the disease, while toire, and flow cytometric analysis of Vβ usage may be
clone 13.6 increased at three months and slightly useful to determine diagnosis and prognosis of SAA
decreased at six, concomitantly with a minimal partial patients, and to monitor their clinical course.
response. In Patient 22, clone 2 completely disappeared at
six months of treatment and the patient achieved hemato- Acknowledgments
logic remission; however, at the time of relapse the clone The authors would like to thank Sachiko Kajigaya and
increased again. Our data suggest that Vβ typing of the Keyvan Keyvanfar (Hematology Branch, NHLBI), Ying Rao
CD8+ CD57+ T-cell population by flow cytometry might (BGI Genomics), and Swee Lay Thein (Sickle Cell Branch,
be a useful biomarker to monitor clonal kinetics during the NHLBI) for assistance; Kinneret Broder (Hematology Branch,
course of AA, but a larger cohort of patients and a more NHLBI) for assistance in obtaining healthy volunteer samples;
sensitive technique, such as deep sequencing, are needed and Barbara Weinstein (Hematology Branch, NHLBI) and Jim
to validate the clinical usefulness. Nichols (Sickle Cell Branch, NHLBI) for obtaining patient sam-
Chronic antigen exposure is required to trigger T-cell acti- ples.
vation, as in persistent viral infections or with antigen
spread during autoimmune and malignant diseases.17 For Funding
viral infections, CDR3 homology in public sequence reper- This research was supported by the Intramural Research
toires, and also cross-reactivity between viruses, is limited Program of the NIH, National Heart, Lung, and Blood Institute.

References Rotoli B. Function and malfunction of 10. Sloand EM, Kim S, Maciejewski JP, et al.
hematopoietic stem cells in primary bone Intracellular interferon- in circulating and
1. Young NS, Calado RT, Scheinberg P. Current marrow failure syndromes. Curr Stem Cell marrow T cells detected by flow cytometry
concepts in the pathophysiology and treat- Res Ther. 2007;2(1):39-52. and the response to imunosuppressive ther-
ment of aplastic anemia. Blood. 6. Maciejewski JP, O’Keefe C, Gondek L, Tiu apy in patients with aplastic anemia. Blood.
2006;108(8):2509-2519. R. Immune-mediated bone marrow failure 2002;100:(4)1185-1191.
2. Young NS. Current concepts in the patho- syndromes of progenitor and stem cells: 11. Maciejewski JP, Selleri C, Anderson S,
physiology and treatment of aplastic ane- molecular analysis of cytotoxic T cell clones. Young NS. Fas antigen expression on
mia. Hematology Am Soc Hematol Educ Folia Histochem Cytobiol. 2007;45(1):5-14. CD34+ human marrow cells is induced by
Program. 2013;2013:76-81. 7. Feng X, Scheinberg P, Wu CO, et al. interferon-gamma and tumor necrosis fac-
3. Maciejewski JP, Selleri C, Sato T, Anderson Cytokine signature profiles in acquired tor-alpha and potentiates cytokine-mediat-
S, Young NS. A severe and consistent deficit aplastic anemia and myelodysplastic syn- ed hematopoietic suppression in vitro.
in marrow and circulating hematopoietic dromes. Haematologica. 2011;96(4):602- Blood. 1995;85:(11)3183-3190.
cells (long-term culture-initiating cells) in 606. 12. Selleri C, Maciejewski JP, Sato T, Young NS.
acquired aplastic anemia. Blood. 8. Risitano AM, Maciejewski JP, Green S, Interferon-γ constitutively expressed in the
1996;88(6):1983-1991. Plasilova M, Zeng W, Young NS. In-vivo stromal microenviroment of human mar-
4. Maciejewski JP, Kim S, Sloand E, Selleri C, dominant immune responses in aplastic row cultures mediates potent hematopoietic
Young NS. Sustained long-term hematologic anaemia: molecular tracking of putatively inhibition. Blood. 1996; 87:(10)4149-4157.
recovery despite a marked quantitative pathogenetic T-cell clones by TCR beta- 13. Hirano N, Butler MO, von Bergwelt-Baildon
defect in the stem cell compartment of CDR3 sequencing. Lancet. 2004; MS, et al. Autoantibodies frequently detect-
patients with aplastic anemia after immuno- 364(9431):355-364. ed in patients with aplastic anemia. Blood.
suppressive therapy. Am J Hematol. 9. Young NS. Hematopoietic cell destruction 2003;102:(13)4567-4575.
2000;65(2):123-131. by immune mechanisms in aquired aplastic 14. Feng X, Chuhjo T, Sugimori C, et al.
5. Risitano AM, Maciejewski JP, Selleri C, anemia. Semin Hematol. 2000;37(1):3-14. Diazepam-binding inhibitor-related protein

768 haematologica | 2018; 103(5)


TCR repertoire of effector memory T cells in AA

1: a candidate autoantigen in acquired aplas- Adv Immunol. 2014;122:1-57. lic T cell responses. Cell Res. 2012;22(1):33-
tic anemia patients harboring a minor popu- 26. Arstila TP, Casrouge A, Baron V, Even J, 42.
latoin of paroxysmal nocturnal hemoglobin- Kanellopoulos J, Kourilsky P. A direct esti- 40. Gargiulo L, Lastraioli S, Cerruti G, et al.
uria-type cells. Blood. 2004; 104:(8)2425- mate of the human alphabeta T cell receptor Highly homologous T-cell receptor beta
2431. diversity. Science. 1999;286(5441):958-961. sequences support a common target for
15. Kook H, Risitano AM, Zeng W, et al. 27. Bretschneider I, Clemente MJ, Meisel C, et autoreactive T cells in most patients with
Changes in T-cell receptor VB repertoire in al. Discrimination of T-cell subsets and T- paroxysmal nocturnal hemoglobinuria.
aplastic anemia: effects of different cell receptor repertoire distribution. Blood. 2007;109(11):5036-5042.
immunosuppressive regimens. Blood. Immunol Res. 2014;58(1):20-27. 41. Risitano AM, Kook H, Zeng W, Chen G,
2002;99(10):3668-3675. 28. Araki K, Youngblood B, Ahmed R. The role Young NS, Maciejewski JP. Oligoclonal and
16. Clemente MJ, Wlodarski MW, Makishima of mTOR in memory CD8 T-cell differenti- polyclonal CD4 and CD8 lymphocytes in
H, et al. Clonal drift demonstrates unexpect- ation. Immunol Rev. 2010;235(1):234-243. aplastic anemia and paroxysmal nocturnal
ed dynamics of the T-cell repertoire in T- 29. Appay V, van Lier RA, Sallusto F, Roederer hemoglobinuria measured by V beta CDR3
large granular lymphocyte leukemia. Blood. M. Phenotype and function of human T spectratyping and flow cytometry. Blood.
2011;118(16):4384-4393. lymphocyte subsets: consensus and issues. 2002;100(1):178-183.
17. Clemente MJ, Przychodzen B, Jerez A, et al. Cytometry A. 2008;73(11):975-983. 42. Filaci G, Fenoglio D, Fravega M et al. CD8+
Deep sequencing of the T-cell receptor 30. Farber DL, Yudanin NA, Restifo NP. Human CD28- T regulatory lymphocytes inhibiting
repertoire in CD8+ T-large granular lympho- memory T cells: generation, compartmen- T cell proliferative and cytotoxic functions
cyte leukemia identifies signature land- talization and homeostasis. Nat Rev infiltrate human cancers. J Immunol 2007;
scapes. Blood. 2013;122(25):4077-4085. Immunol. 2014;14(1):24-35. 179:(7)4323-4334.
18. Calis JJ, Rosenberg BR. Characterizing 31. World Medical Association. World Medical 43. Characiejus D, Pasukoniene V,
immune repertoires by high throughput Association Declaration of Helsinki: ethical Jonusauskaite R, et al. Peripheral blood
sequencing: strategies and applications. principles for medical research involving CD8high CD57+ lymphocyte levels may
Trends Immunol. 2014;35(12):581-590. human subjects. JAMA. 2013;310(20):2191- predict outcome in melanoma patients treat-
19. Langerak AW, van Den Beemd R, Wolvers- 2194. ed with adjuvant interferon-alpha.
Tettero IL, et al. Molecular and flow cyto- 32. International Agranulocytosis and Aplastic Anticancer Res. 2008;28:(2B)1139-1142.
metric analysis of the Vbeta repertoire for Anaemia Study. Incidence of aplastic ane- 44. Kook H, Zeng W, Guibin C, Kirby M, Young
clonality assessment in mature mia: the relevance of diagnostic criteria. By NS, Maciejewski JP. Increased cytotoxic T
TCRalphabeta T-cell proliferations. Blood. the International Agranulocytosis and cells with effector phenotype in aplastic ane-
2001;98(1):165-173. Aplastic Anemia Study. Blood. mia and myelodysplasia. Exp Hematol.
20. Wlodarski MW, Gondek LP, Nearman ZP, et 1987;70(6):1718-1721. 2001;29:(11)1270-1277.
al. Molecular strategies for detection and 33. Camitta BM, Thomas ED, Nathan DG, et al. 45. Boucher N, Dufeu-Duchesne T, Vicaut E,
quantitation of clonal cytotoxic T-cell Severe aplastic anemia: a prospective study Farge D, Effros RB, Schächter F. CD28
responses in aplastic anemia and myelodys- of the effect of early marrow transplantation expression in T cell aging and human
plastic syndrome. Blood. 2006;108(8):2632- on acute mortality. Blood. 1976;48(1):63-70. longevity. Exp Gerontol. 1998;33(3):267-
2641. 34. Bolotin DA, Shugay M, Mamedov IZ, et al. 282.
21. Schuster FR, Hubner B, Führer M, et al. MiTCR: software for T-cell receptor 46. Wikby A, Ferguson F, Forsey R, et al. An
Highly skewed T-cell receptor V-beta chain sequencing data analysis. Nat Methods. immune risk phenotype, cognitive impair-
repertoire in the bone marrow is associated 2013;10(9):813-814. ment, and survival in very late life: impact of
with response to immunosuppressive drug 35. Benjamini Y, Yekutieli D. The control of the allostatic load in Swedish octogenarian and
therapy in children with very severe aplastic false discovery rate in multiple testing under nonagenarian humans. J Gerontol A Biol Sci
anemia. Blood Cancer J. 2011;1(3):e8. dependency. Ann Stat. 2001; 29:(4)1165- Med Sci. 2005;60(5):556-565.
22. Schatz DG, Ji Y. Recombination centres and 1188. 47. Fozza C, Barraqueddu F, Corda G, et al.
the orchestration of V(D)J recombination. 36. Strioga M, Pasukoniene V, Characiejus D. Study of the T-cell receptor repertoire by
Nat. Rev. Immunol. 2011;11:(4)251-263. CD8+ CD28- and CD8+ CD57+ T cells and CDR3 spectratyping. J Immunol Methods.
23. Gauss GH, Lieber MR. Mechanistic con- their role in health and disease. 2017;440:1-11.
straints on diversity in human V(D)J recom- Immunology. 2011;134(1):17-32. 48. Zeng W, Maciejewski JP, Chen G, Young NS.
bination. Mol Cell Biol. 1996;16(1):258-269. 37. Nickel RS, Horan JT, Fasano RM, et al. Limited heterogeneity of T cell receptor BV
24. Teng G, Papavasiliou FN. Immunoglobulin Immunophenotypic parameters and RBC usage in aplastic anemia. J Clin Invest.
somatic hypermutation. Annu Rev Genet. alloimmunization in children with sickle cell 2001;108(5):765-773.
2007;41:107-120. disease on chronic transfusion. Am J 49. Le Gal FA, Ayyoub M, Dutoit V, et al.
25. Matthews AJ, Zheng S, DiMenna LJ, Hematol. 2015;90(12):1135-1141. Distinct structural TCR repertoires in natu-
Chaudhuri J. Regulation of immunoglobulin 38. Hill MO. Diversity and evenness: a unifying rally occurring versus vaccine-induced
class-switch recombination: choreography notation and its consequences. Ecology. CD8+ T-cell responses to the tumor-specific
of noncoding transcription, targeted DNA 1973;54:(2)427-432. antigen NY-ESO-1. J Immunother.
deamination, and long-range DNA repair. 39. Li H, Ye C, Ji G, Han J. Determinants of pub- 2005;28(3):252-257.

haematologica | 2018; 103(5) 769


ARTICLE Rec Cell Biology & its Disorders

Safety and efficacy of plerixafor dose


Ferrata Storti
Foundation
escalation for the mobilization of CD34+
hematopoietic progenitor cells in patients
with sickle cell disease: interim results
Farid Boulad,1,2 Tsiporah Shore,3 Koen van Besien,3 Caterina Minniti,4 Mihaela
Barbu-Stevanovic,5 Sylvie Wiener Fedus,6 Fabiana Perna,2 June Greenberg,7
Danielle Guarneri,7 Vijay Nandi,5 Audrey Mauguen,8 Karina Yazdanbakhsh,5
Haematologica 2018 Michel Sadelain2 and Patricia A. Shi4,5

Department of Pediatrics, BMT Service, Memorial Sloan Kettering Cancer Center, New
Volume 103(5):770-777 1

York; 2Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York;
3
Bone Marrow and Hematopoietic Stem Cell Transplant Program, Weill Cornell
Medicine/ New York Presbyterian Hospital, New York; 4Sickle Cell Program, Division of
Hematology, Albert Einstein College of Medicine, Bronx; 5Lindsley F. Kimball Research
Institute, New York Blood Center, NY; 6Laboratory Medicine, Memorial Sloan Kettering
Cancer Center, New York; 7Division of Hematology and Oncology, Weill Cornell Medicine
/New York Presbyterian Hospital, NY and 8Department of Epidemiology and
Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA

ABSTRACT

G
ene therapy for sickle cell disease is limited by the yield of
hematopoietic progenitor cells that can be harvested for transduc-
tion or gene editing. We therefore performed a phase I dose-esca-
lation study of the hematopoietic progenitor cell mobilizing agent pler-
ixafor to evaluate the efficacy and safety of standard dosing on periph-
eral blood CD34+ cell mobilization. Of 15 patients enrolled to date, only
one was chronically transfused and ten were on hydroxyurea. Of eight
patients who achieved a CD34+ cell concentration >30 cells/μL, six were
on hydroxyurea. There was no clear dose response to increasing plerix-
afor dosage. There was a low rate of serious adverse events; two patients
developed vaso-occlusive crises, at the doses of 80 μg/kg and 240 μg/kg.
Correspondence:
pshi@nybc.org Hydroxyurea may have contributed to the limited CD34+ mobilization
by affecting baseline peripheral blood CD34 counts, which correlated
strongly with peak peripheral blood CD34 counts. Plerixafor administra-
Received: December 22, 2017. tion did not induce significant increases in the fraction of activated neu-
trophils, monocytes, or platelets. However, increased neutrophils posi-
Accepted: January 23, 2018. tive for activated β2 integrin and Mac-1 were associated with serious
Pre-published: February 1, 2018. adverse events. In summary, plerixafor was well tolerated but did not
achieve consistent CD34+ cell mobilization in this cohort of patients,
most of whom were being actively treated with hydroxyurea and only
doi:10.3324/haematol.2017.187047 one was chronically transfused. The study will continue with escalation
of the dose of plerixafor and modification of hydroxyurea administra-
Check the online version for the most updated tion. Clinicaltrials.gov identifier: NCT02193191.
information on this article, online supplements,
and information on authorship & disclosures:
www.haematologica.org/content/103/5/770
Introduction
©2018 Ferrata Storti Foundation Autologous gene therapy holds considerable promise for the treatment of
Material published in Haematologica is covered by copyright. patients with sickle cell disease (SCD).1,2 However, its successful application
All rights are reserved to the Ferrata Storti Foundation. Use of requires an adequate number of hematopoietic progenitor cells (HPC) for gene
published material is allowed under the following terms and
conditions:
transfer or gene editing.3 Steady-state bone marrow has been the historical source
https://creativecommons.org/licenses/by-nc/4.0/legalcode. of HPC for SCD gene therapy, but its harvest requires general anesthesia and has
Copies of published material are allowed for personal or inter- been complicated in current gene therapy trials by the need for repeated bone mar-
nal use. Sharing published material for non-commercial pur- row harvests and a high rate of adverse events.4 Granulocyte colony-stimulating
poses is subject to the following conditions:
factor (G-CSF) is a standard method of mobilizing HPC but its use in SCD patients
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- has been associated with vaso-occlusive complications and even death.5 The
mercial purposes is not allowed without permission in writing mechanism of action of G-CSF involves activation of neutrophils,6 and is also asso-
from the publisher. ciated with endothelial cell, platelet, and coagulation system activation,7-11 all of
which may play a crucial role in sickle cell vaso-occlusion.12
In contrast to G-CSF, plerixafor is a bicyclam reversible small molecule inhibitor

770 haematologica | 2018; 103(5)


Safety and efficacy of plerixafor in SCD patients

Table 1. Patients’ characteristics.


Dose level Subject ID Ethnicity Age Gender Genotype Treatment Clinical complications (in addition to ACS)
yrs regimen
1 African- 33 M SS HU 16 mg/kg Avascular necrosis, retinopathy
2 American 21 F SS HU 26 mg/kg ≥ 3 vaso-occlusive crises per year
3 (1*) 29 M SS; thal(1/4) No HU ≥ 3 vaso-occlusive crises per year
80 μg/kg 4 Hispanic 32 M SS; thal (1/4) No HU Deep venous thrombosis, priapism
5 34 M SS; thal (1/4) HU 28 mg/kg Leg ulcers, retinopathy
6 African- 25 F SS; thal (1/4) HU 16 mg/kg Leg ulcers, retinal artery occlusion,
American retinopathy
7 25 F SS HU 25 mg/kg Cerebral aneurysms (2-3 mm)
160 μg/kg 8 (1*) African- 37 M SS HU 27 mg/kg Avascular necrosis, retinopathy
9 American 32 F SS Chronic transfusion Avascular necrosis, iron overload,
– No HU leg ulcers
10 46 M SS; thal (1/4) No HU Leg ulcers, priapism
12 38 M SS HU 17 mg/kg Avascular necrosis, leg ulcers, priapism
240 μg/kg 13 African- 23 F SS HU 27 mg/kg Avascular necrosis, retinopathy
14 American 27 M SS HU 23 mg/kg Avascular necrosis, priapism
3 (2*) 31 M SS- thal (1/4) No HU ≥ 3 vaso-occlusive crises per year
8 (2*) 38 M SS HU 27 mg/kg Avascular necrosis, retinopathy
*Indicates first and second enrollments for indicated subject. ACS: acute chest syndrome,; HU: hydroxyurea.

of the chemokine receptor CXCR4 and prevents binding drug-induced mobilization.23,24 However, Richard et al.
of its ligand CXCL12 or stromal cell derived factor-1a to showed that two of the three SCD patients whose
induce HPC mobilization.13 We hypothesized that plerix- hydroxyurea was withdrawn for 2 weeks developed
afor’s mechanism of action would lead to less marked painful crises following the withdrawal. Given these con-
increases in white blood cell (WBC) counts and therefore siderations, we designed a prospective phase I dose esca-
less cell and coagulation system activation in SCD and lation study of both the safety and efficacy of plerixafor in
supported this with data from a pre-clinical study involv- patients with SCD in which the patients continued on
ing a sickle cell mouse model.14 Nevertheless, the safety of their standard outpatient treatment used for disease con-
plerixafor in SCD patients remains a matter of concern trol. We have completed the dosing cohorts through to the
because of possible activation of WBC and neutrophils standard plerixafor dose of 240 μg/kg and report the inter-
which could still lead to vaso-occlusive complications and im results here.
the risk of early death in SCD.15-19 As CXCR4 is expressed
on most WBC and is involved in the retention of these
cells in bone marrow, a standard dose of plerixafor of 240 Methods
μg/kg increases all major WBC subsets (neutrophils, lym-
phocytes, monocytes) in normal donors about 3- to 4- Study design
fold.20 Notably, however, in SCD patients who received G- This study is conducted under FDA IND 122657, registered in
CSF, not all patients who had highly elevated WBC counts ClinicalTrials.gov as NCT02193191, and approved by the
experienced vaso-occlusive complications, and conversely, Institutional Review Boards of Memorial Sloan Kettering, Weill
not all patients who experienced vaso-occlusive complica- Cornell Medical College and the New York Blood Center. The
tions had highly elevated WBC counts,5 suggesting that study design is a 3 + 3 dose escalation study with six levels of esca-
WBC activation rather than WBC count per se may con- lation: doses of 80, 160, 240, 320, 400, and 480 μg/kg. There are
tribute to vaso-occlusion in SCD. two primary endpoints: (i) efficacy, defined by the achievement of
Another issue of concern is whether enough peripheral a HPC mobilization level of 30 CD34+ cells/μL; and (ii) safety,
blood CD34+ cells can be mobilized in SCD patients with defined by the occurrence of serious adverse events (≥ grade 3)
plerixafor. The mean and median peak CD34+ counts that are at least possibly plerixafor-related (including vaso-occlu-
using plerixafor alone in normal donors are only sive events).
~25/μL.13,20 SCD patients might mobilize particularly well, At any dose level, the occurrence of at least one grade 3 serious
in that SCD patients might have increased circulating HPC adverse event results in the addition of three more patients to the
even at steady state, although more so during a crisis.21,22 initial three-patient dosing cohort. The occurrence of two grade 3
Furthermore, SS and Sβ0 patients tend to be autosplenec- serious adverse events at a particular dose-level signifies that the
tomized, and data from patients with thalassemia showed maximal tolerated dose has been exceeded and that the previous
that splenectomized patients mobilized about twice as dose-level is the maximum tolerated dose. The trial will be
many peripheral blood CD34 cells with plerixafor alone as stopped upon the occurrence of one grade 4 or 5 serious adverse
non-splenectomized patients.23 event at least possibly related to plerixafor. Patients are followed
Another consideration when using plerixafor is whether for adverse events for 1 month after administration of the plerix-
to withhold hydroxyurea, the recommended standard of afor. This design provides the following probabilities of escalation
care for most SCD patients. Hydroxyurea may inhibit based on the true chance of a dose-limiting toxicity at a specific
mobilization and withholding hydroxyurea for 2 weeks dose level:
leads to a degree of spontaneous mobilization that abets True probability of toxicity 0.10 0.20 0.30 0.40 0.50 0.60

haematologica | 2018; 103(5) 771


F. Boulad et al.

Figure 1. Peripheral blood white blood cell, absolute neutrophil, and CD34 cell counts. There is a trend of increasing white blood cell count (WBC, P=0.05) and
absolute neutrophil count (ANC, P=0.03), but not CD34 concentration (P=0.65) with increasing dose of plerixafor. The graphs show the peripheral blood WBC, ANC,
and CD34 concentrations of 15 patients with SCD treated whith 80 (circles), 160 (squares) and 240 (triangles) µg/Kg of plerixafor prior to administration of plerixafor
(PRE), 6-12 h and 20-24 h after plerixafor. Patients on hydroxyurea are represented by filled circles, squares and triangles, patients off hydroxyurea are represented
by open circles, squares and triangles.

Probability of escalation: 0.91 0.71 0.49 0.31 0.17 0.08 Dickinson Biosciences, San Jose, CA, USA) and FACS Diva soft-
For the efficacy endpoint, the dose escalation will continue to ware (BD Biosciences). Samples are stained and analyzed within
480 μg/kg unless all patients at a preceding dose level achieve a 2-12 h of collection using a modification of the International
peripheral blood CD34+ concentration of at least 30 cells/μL. In the Society of Hematotherapy and Graft Engineering (ISHAGE)
present dose-escalation phase, no leukapheresis is performed. If method (see Online Supplementary Methods).
and when the efficacy endpoint is safely reached, the study will
proceed to a leukapheresis phase (including preclinical transduc- CD34+CD38– enumeration
tion and editing) in three patients. Mononuclear cells are isolated from 2 mL peripheral blood by
Eligible subjects are adults with SS or Sβ0 disease, normal renal Ficoll-Hypaque Plus density centrifugation. CD34+ cells are puri-
and liver function, hemoglobin concentration ≥6 g/dL, WBC count fied by positive selection (MidiMACS™ LS Columns, Miltenyi)
≥3,000/μL, absolute neutrophil count (ANC) ≥1,500/μL, and and stained with CD34 (BD PharMingen) and CD38
platelet count of ≥150,000/μL. Eligible subjects are admitted to the (Invitrogen).
Clinical Research Center at Weill Cornell Medical College. A sin-
gle subcutaneous injection of plerixafor (Sanofi-Genzyme) is
administered in the evening between 8-9 pm. The protocol calls Research cell and coagulation activation studies
for peripheral blood sampling at three time points (baseline, 0-2 h Peripheral blood samples drawn at baseline and after 12 h are
prior to plerixafor; peak between 6-12 h after the plerixafor dose; stained within 1 h of collection for activation markers relevant
at the presumed return to baseline between 20-24 h after the to sickle vaso-occlusion12,25-28 and assessed by flow cytometry
dose): for reasons of feasibility and patient comfort issues, the (BD FACSCanto™). For CD16b+ (1D3, Beckman Coulter) neu-
peak sample was consistently drawn at a mean of 12 ± 1 h after trophils: activated β2 integrin (clone 24, abcam), activated Mac-
plerixafor administration and the return to baseline sample at a 1 (CBRM1/5, eBioscience), E-selectin-Fc chimera (724-ES, R&D
mean of 20 ± 0.29 h after the dose. Since patients have pre-existing Systems), L-selectin (DREG-56, eBioscience), Mac-1/CD11b
anemia, for reasons of safety no more than a total of 105 mL of (ICRF44, BD Pharmingen), and LFA-1/CD11a (HI111, BD
blood is drawn at all three time points combined. Pharmingen). For CD14+ (M5E2, BD Pharmingen) monocytes:
tissue factor (HTF-1, BD Pharmingen). For CD41+ (HIP2, BD
Peripheral blood CD34 testing Pharmingen) platelets: CD16b (1D3, Beckman Coulter) and
Flow cytometric evaluation of the collected peripheral blood CD14 (M5E2, BD Pharmingen). The percentages of positive cells
is performed using a FACS Canto flow cytometer (Becton and median fluorescent intensity (MFI) are assessed for each

772 haematologica | 2018; 103(5)


Safety and efficacy of plerixafor in SCD patients

A B C

Figure 2. Correlation between post-plerixafor CD34+ cell counts and baseline cell counts. The CD34+ level at 12 h correlated positively with the baseline level of
CD34+ (P=0.0006) but not baseline levels of absolute neutrophil count (ANC, P=0.66) or white blood cells (WBC, P=0.49). The graphs show the association between
the value of peripheral blood CD34 concentration at 12 h after plerixafor and the baseline values of (A) CD34, (B) ANC and (C) WBC in 15 patients with SCD treated
with 80 (circles), 160 (squares) and 240 (triangles) μg/Kg of plerixafor. Patients on hydroxyurea are represented by filled circles, squares and triangles, patients off
hydroxyurea are represented by open circles, squares and triangles.

patient, with calculation of the absolute number of positive cells Medical Center (New York, USA) and one patient from
by multiplying the percentage of positive cells by the relevant The Mount Sinai Hospital (New York, USA) (Table 1).
number of cells obtained from a concurrent complete blood Two patients enrolled at dose levels 1 and 2 were subse-
count. For plasma studies, whole blood is centrifuged at 2500 quently re-enrolled in the study at a higher plerixafor
rpm for 15 min at 4°C and plasma frozen at ≤ -80°C until batch dose (dose level 3). All subjects had a past history of mod-
testing. The following coagulation system activation markers erate to severe acute chest syndrome, defined by requir-
relevant to sickle vaso-occlusion are tested:26,29 prothrombin frag- ing treatment with simple or exchange transfusion.
ment 1.2 (Enzygnost F1+2, Dade-Behring) and factor VIII (STA®- Importantly, for safety and feasibility, patients were con-
ImmunoDef VIII, Stago). tinued on their standard outpatient treatment being used
to control their disease. Ten of 15 patients were on
Statistical analysis hydroxyurea, with a median HbF level of 12.4% (Online
Absolute concentrations as well as the fold increases (ratio of Supplementary Table S1) and median baseline ANC of
peak at 12 h to baseline absolute concentrations) of standard 4100/μL (Online Supplementary Table S2). Only one of the
clinical as well as research parameters were analyzed. For 15 subjects was receiving chronic transfusion therapy,
patient 8(2), to avoid computing correlations on an infinite with a HbF of 1.2% and HbA of 54%; this patient was
value, we replaced the baseline value of 0 by 0.2 in the analyses also on deferasirox for the treatment of transfusion-relat-
using CD34+ ratio (10/0.2 = 50). When applying non-parametric ed iron overload. HbA was absent in all other patients. In
statistics, the chosen value does not affect the results, as long as the non-transfused patients, HbF levels correlated strong-
it is close to 0. The presence of a trend in increases of CD34+, ly with hemoglobin concentration, hematocrit, and retic-
ANC, and WBC counts (both at 12 h and fold increases) with ulocyte counts. Of nine patients for whom splenic imag-
increasing dose was tested using the non-parametric Cuzick test ing was available, seven had splenic atrophy (Online
for trend. Correlations between baseline values, and values at 12 Supplementary Table S1).
h or fold increases were estimated using the Kendall tau. A dif-
ference in the distribution of 12 h and fold increases according Efficacy of CD34+ mobilization
to the administration of hydroxyurea was investigated using a Absolute WBC counts, neutrophil counts and CD34+
Wilcoxon test. cell concentrations increased from baseline in all patients
For analyses of cell activation and coagulation, Wilcoxon (Figure 1). Absolute monocyte and lymphocyte counts
signed rank paired testing was performed on the combined dose also increased from baseline (Online Supplementary Table
cohorts for differences between 12 h and baseline values, where- S2). Our target goal of mobilizing at least 30 CD34+
as the presence of significant fold differences between dose lev- cells/μL was, however, reached in only 50% of patients
els was examined with the Kruskal-Wallis test. Correlations given the plerixafor dose of 80 μg/kg, 33% of patients
between values were estimated using the Kendall tau. Two- given 160 μg/kg, and 33% of patients given 240 μg/kg.
tailed P values <0.05 were considered statistically significant. Peak ANC (P=0.03) and WBC count (P=0.05), but not
CD34+ cell count (P=0.65), increased with increasing dose
level. As previously reported in healthy donors,13,30,31 there
Results was a strong correlation of peak CD34+ count with base-
line CD34+ concentration (Kendall tau=0.68, P=0.0006)
Patients’ characteristics but no correlation was observed with baseline ANC
Fifteen subjects have been recruited to date for the (Kendall tau=0.09, P=0.66) or baseline WBC count
study at the first three dose levels of 80, 160 and 240 (Kendall tau=0.13, P=0.49) (Figure 2). There was also no
μg/kg. Fourteen patients were enrolled from Montefiore correlation, as previously reported,13 with baseline platelet

haematologica | 2018; 103(5) 773


F. Boulad et al.

A B C

Figure 3. Association of hydroxyurea treatment with post-plerixafor cell counts. The use or not of hydroxyurea was not associated with different levels of CD34+
(P=0.66), absolute neutrophil count (ANC, P=0.66), or white blood cell count (WBC, P=0.57) 12 h after plerixafor. Peripheral blood (A) CD34 concentrations, (B) ANC,
and (C) WBC at 12 h after plerixafor in 15 patients with SCD treated with 80 (circles), 160 (squares) and 240 (triangles) μg/kg of plerixafor. Patients on hydroxyurea
are represented by filled circles, squares and triangles, patients off hydroxyurea are represented by open circles, squares and triangles.

count (Kendall tau=0.30, P=0.13) or donor age (Kendall cytes at the 240 µg/kg dose, the median fold-changes in
tau=0.14, P=0.49). percentage of cells at every dose cohort were ≤1.1, arguing
There was a significant increase in the median CD34+ against generalized plerixafor-mediated cell activation
fold increase with dose (P=0.01) (Online Supplementary (Figure 4A). Median fold increases in absolute numbers of
Figure S1). A trend was observed for ANC ratio, although activated β2 integrin-positive (aβ2+) neutrophils, activated
not statistically significant (1.6-, 1.8-, and 2.1-fold increas- Mac-1-positive (aMac-1+) neutrophils, and TF+ monocytes
es, P=0.08). No trend was seen for WBC ratio (P=0.13). (160 μg/kg, 240 μg/kg) were close to 2 (Figure 4B). There
With the caveat of statistical adjustment for patient 8(2)'s were strong correlations between the fold increase in
baseline CD34+ count of 0/μL, there was no correlation absolute numbers of neutrophils and fold increases in aβ2+
between CD34+ fold increase and baseline CD34+ (Kandall neutrophils (Kendall tau=0.85, P<0.001) and aMac-1+ neu-
tau= -0.32, P=0.11), baseline ANC (Kendall tau=0.19, trophils (Kendall tau=0.46, P=0.02). The absolute numbers
P=0.32), or baseline WBC (Kendall tau=0.22, P=0.25) of aβ2+ and aMac-1+ neutrophils were significantly
(Online Supplementary Figure S2). increased (Online Supplementary Figure S4A,B), and the two
Hydroxyurea was not associated with differences in patients with serious adverse events (gray arrows) had rel-
peak absolute CD34+ concentration (P=0.95), peak ANC atively high absolute numbers of aβ2+ and aMac-1+ neu-
(P=0.59) or peak WBC (P=0.68) concentrations (Figure 3); trophils, albeit not the highest. There was also a significant
or with differences in the fold increases of CD34+ cell increase in plasma prothrombin fragment 1.2 concentra-
(P=0.64), ANC (P=0.12), or WBC (P=0.36) concentrations tions (Online Supplementary Figure S4C), but the two
(Online Supplementary Figure S3). patients with serious adverse events had absolute concen-
In a subset of six patients, CD34+CD38– cells were enu- trations and fold increases at 12 h that were lower than the
merated (Online Supplementary Table S3), showing a medi- median and mean for that measure. Both patients with
an 3-fold increase in CD34+CD38– concentrations at 12 h. serious adverse events had relatively high fold-increases in
L-selectinneg neutrophils and one had a large fold increase in
Safety of plerixafor TF+ monocytes (Figure 5), but their absolute numbers of L-
There were no significant changes in hemoglobin con- selectinneg neutrophils and TF+ monocytes were not partic-
centration, hematocrit, or platelet counts with plerixafor ularly high (Online Supplementary Figure S4D,E). There were
treatment (data not shown, baseline values in Online significant decreases for five parameters: percentage of
Supplementary Table S1). Due to the occurrence of one seri- aβ2+ neutrophils, MFI of aβ2 neutrophils, percentage of TF+
ous adverse event at the 80 μg/kg dose and another one at monocytes, and percentage and absolute number of
the 240 μg/kg dose, an additional three patients were platelet-neutrophil aggregates (Online Supplementary Figure
enrolled at each of these dose levels. The serious adverse S5). There were no significant changes in the MFI of aMac-
events were both pain crises, possibly related to plerixafor 1+, Mac-1, LFA-1, or L-selectin on neutrophils (data not
(Online Supplementary Table S4), but also associated with shown).
other possibly contributory events. Patient 13 with a seri-
ous adverse event had the second highest peak ANC (and
third highest peak WBC count) in the study, but high Discussion
WBC count and ANC were not consistently associated
with serious adverse events. Eight of 15 patients (53%) with SCD treated with pler-
There were no significant differences between dose lev- ixafor reached the peripheral blood CD34 cell target count
els for any of the activation markers of vaso-occlusion test- of at least 30 CD34+ cells/μL, including three of six
ed. With the exception of tissue factor-positive (TF+) mono- patients treated at a dose of 240 μg/kg. This is in contrast

774 haematologica | 2018; 103(5)


Safety and efficacy of plerixafor in SCD patients

Figure 4. Fold changes in the percent-


age of cells expressing activation mark-
ers, according to plerixafor dose. (A)
Other than % TF+ monocytes, median
fold changes in % cells were ≤1.1. (B)
Median fold changes in absolute num-
B ber of cells for TF+ mono, aβ2+ PMN, and
aMac-1+ PMN were closer to 2 than 1.
Fold changes are ratios of 12 h to base-
line values. Horizontal lines represent
medians. Filled shapes represent
hydroxyurea-treated patients, open
shapes represent patients not treated
with hydroxyurea. Gray arrows point to
the values for the two SAE patients who
had serious adverse events. TF+ mono:
tissue-factor-positive monocytes; E-Sel+
PMN: E-selectin-positive neutrophils;
aβ2+ PMN: activated β2 integrin-positive
neutrophils, aMac-1+ PMN: activated
Mac-1-positive neutrophils; L-Selneg PMN:
L-selectin-negative neutrophils; PMA:
platelet-monocyte aggregates; PNA:
platelet-neutrophil aggregates.

with the findings of a recent study by Tisdale et al., in related myelosuppression.24,33 Patient #8, a subject re-
which mobilization was effective in seven of seven SCD enrolled in the study, was particularly instructive regard-
subjects (100%) at a dose of 240 μg/kg.4 It should be noted ing this hypothesis. This patient was clinically stable on
that patients in the National Institutes of Health study hydroxyurea at a dose of 27 mg/kg and was enrolled twice
were off hydroxyurea and had been transfused for at least at an interval of 13 months. At the time of his second
2 months to achieve a HbS <20-30%4,32 while in our study, treatment, however, he had a markedly lower baseline
ten of the 15 patients were on stable doses of hydroxyurea ANC (1900/μL down from 6300/μL) and platelet count
(for at least 1 year) and only one patient was on chronic (217,000/μL down from 400,000/μL), probably related to
transfusion. Although hydroxyurea, a ribonucleotide oscillatory non-toxic hematopoiesis seen in SCD with
reductase inhibitor, causes myelosuppression and was chronic and dose-intensive treatment with hydroxyurea
recently found to reduce CD34 counts in peripheral blood (ANC oscillations between 2,000-6,000/μL as determined
and bone marrow,33 there is no definitive evidence that from review of his clinical laboratory records).38 This
hydroxyurea negatively affects numbers or quality of cell myelosuppression was associated with a baseline CD34
cycle-quiescent hematopoietic stem cells or immature concentration of 0/μL rather than 1/μL, possibly contribut-
bone marrow progenitors as opposed to more mature ing to the relatively low 12 h CD34+ concentration of
myeloid-erythroid progenitors.33-37 Indeed, in our study, 10/μL at the 240 μg/kg dose as compared to 27/μL at the
although we did not achieve consistent efficiency in CD34 160 μg/kg dose. In brief, because hydroxyurea can
cell mobilization, no correlation was found between decrease ANC and platelet count,39 hydroxyurea-related
hydroxyurea use, and absolute or fold increases in CD34+ myelosuppression may have contributed to the relatively
cells/μL. poor CD34+ mobilization obtained in this cohort.
We observed wide inter-donor variability in CD34 However, avoiding hydroxyurea withdrawal might lower
mobilization with plerixafor, as previously reported in the risk of pain crises;24 we, therefore, plan to explore tim-
normal donors (CD34 peaks between 4-157/μL )13 and in ing plerixafor administration to the peak rather than nadir
patients with SCD (CD34 peaks between 50-200/μL).4 of hydroxyurea-related oscillatory hematopoiesis. Finally
However, we also observed a strong correlation between with regards to hydroxyurea therapy, data from the six
baseline CD34+ and peak CD34+ concentrations, as previ- patients in whom we enumerated CD34+CD38– cells sug-
ously reported with both G-CSF and plerixafor mobiliza- gest that hydroxyurea may not adversely affect HSC,
tion in healthy donors (Kendall tau=0.68, P=0.0006).13,30,31 given that all patients except one (patient 10) were on
Factors contributing to baseline CD34 count remain hydroxyurea and a median 3-fold increase at 12 h was
unclear, but our data and others’ suggest that baseline observed. Only 0.2-2.8% of CD34+ cells were CD38-neg-
CD34+ concentration may be affected by hydroxyurea- ative, but this may be consistent with plerixafor’s effect in

haematologica | 2018; 103(5) 775


F. Boulad et al.

normal healthy donors, in that fewer HPC may be mobi- sion may be more effective than continuing standard of
lized by plerixafor than by G-CSF, where up to 50% of G- care. Whether red blood cell transfusion will remain more
CSF-mobilized CD34+ cells are CD38-negative.13,40 effective as we escalate the plerixafor dose (as safety
Because we enrolled only one patient on chronic trans- allows) to 480 μg/kg, with protocol revisions for hydrox-
fusion, we cannot assess any correlation between transfu- yurea-treated patients, is unknown. Finally, potential can-
sion and CD34 mobilization, although notably this didates for SCD gene therapy may not be able to receive
patient had the second highest baseline and highest peak regular red blood cell transfusions (e.g. if they have red cell
CD34+ cell counts in our study. Other studies of plerixafor alloimmunization or a history of hyperhemolysis) or may
in SCD4,32 have initiated chronic transfusion based on the not be willing to do so (e.g. Jehovah Witnesses), even for
hypothesis that the inflammatory nature of SCD affects the relatively short duration of 2-3 months.
the bone marrow and transfusion assuages bone marrow This study has several limitations. Firstly, despite this
inflammation and stress erythropoiesis. Although replica- study being the largest study to date of plerixafor adminis-
tive and oxidative stress of HPC in bone marrow may tration in SCD patients, overall the number of patients
occur,41-44 there is limited evidence that HPC are damaged involved remains small; thus comparisons, for example,
in SCD.36 Five of our patients had HbF-associated increas- between hydroxyurea-treated and non-hydroxyurea-treat-
es in hemoglobin concentration and hematocrit to more ed patients, may not be representative of the actual popu-
than 10 g/dL and 30%, respectively (similar to values in lations. Secondly, we measured WBC, ANC and CD34
chronically transfused patients) but HbF levels did not cor- mobilization in this study only at ~12 and ~20 h after pler-
relate with CD34 cell mobilization. ixafor administration. It is possible that an initial peak
Based on our data, it is possible that continued dose esca- could have occurred at an earlier time (6-9 h) after plerix-
lation could result in greater efficacy of mobilization, since afor and could, therefore, have been missed. Nevertheless,
we observed a dose-related response in the median CD34+ CD34 cell concentrations remain at ~70% of peak levels at
cell fold increase (P=0.01), as also observed in healthy 12 h.20,45,48 Our current study will be amended to include the
donors.45 Patient 3, a repeat enrollment who had never addition of earlier post-plerixafor assessments. Thirdly, we
been on hydroxyurea and was clinically stable, is instruc- determined peripheral blood CD34 cell mobilization in the
tive in that his 12 h peak CD34+ cell count following a pler- 15 patients treated with plerixafor, without performing
ixafor dose of 80 μg/kg was only 8/μL whereas at the dose apheresis. However, there is a well-described correlation
of 240 μg/kg it was 40/μL, even though his baseline CD34+ between peripheral blood CD34 cell concentration and the
cell concentrations (1/μL and then 2/μL) were similar, sug- ultimate CD34 cell dose obtained after apheresis. It is pos-
gesting a dose-response to plerixafor. Notably, his two sible that technical adjustments may be required for this
periods in the study were separated by 19 months, sug- equation in the context of SCD. Finally, other than enu-
gesting that, as with healthy donors,46 intra-individual merating CD34+CD38- cells, we did not further character-
CD34+ cell counts in stable SCD patients not on hydrox- ize CD34+ cells to study “stemness”, for example by deter-
yurea may remain stable over time. Based on these data mining glycophorin A positivity and CD34 dimness.4
and given the safety and continued dose response between CD34+ or CD34+CD38- enumeration is not specific for
240 μg/kg and 480 μg/kg observed in healthy donors,20 we HSC49 and it is, therefore, unclear whether patients had a
plan to continue dose escalation in SCD patients through true increase in HSC, as opposed to more mature lineage-
to the 480 μg/kg dose, barring significant adverse events. committed CD34+ progenitors, which are either mobilized
Adding the CXCR2 agonist, GROβ, might be useful.47 or present in bone marrow.42 We plan to characterize the
Only two of 15 patients (13%) developed serious adverse CD34+ cells further as we move forward in the study,
events as compared to three of seven patients (43%) in the which is currently enrolling at the 320 μg/kg dose level.
study of plerixafor mobilization in SCD by Tisdale et al.,4 Despite mobilization of HSC possibly being less efficient
although this must be qualified by the fact that the patients with plerixafor than with G-CSF, plerixafor-mobilized
in the study by Tisdale et al. also underwent leukapheresis. HSC may have an engraftment advantage over G-CSF-
Our low rate of serious adverse events could, however, also mobilized HSC with regard to better retention of CXCR4,
be due to chronic hydroxyurea therapy and the subsequent which facilitates homing.32
lower WBC and ANC peaks. As the fraction of activated
neutrophils did not increase significantly with plerixafor, Acknowledgments
our low serious adverse event rate may be related to mod- The authors would like to thank our study subjects for their
eration of ANC elevations by hydroxyurea, reducing the participation; the Doris Duke Charitable Foundation for a 2011
absolute number of activated cells. Given the still uncertain Innovation in Clinical Research Award for trial support (to PAS
risks of morbidity, as seen with G-CSF, the use of plerixafor and MS); Sanofi-Genzyme for provision of plerixafor; Jena
in SCD requires further evaluation. Simon for referring one study patient; W. Beau Mitchell for assis-
In summary, our present data suggest that, with regards tance with platelet activation studies; and Henny Billett, Narla
the efficacy of CD34 mobilization, red blood cell transfu- Mohandas, and Beth Shaz for departmental support.

2. Ribeil JA, Hacein-Bey-Abina S, Payen E, et Successful Plerixafor-mediated mobilization,


References al. Gene therapy in a patient with sickle cell apheresis, and lentiviral vector transduction
disease. N Engl J Med. 2017;376 (9):848-855. of hematopoietic stem cells in patients with
1. Mansilla-Soto J, Riviere I, Sadelain M. 3. Sadelain M, Boulad F, Lisowki L, Moi P, severe sickle cell disease. Blood. 2017;130
Genetic strategies for the treatment of sickle Riviere I. Stem cell engineering for the treat- (Suppl 1):990.
cell anaemia. Br J Haematol. 2011;154(6): ment of severe hemoglobinopathies. Curr 5. Fitzhugh CD, Hsieh MM, Bolan CD, Saenz
715-727. Mol Med. 2008;8(7):690-697. C, Tisdale JF. Granulocyte colony-stimulat-
4. Tisdale JF, Pierciey Jr. JF, Kanter J, et al. ing factor (G-CSF) administration in individ-

776 haematologica | 2018; 103(5)


Safety and efficacy of plerixafor in SCD patients

uals with sickle cell disease: time for a mora- randomized crossover trial. Haematologica. dental overdose of hydroxyurea in a young
torium? Cytotherapy. 2009;11(4):464-471. 2017;102(3):600-609. child with sickle cell anemia. Pediatr Blood
6. Petit I, Szyper-Kravitz M, Nagler A, et al. G- 21. Lamming CE, Augustin L, Blackstad M, Cancer. 2012;59(1):170-172.
CSF induces stem cell mobilization by Lund TC, Hebbel RP, Verfaillie CM. 35. Castro O, Nouraie M, Oneal P.
decreasing bone marrow SDF-1 and up-reg- Spontaneous circulation of myeloid-lym- Hydroxycarbamide treatment in sickle cell
ulating CXCR4. Nat Immunol. 2002;3(7): phoid-initiating cells and SCID-repopulating disease: estimates of possible leukaemia risk
687-694. cells in sickle cell crisis. J Clin Invest. and of hospitalization survival benefit. Br J
7. Cella G, Marchetti M, Vignoli A, et al. Blood 2003;111(6):811-819. Haematol. 2014;167(5):687-691.
oxidative status and selectins plasma levels 22. Croizat H, Ponchio L, Nicolini FE, Nagel 36. Brunson A, Keegan THM, Bang H, Mahajan
in healthy donors receiving granulocyte- RL, Eaves CJ. Primitive haematopoietic A, Paulukonis S, Wun T. Increased risk of
colony stimulating factor. Leukemia. progenitors in the blood of patients with leukemia among sickle cell disease patients
2006;20(8):1430-1434. sickle cell disease appear to be endoge- in California. Blood. 2017;130(13):1597-
8. de Haas M, Kerst JM, van der Schoot CE, et nously mobilized. Br J Haematol. 2000;111 1599.
al. Granulocyte colony-stimulating factor (2):491-497. 37. Drasar ER, Jiang J, Gardner K, et al.
administration to healthy volunteers: analy- 23. Yannaki E, Papayannopoulou T, Jonlin E, et Leucocyte telomere length in patients with
sis of the immediate activating effects on cir- al. Hematopoietic stem cell mobilization for sickle cell disease. Br J Haematol. 2014;165
culating neutrophils. Blood. 1994;84(11): gene therapy of adult patients with severe (5):725-727.
3885-3894. beta-thalassemia: results of clinical trials 38. Baird JH, Minniti CP, Lee JM, et al.
9. Falanga A, Marchetti M, Evangelista V, et al. using G-CSF or plerixafor in splenectomized Oscillatory haematopoiesis in adults with
Neutrophil activation and hemostatic and nonsplenectomized subjects. Mol Ther. sickle cell disease treated with hydroxycar-
changes in healthy donors receiving granulo- 2012;20(1):230-238. bamide. Br J Haematol. 2015;168(5):737-746.
cyte colony-stimulating factor. Blood. 24. Richard RE, Siritanaratkul N, Jonlin E, 39. Zimmerman SA, Schultz WH, Davis JS, et
1999;93(8):2506-2514. Skarpidi E, Heimfeld S, Blau CA. Collection al. Sustained long-term hematologic efficacy
10. Spiel AO, Bartko J, Schwameis M, et al. of blood stem cells from patients with sickle of hydroxyurea at maximum tolerated dose
Increased platelet aggregation and in vivo cell anemia. Blood Cells Mol Dis. in children with sickle cell disease. Blood.
platelet activation after granulocyte colony- 2005;35(3):384-388. 2004;103(6):2039-2045.
stimulating factor administration. A ran- 25. Manwani D, Chen G, Carullo V, et al. 40. Worel N, Frank N, Frech C, Fritsch G.
domised controlled trial. Thromb Haemost. Single-dose intravenous gammaglobulin can Influence of plerixafor on the mobilization
2011;105(4):655-662. stabilize neutrophil Mac-1 activation in sick- of CD34+ cell subpopulations and lympho-
11. Canales MA, Arrieta R, Gomez-Rioja R, le cell pain crisis. Am J Hematol. cyte subtypes. Transfusion. 2017;57(9):
Diez J, Jimenez-Yuste V, Hernandez- 2015;90(5):381-385. 2206-2215.
Navarro F. Induction of a hypercoagulability 26. Jakubowski JA, Zhou C, Jurcevic S, et al. A 41. Javazon EH, Radhi M, Gangadharan B, Perry
state and endothelial cell activation by gran- phase 1 study of prasugrel in patients with J, Archer DR. Hematopoietic stem cell func-
ulocyte colony-stimulating factor in periph- sickle cell disease: effects on biomarkers of tion in a murine model of sickle cell disease.
eral blood stem cell donors. J Hematother platelet activation and coagulation. Thromb Anemia. 2012;2012:387385.
Stem Cell Res. 2002;11(4):675-681. Res. 2014;133(2):190-195. 42. Leonard A, Bonifacino A, Dominical VM, et
12. Zhang D, Xu C, Manwani D, Frenette PS. 27. Canalli AA, Proenca RF, Franco-Penteado CF, al. Bone marrow characterization in sickle
Neutrophils, platelets, and inflammatory et al. Participation of Mac-1, LFA-1 and VLA- cell disease: inflammation and stress ery-
pathways at the nexus of sickle cell disease 4 integrins in the in vitro adhesion of sickle thropoiesis lead to suboptimal CD34 recov-
pathophysiology. Blood. 2016;127(7):801- cell disease neutrophils to endothelial layers, ery compared to normal volunteer bone
809. and reversal of adhesion by simvastatin. marrow. Blood. 2017;130(Suppl 1):966.
13. Schroeder MA, Rettig MP, Lopez S, et al. Haematologica. 2011;96(4):526-533. 43. Dallalio G, Brunson CY, Means RT, Jr.
Mobilization of allogeneic peripheral blood 28. Wun T, Styles L, DeCastro L, et al. Phase 1 Cytokine concentrations in bone marrow of
stem cell donors with intravenous plerixafor study of the E-selectin inhibitor GMI 1070 in stable sickle cell anemia patients. J Investig
mobilizes a unique graft. Blood. 2017;129 patients with sickle cell anemia. PLoS One. Med. 2007;55(2):69-74.
(19):2680-2692. 2014;9(7):e101301. 44. Colella MP, Santana BA, Conran N, et al.
14. Choi E, Branch C, Cui MH, et al. No evidence 29. Nur E, van Beers EJ, Martina S, et al. Plasma Telomere length correlates with disease
for cell activation or brain vaso-occlusion levels of pentraxin-3, an acute phase protein, severity and inflammation in sickle cell dis-
with plerixafor mobilization in sickle cell are increased during sickle cell painful crisis. ease. Rev Bras Hematol Hemoter. 2017;39
mice. Blood Cells Mol Dis. 2016;57:67-70. Blood Cells Mol Dis. 2011;46(3):189-194. (2):140-145.
15. Charache S. Mechanism of action of 30. de la Rubia J, Lorenzo JI, Torrabadella M, 45. Liles WC, Broxmeyer HE, Rodger E, et al.
hydroxyurea in the management of sickle Marin P, Insunza A, Sanz MA. Basal Mobilization of hematopoietic progenitor
cell anemia in adults. Semin Hematol. CD34(+) cell count predicts peripheral blood cells in healthy volunteers by AMD3100, a
1997;34(3 Suppl 3):15-21. progenitor cell mobilization and collection CXCR4 antagonist. Blood. 2003;102(8):
16. Castro O, Brambilla DJ, Thorington B, et al. in healthy donors after administration of 2728-2730.
The acute chest syndrome in sickle cell dis- granulocyte colony-stimulating factor. 46. Eidenschink L, DiZerega G, Rodgers K,
ease: incidence and risk factors. The Haematologica. 2004;89(12):1530-1532. Bartlett M, Wells DA, Loken MR. Basal lev-
Cooperative Study of Sickle Cell Disease. 31. Martino M, Gori M, Pitino A, et al. Basal els of CD34 positive cells in peripheral blood
Blood. 1994;84(2):643-649. CD34(+) Cell count predicts peripheral differ between individuals and are stable for
17. Steinberg MH, Barton F, Castro O, et al. blood stem cell mobilization in healthy 18 months. Cytometry B Clin Cytom.
Effect of hydroxyurea on mortality and mor- donors after administration of granulocyte 2012;82(1):18-25.
bidity in adult sickle cell anemia: risks and colony-stimulating factor: a longitudinal, 47. Hoggatt J, Singh P, Tate TA, et al. Rapid
benefits up to 9 years of treatment. JAMA. prospective, observational, single-center, mobilization reveals a highly engraftable
2003;289(13):1645-1651. cohort study. Biol Blood Marrow hematopoietic stem cell. Cell. 2018;172(1-
18. Litos M, Sarris I, Bewley S, Seed P, Okpala I, Transplant. 2017;23(7):1215-1220. 2):191-204.e10.
Oteng-Ntim E. White blood cell count as a 32. Kanter J, Walters MC, Hsieh MM, et al. 48. Lemery SJ, Hsieh MM, Smith A, et al. A
predictor of the severity of sickle cell disease Interim results from a phase 1/2 clinical pilot study evaluating the safety and CD34+
during pregnancy. Eur J Obstet Gynecol study of lentiglobin gene therapy for severe cell mobilizing activity of escalating doses of
Reprod Biol. 2007;133(2):169-172. sickle cell disease. Blood. 2017;128 plerixafor in healthy volunteers. Br J
19. Platt OS, Brambilla DJ, Rosse WF, et al. (22):1176. Haematol. 2011;153(1):66-75.
Mortality in sickle cell disease. Life 33. Uchida N, Fujita A, Hsieh MM, et al. Bone 49. Notta F, Doulatov S, Laurenti E, Poeppl A,
expectancy and risk factors for early death. marrow as a hematopoietic stem cell source Jurisica I, Dick JE. Isolation of single human
N Engl J Med. 1994;330(23):1639-1644. for gene therapy in sickle cell disease: evi- hematopoietic stem cells capable of long-
20. Pantin J, Purev E, Tian X, et al. Effect of high- dence from Rhesus and SCD patients. Hum term multilineage engraftment. Science.
dose plerixafor on CD34(+) cell mobiliza- Gene Ther Clin Dev. 2017;28(3):136-144. 2011;333(6039):218-221.
tion in healthy stem cell donors: results of a 34. Miller ST, Rey K, He J, et al. Massive acci-

haematologica | 2018; 103(5) 777


ARTICLE Red Cell Biology & its Disorders

Plerixafor enables safe, rapid, efficient


Ferrata Storti
Foundation
mobilization of hematopoietic stem cells
in sickle cell disease patients after
exchange transfusion
Chantal Lagresle-Peyrou,1,2,3* François Lefrère,4* Elisa Magrin,1,4*
Jean-Antoine Ribeil,1,4* Oriana Romano,3,5,6 Leslie Weber,2,3,7 Alessandra
Magnani,1,4 Hanem Sadek,1,2,3 Clémence Plantier,1,4 Aurélie Gabrion,1,4
Brigitte Ternaux,1,4 Tristan Félix,3,5 Chloé Couzin,1,4 Aurélie Stanislas,1,4
Jean-Marc Tréluyer,8 Lionel Lamhaut,9,10 Laure Joseph,4 Marianne Delville,2,3,4
Haematologica 2018
Annarita Miccio,3,5# Isabelle André-Schmutz1,2,3# and Marina Cavazzana1,2,3,4#
Volume 103(5):778-786

1
Biotherapy Clinical Investigation Center, Groupe Hospitalier Universitaire Ouest,
Assistance Publique-Hôpitaux de Paris, INSERM CIC 1416, France; 2Laboratory of
Human Lymphohematopoiesis, INSERM UMR 1163, Imagine Institute, Paris, France:
3
Paris Descartes University – Sorbonne Paris Cité, Imagine Institute, France
4
Department of Biotherapy, Necker Children’s Hospital, Assistance Publique-Hôpitaux
de Paris, France; 5Laboratory of Chromatin and Gene Regulation during Development,
INSERM UMR1163, Imagine Institute, Paris, France; 6Department of Life Sciences,
University of Modena and Reggio Emilia, Modena, Italy; 7Paris Diderot University –
Sorbonne Paris Cité, France; 8Mère-Enfant Clinical Investigation Center, Groupe
Hospitalier Necker Cochin, Assistance Publique-Hôpitaux de Paris, France; 9Intensive
Care Unit, Anaesthesia and SAMU de Paris, Necker Hospital, Assistance Publique-
Hôpitaux de Paris, France and 10Paris Descartes University – Sorbonne Paris Cité,
France.
*CLP, FL and EM and JAR contributed equally to this work, in alphabetical order
#
AM, IAS and MC contributed equally to this work

Correspondence:
ABSTRACT
isabelle.andre-schmutz@inserm.fr

S
ickle cell disease is characterized by chronic anemia and vaso-occlu-
sive crises, which eventually lead to multi-organ damage and pre-
Received: November 15, 2017.
mature death. Hematopoietic stem cell transplantation is the only
curative treatment but it is limited by toxicity and poor availability of
Accepted: February 13, 2018. HLA-compatible donors. A gene therapy approach based on the autolo-
Pre-published: February 22, 2018. gous transplantation of lentiviral-corrected hematopoietic stem and pro-
genitor cells was shown to be efficacious in one patient. However, alter-
ations of the bone marrow environment and properties of the red blood
doi:10.3324/haematol.2017.184788 cells hamper the harvesting and immunoselection of patients’ stem cells
from bone marrow. The use of Filgrastim to mobilize large numbers of
Check the online version for the most updated
information on this article, online supplements, hematopoietic stem and progenitor cells into the circulation has been
and information on authorship & disclosures: associated with severe adverse events in sickle cell patients. Thus, broad-
www.haematologica.org/content/103/5/778 er application of the gene therapy approach requires the development of
alternative mobilization methods. We set up a phase I/II clinical trial
whose primary objective was to assess the safety of a single injection of
©2018 Ferrata Storti Foundation Plerixafor in sickle cell patients undergoing red blood cell exchange to
Material published in Haematologica is covered by copyright. decrease the hemoglobin S level to below 30%. The secondary objective
All rights are reserved to the Ferrata Storti Foundation. Use of was to measure the efficiency of mobilization and isolation of
published material is allowed under the following terms and hematopoietic stem and progenitor cells. No adverse events were
conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode. observed. Large numbers of CD34+ cells were mobilized extremely
Copies of published material are allowed for personal or inter- quickly. Importantly, the mobilized cells contained high numbers of
nal use. Sharing published material for non-commercial pur- hematopoietic stem cells, expressed high levels of stemness genes, and
poses is subject to the following conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
engrafted very efficiently in immunodeficient mice. Thus, Plerixafor can
sect. 3. Reproducing and sharing published material for com- be safely used to mobilize hematopoietic stem cells in sickle cell
mercial purposes is not allowed without permission in writing patients; this finding opens up new avenues for treatment approaches
from the publisher. based on gene addition and genome editing. Clinicaltrials.gov identifier:
NCT02212535.

778 haematologica | 2018; 103(5)


Stem cell mobilization in sickle cell patients

Introduction patients,6-7 the recovery of HSPC from SCD patients’ BM


is peculiarly low (M. Cavazzana, unpublished data). In our
Sickle cell disease (SCD) is caused by a point mutation ongoing gene therapy trial (HGB-205, ClinicalTrials.gov
in the coding region of the HBB (β-globin) gene. As a number, NCT02151526), two BM harvests (each requiring
result, an abnormal β-globin protein is incorporated into an exchange transfusion program before general anesthe-
hemoglobin tetramers. These mutant tetramers polymer- sia) were needed to obtain enough cells to transplant the
ize when the local oxygen tension is low. The sickle three SCD patients enrolled.4
hemoglobin (HbS) polymers rigidify red blood cells, Mobilization with granulocyte colony-stimulating fac-
change these cells’ shape, and are responsible for structur- tor (G-CSF, Filgrastim) is widely used to increase the har-
al damage to the red blood cell membrane. In turn, this vest of HSPC (relative to that obtained in a BM aspirate).
modifies the cells’ rheological properties, alters their flow However, attempts to mobilize HSPC with Filgrastim in
in the microcirculation, and thus causes ischemia, stroke, SCD have led to severe adverse events, which hamper the
multi-organ damage, severe acute and chronic pain, and cytokine’s use in this setting. The first report of a severe
chronic hemolytic anemia. Progressive chronic organ com- adverse event following mobilization with low-dose
plications become the main cause of morbidity and mor- Filgrastim (2.5 μg/kg/day) concerned a SCD patient who
tality in the third decade of life.1 SCD is endemic in Africa, developed acute chest syndrome and an elevated white
and the World’s Health Organization considers that 7% of blood cell count (63,400/mm3) as early as 3 days after the
the world population carries the trait. first injection.8 The temporal relationship between
The only curative treatment for SCD is allogeneic Filgrastim administration, the rapid rise in the white blood
hematopoietic stem cell transplantation (HSCT) from cell count and the severe adverse event were strongly sug-
matched sibling donors; the disease-free survival rate 6 gestive of a causal link. Two other severe adverse events
years after transplantation is reportedly >90%.2,3 Given (multi-organ failure and a death) were reported in 2001.9-10
the limited availability of suitable donors and the increase Between 2003 and 2008, eight other patients requiring
in toxicity with age, HSCT is only applied with great cau- autologous HSCT for a malignant hematopoietic disease
tion in adult SCD patients (the main target population for were mobilized with Filgrastim after additional precau-
curative treatment). tions had been taken: reduction of the HbS level to below
We recently demonstrated that gene therapy is applica- 30% via red blood cell exchange, and carefully monitoring
ble to SCD patients, and that the associated toxicity and of peripheral leukocytosis and the blood ion profile.11-13
morbidity rates seem to be lower than those for allogeneic Although all eight patients experienced bone pain, hyper-
HSCT, at least in the first treated patient.4 tension or migraine, no severe adverse events were report-
As is the case with all genetic diseases, the success of ed - providing evidence that SCD patients can be mobi-
gene therapy in SCD relies on several key factors; these lized without any major complications. The median
include the source, quality and number of transduced [interquartile range] circulating CD34+ cell count was
cells, the choice of the conditioning regimen, the level of 24.4/μL [21.2-48.6].12-14
therapeutic transgene expression, and the quality of the As an alternative to Filgrastim, Plerixafor (formerly
bone marrow (BM) microenvironment at the time of har- known as AMD3100) can effectively mobilize HSPC; the
vest and transplantation. It is generally acknowledged that CD34+ cell count in the circulation can reach 15 to 40/μL,
2 to 3x106 CD34+ hematopoietic stem and progenitor cells depending on the dose and the study.15,16 In contrast to
(HSPC)/kg are required for a successful outcome in autol- Filgrastim (which acts by activating monocytes and neu-
ogous HSCT.5 Considering the typical proportion of HSPC trophils in both peripheral blood and the BM), Plerixafor
that can be corrected in gene therapy clinical trials (~50% directly inhibits the binding of stroma-cell-derived factor-
of CD34+ HSPC) and an average recovery of 70% of 1a to its CXC chemokine receptor on HSPC – thus releas-
CD34+ cells post-selection, a minimum harvest of ~6x106 ing stem cells from the BM niches. This mechanistic dif-
CD34+ cells/kg would be required. For reasons that have ference explains the small increase in the white blood cell
not been completely elucidated, as for thalassemic count and the short time interval between Plerixafor

Table 1. Clinical parameters of patients treated with Plerixafor.


SCD Pler 1 (19 years) SCD Pler 2 (20 years) SCD Pler 3 (21 years)
Normal values D-1 D+1 D+7 D+30 D-1 D+1 D+7 D+30 D-1 D+1 D+7 D+30
HbS (HPLC G8) 0% 12·1% 12·6% 15·5% 36·0% 20·2% 15·8% ND 37·1% 6·2%*,** 13·2%** 25·2%** 36·8%**
Total bilirubin (mol/L) 0-17 112 105 109 132 32 35 ND 32 30 30 31 53
Conjugated bilirubin (mol/L) 0-5 8 7 8 9 11 13 ND 12 10 10 10 6
Lactate dehydrogenase (U/L) 125-243 399 409 361 429 566 584 ND 605 378 383 426 501
White blood cells (109/L) 4·0-10·0 9·9 20·1 11·8 13·2 11·9 11·2 ND 6·2 14·0 11·3 9·6 10·4
Neutrophils (109/L) 1·5-7 4·9 14·9 7·8 8·2 7·6 9·2 ND 2·6 9·6 9·3 6·4 6·5
Monocytes (109/L) 0·2-1 2·0 2·4 1·7 2·3 0·9* 0·9 ND 0·7 1·8* 1·3 1·5 1·8
Ferritin (μg/L) 22-275 2441 ND 2674 3111 ND 1009 1473 1962 ND 381 365 446
Liver quantification < 36 340 (±50) 120 (±30) 55 (±30)
(at inclusion period) (μmol/g)
HPLC: High-performance-liquid-chromatography; D: day. ND: not determined. * D+3. **Capillary 3 (and not G8).

haematologica | 2018; 103(5) 779


C. Lagresle-Peyrou et al.

administration and HSPC mobilization. Furthermore, Methods


Plerixafor synergistically augments Filgrastim-induced
mobilization of HSPC,17 and so has been widely combined Study design and human samples
with Filgrastim (i) in Filgrastim-non-responsive patients, This open-label phase I/II trial (ClinicalTrials.gov number,
(ii) as an adjunct to increase the overall recovery of CD34+ NCT02212535) was sponsored by the Assistance Publique
cells for autologous or allogenic HSCT, and (iii) in gene Hôpitaux de Paris. The protocol was reviewed and approved by
therapy trials for β-thalassemia and Wiskott-Aldrich syn- the French Drug Agency (Agence Nationale de Sécurité du
drome.18-20 In view of all the above data, we initiated a Médicament) and the local independent ethics committee (Comité de
phase I/II clinical trial of Plerixafor as a single mobilizing Protection des Personnes Ile-de-France II, Paris, France). The trial was
agent (NCT02212535). Our objective was to demonstrate performed in accordance with the Declaration of Helsinki. Adult
that SCD patients can be safely mobilized under well- SCD patients and healthy donors (for control samples) provided
defined, controlled clinical and biological conditions. their written, informed consent (Online Supplementary Table S1).
Moreover, this clinical trial provided an estimate of the The study was designed to demonstrate the safety and efficacy of
overall recovery of CD34+ HSPC after apheresis in adult the mobilization and harvesting of peripheral HSPC following a
Plerixafor-mobilized SCD patients – thus providing a basis single injection of 0.24 mg/kg Plerixafor in adult SCD patients,
for the wider use of this protocol for gene-addition or with the inclusion criteria described in the Online Supplementary
genome-editing approaches. Methods.

Figure 1. Plerixafor is highly efficient at mobilizing


hematopoietic stem and progenitor cells from sick-
le cell disease patients. (A) Changes in white blood
cell (WBC) and (B) CD34+ hematopoietic stem/prog-
enitor cell (HSPC) counts over the 66 h following
Plerixafor administration in SCD Pler 1 (red
squares), SCD Pler 2 (blue circles) and SCD Pler 3
(green triangles). Arrows indicate time and duration
of apheresis. (C) Number of hematopoietic stem
cells (HSC, black bars) and multipotent progenitors
(MPP, dotted bars) per 1,000 CD34+ cells in sam-
ples of various origins. HD: healthy donor, BM: bone
marrow, SCD: sickle cell disease, Pler: Plerixafor,
Filg: Filgrastim.

780 haematologica | 2018; 103(5)


Stem cell mobilization in sickle cell patients

The patients were carefully monitored in the Intensive Care River Laboratories) were housed in a pathogen-free facility. All
Unit and a few hours before the procedure started, they were experiments were performed in compliance with the French
given prophylaxis for vaso-occlusive crisis [hyperhydration 60 Ministry of Agriculture’s regulations on animal experiments, and
mL/kg/day of a 0.9% saline solution and oxygen therapy (2 were approved by the regional Animal Care and Use Committee
L/min)], as recommended by French guidelines.21 These guidelines (APAFIS#2101-2015090411495178 v4). Six- to 8-week-old mice
recommend oxygen therapy to prevent sickling phenomena in were conditioned with intraperitoneally injected busulfan (Sigma,
particularly stressful conditions. As mobilization can be consid- 25 mg/kg body weight/day) 72 h, 48 h and 24 h before transplan-
ered stressful, we decided to treat all patients with oxygen thera- tation. CD34+ cells (300,000 cells/mouse) from Filgrastim-mobi-
py. Baseline O2 saturation levels were normal and remained stable lized healthy donors or Plerixafor-mobilized cells from the three
during the procedure. The patients received a subcutaneous injec- SCD patients were transplanted into the NSG mice via retro-
tion of 0.24 mg/kg of Plerixafor. We then monitored whole blood orbital sinus injection. Neomycin was added to the animals’ acid-
HbS level, plasma bilirubin, conjugated bilirubin and lactate dehy- ified drinking water. Engraftment was analyzed as described in the
drogenase levels, and white blood cell, neutrophil and monocyte Online Supplementary Methods.
counts for 30 days after the Plerixafor injection. Plerixafor-mobi-
lized HSPC in peripheral blood were collected by using a COBE® Statistical analysis
Spectra Optia apheresis system (Terumo BCT, Lakewood, CO, Statistical tests are indicated in the Figure legends. The thresh-
USA) with modifications (Online Supplementary Material and old for statistical significance was set at P<0.05.
Methods). The apheresis lasted from 4 to 6 h. Product volumes and
total blood volumes are indicated in Table 2. Egress of the CD34+
cells was monitored hourly for at least 24 h after the Plerixafor Results
injection: total CD34+/kg collected and CD34+ collection efficiency
(CE1) [total CD34+ collected/[L processed x (CD34+ pre + CD34+ Patients’ characteristics
post)/2] were evaluated following apheresis (Table 2). Four SCD homozygous patients followed at Necker-
Enfants Malades hospital and meeting our inclusion crite-
Flow cytometry analysis ria were enrolled between May 2015 and January 2017.
Cells were stained with specific antibodies (Online The first one was excluded from the study during the
Supplementary Table S2) and analyzed on a BD FACSCanto™ II sys- enrollment stage because of elevated granulocyte counts
tem, with gating on viable, 7AAD-negative cells. The data were exceeding the limits established in the protocol (<10x109
processed using FlowJo software (version 10.2, Treestar, Ashland, granulocytes/L). The other three patients (P1 - P3) had
OR, USA). been monitored in a reference center after the diagnosis of
SCD within the first 4 years of life. All suffered from
RNA-Seq extraction severe SCD, with a history of acute chest syndrome and
Total RNA was extracted from 0.1-1x106 CD34+ cells using an more than two vaso-occlusive crises per year requiring
RNeasy Micro kit (QIAGEN). RNA-Seq libraries were prepared hospitalization. P1 had undergone cholecystectomy and
from ~10 ng of total RNA, using the Ovation Human FFPE RNA-Seq tonsillectomy. P2 had papillary necrosis and osteonecrosis
Multiplex System kit (Nugen) after DNase treatment (ArcticZymes) of both femoral heads. P3 had undergone cholecystecto-
and >40 million paired-end reads/sample. Samples were generated my, and had chronic asthma and a history of osteomyelitis
on a HiSeq 2500 instrument (Illumina). RNA-Seq analysis was per- events. P1 and P2 were transfused monthly because years
formed as described in the Online Supplementary Methods. of hydroxyurea treatment had proven to be ineffective.
Hydroxyurea treatment was stopped in P3 3 months
Transplantation into non-obese diabetic severe before mobilization. P3 was then transfused monthly until
combined immunodeficiency gamma mice mobilization. In view of iron overload caused by the
The non-obese diabetic severe combined immunodeficiency transfusions (Table 1), treatment with deferasirox (P1) and
gamma (NSG) mice (NOD.CgPrkdcscid Il2rgtm1Wj/SzJ, Charles deferiprone (P2) was ongoing.

Table 2. Characteristics of apheresis and CD34+ immunoselection.


Apheresis SCD Pler 1 SCD Pler 2 SCD Pler 3
Total blood volumes in liters (liters 3.27 (15.8) 4 (21) 4 (17.9)
processed by the apheresis device)
Product volumes (mL) 278 382 322
Hematocrit value (%)* 4.8 5.8 8.2
CE1** 0.24 0.30 0.29
Total CD34+ cells collected by apheresis x106 354 412 292
Total CD34+ cells collected by apheresis x106/kg 4.6 (77) 5.8 (71) 4.5 (65)
of body weight

CD34+ cell product after immunoselection


Recovery 82% 92% 31%
Purity post-selection 95% 94.7% 79.5%
*Normal value: 2-3%; **CE1 = total CD34+ collected/ [L processed X (CD34+pre+CD34+post)/2]

haematologica | 2018; 103(5) 781


C. Lagresle-Peyrou et al.

Absence of Plerixafor-related toxicity numbers of circulating hematopoietic progenitors,22 were


Prior to Plerixafor injection, patients underwent several comparable to those of healthy controls (data not shown).
erythrocytoapheresis sessions in order to decrease their Vital parameters and blood O2 saturation remained stable
HbS levels to below 30% (Table 1). Each patient received and normal during all the procedure. Monitoring of the
a single, subcutaneous injection of Plerixafor (0.24 mg/kg). patients between discharge and the end of the follow-up
The HbS values remained low until day +1 and increased period (6 months after treatment) was uneventful.
to pre-treatment levels thereafter (Table 1). No adverse
effects were observed, other than moderate hypokalemia Efficacy of Plerixafor in mobilizing hematopoietic stem
related to the anticoagulant citrate infusion during the and progenitor cells
apheresis; this was corrected by a 2-day course of potassi- Baseline CD34+ cell counts were 7, 10 and 10/μL in P1,
um chloride (Table 1). The white blood cell and neutrophil P2 and P3, respectively. The patients exhibited a very fast,
counts rose to 30.4 ± 2.8x109/μL and 17.8 ± 3.5x109/L, intense increase in peripheral blood CD34+ cell count,
respectively, during the first 3 h, remained stable and then exceeding 80 CD34+/μL 3 h after the Plerixafor injection
(within 24 to 36 h of Plerixafor injection) returned to pre- (Figure 1B). Levels greater than 50 CD34+/μL were main-
treatment values (Figure 1A and Online Supplementary tained for 6 h, then decreased and returned to normal pre-
Figure S1). Bilirubin, conjugated bilirubin and lactate dehy- treatment values (Figure 1B, and data not shown for day 30
drogenase values and monocyte counts remained stable and day 60). Apheresis was performed with the technical
up to day +30. Serum levels of inflammatory cytokines, adjustments described in the Online Supplementary
including interleukin-8, which is increased in SCD Methods. The quantities of CD34+ cells harvested by
patients during acute crises and associated with higher apheresis (4.6 x 106, 5.8x106 and 4.5x106/kg body weight

A B

Figure 2. Analysis of the transcriptomic profiles of hematopoietic stem and progenitor cells from different sources (A) Hierarchical clustering analysis of HD BM,
SCD BM, SCD Plerixafor-mobilized (Pler), HD Plerixafor-mobilized and HD Filgrastim-mobilized (Filg) HSPC (cluster method: average; distance: correlation). The color
of the sample name indicates the classification. (B) Gene ontology analysis of differentially expressed genes. The most enriched biological process categories are
shown on the y-axis. The x-axis shows sample comparisons, as defined in Table 3. The orange and green color gradients correspond to the statistical significance of
the enrichment [expressed as –log10 (qvalue)] in up- and downregulated genes, respectively. The first color bar at the top indicates comparisons between HSPC from
different types of source (dark red) or the same type of source (light red). The second color bar at the top indicates comparisons between HSPC from different types
of donor (dark blue) or the same type of donor (light blue). (C) Heat map of genes involved in HSC and progenitor biology. A proportion of the HSC markers were highly
expressed in SCD Plerixafor-mobilized HSPC compared with the other samples. The row Z-score is plotted on a red-blue color scale, where red indicates high expres-
sion and blue indicates low expression. The color bar at the top indicates the sample classification. HD: healthy donor; BM: bone marrow; HSPC: hematopoietic stem
and progenitor cells; HSC: hematopoietic stem cells; SCD: sickle cell disease; Pler: Plerixafor; Filg: Filgrastim.

782 haematologica | 2018; 103(5)


Stem cell mobilization in sickle cell patients

for P1, P2 and P3, respectively) were high in all three HSC per 1000 CD34+ cells to be >25 in Plerixafor-mobi-
patients despite a limited collection efficiency (Table 2). lized SCD samples and <5 in all other samples, suggesting
This enabled the cryopreservation of 3x106 unselected that Plerixafor mobilizes HSC with an unexpectedly high
CD34+ cells/kg as a back-up for the upcoming gene thera- efficacy in SCD patients.
py trial (used in the case of absence of engraftment) and The frequency of erythroid and granulocyte/monocyte
the immunoselection and further analyses of mobilized colony-forming cells was similar in Plerixafor-mobilized
CD34+ cells as detailed below. Following CD34+ selection, and BM SCD samples and in the range of that observed in
CD34+ cell purity was in the normal range (Table 2). Filgastrim-mobilized HSPC (Online Supplementary Figure
CD34+ recovery was high in P1 and P2 (82% and 92%, S3A and data not shown). Upon erythroid differentiation,
respectively) and lower in P3 (31%). Plerixafor-mobilized as well as BM SCD HSPC gave rise to
>90% of mature GYPA+CD36lowCD71low enucleated red
Characterization of mobilized hematopoietic stem blood cells (Online Supplementary Figure S3B-D).
and progenitor cells The transcriptome of highly purified CD34+ HSPC from
To determine the hematopoietic differentiation capacity the different sources was analyzed using RNA-Seq.
and self-renewal potential of the mobilized CD34+ cells, Unsupervised gene expression analysis showed that the
we performed a number of phenotypic, transcriptomic samples clustered into two major groups, based on cell
and functional analyses. origin: the “BM” group encompassed BM samples from
Hematopoietic stem cells (HSC) and their immediate SCD patients and healthy donors, whereas the “mobi-
progeny (multipotent progenitors) within the CD34+ sub- lized” group encompassed Plerixafor-mobilized HSPC
set are negative for lineage, CD38, and CD45RA markers from SCD patients and healthy donors and Filgrastim-
and positive for CD13323-25 and can, therefore, be detected mobilized HSPC from healthy donors (Figure 2A).
using flow cytometry. We compared the numbers of HSC Next, we identified differentially expressed genes
and multipotent progenitors among mobilized SCD among the different populations (false discovery rate
CD34+ cells with the values for (i) the BM of healthy <0.05, Table 3). In a comparison of samples from SCD
donors and SCD patients, and (ii) samples from healthy patients and healthy donors, we observed that the genes
donors mobilized with either Filgrastim or Plerixafor upregulated in SCD samples are involved in inflammatory
(Online Supplementary Table S1, Figure 1C and Online and immune responses (e.g. defense response to other
Supplementary Figure S2). We estimated the number of organisms, type I interferon signaling pathway, cytokine

Figure 3. Plerixafor-mobilized CD34+ cells from sickle cell disease patients engraft to the same degree as Filgrastim-mobilized CD34+ cells from healthy donors
in NSG mice. NSG mice were sacrificed 3 to 4 months after the injection of SCD (SCD Plerixafor, n=3) or HD (HD Filgrastim, n=2) CD34+ cells. (A) Bone marrow cells
and (B) splenocytes were isolated, stained and analyzed by flow cytometry. The chimerism (defined as % human CD45+cells/total CD45+cells) and the numbers of
human B lymphocytes (CD19+IgM+), granulocytes (CD11b+CD15+), and monocytes (CD11b+CD14+) were evaluated in each group of mice (red circles and red triangles
SCD Pler1; blue circles and blue triangles SCD Pler2; green circles and green triangles SCD Pler3; the two HD Filg control are represented by gray squares/gray dia-
mond and black squares/black diamonds, respectively). Each dot represents an individual mouse. HD: healthy donor; SCD: sickle cell disease; Pler: Plerixafor; Filg:
Filgrastim.

haematologica | 2018; 103(5) 783


C. Lagresle-Peyrou et al.

production, leukocyte migration, phagocytosis and adap- Table 3. Number of differentially expressed (up- or downregulated)
tive immune response) (Figure 2B). Major differences in genes in HSPC from different sources (false discovery rate < 0.05).
gene expression (>900 differentially expressed genes) Comparison Differentially Upregulated Downregulated
were observed when comparing mobilized and BM sam- expressed
ples (Table 3). The genes downregulated in mobilized ver- SCD BM vs. HD BM 134 52 82
sus BM HSPC are involved in cell cycle-related processes
SCD Pler vs. HD Pler 165 133 32
(e.g. DNA replication, chromosome segregation, and
nuclear division) – confirming that mobilized samples SCD Pler vs. HD Filg 265 188 77
contain more quiescent cells, presumably HSC, than pro- SCD Pler vs. HD BM 1685 761 924
genitors (Figure 2B, Online Supplementary Figure S4A and SCD Pler vs. SCD BM 1544 753 791
Online Supplementary Table S2). Interestingly, mobilized HD Pler vs. HD BM 923 315 608
populations poorly expressed genes that are typical of
committed hematopoietic progenitors, relative to BM HD Filg vs. HD BM 1893 789 1104
samples (Figure 2C, Online Supplementary Figure S4A and HD Pler vs. HD Filg 47 20 27
Online Supplementary Table S2). Conversely, a large propor- HD Filg vs. SCD BM 1683 800 883
tion of the genes involved in HSC biology were strongly HD Pler vs. SCD BM 1024 398 626
expressed in mobilized HSPC (Figure 2C, Online SCD: sickle cell disease. HD: healthy donor. BM: bone marrow. Pler: Plerixafor. Filg:
Supplementary Figure S4A and Online Supplementary Table Filgrastim.
S2). Importantly, some HSC markers (e.g. THY1, HLF and
FLT3) were more strongly expressed in Plerixafor-mobi-
lized SCD samples – confirming the latter’s high HSC con-
tent – than in BM samples and Filgrastim- or Plerixafor- dure.26 Because of the risks incurred by the patients and
mobilized HSPC from healthy donors (Figure 2C). the absence of a direct benefit, the trial was restricted to
Fewer than 300 genes were differentially expressed patients with <10x109 granulocytes/L, knowing their role
between Filgrastim- or Plerixafor-mobilized samples in vaso-occlusive crises.
(Table 3). Of note, genes encoding transcriptional regula- We decided to discontinue hydroxyurea treatment 3
tors and surface markers of plasmacytoid dendritic cell months before the mobilization in P3 and to submit all the
progenitors are upregulated in Plerixafor-mobilized sam- patients to monthly transfusions. The rationale for this
ples compared to Filgrastim-mobilized samples (Online decision was based on the following observations: (i)
Supplementary Figure S4B). FACS analyses showed the hydroxyurea has no beneficial role in CD34+ cell mobiliza-
appearance of a CD133-CD34dim cell population preferen- tion in thalassemic patients;27 (ii) hydroxyurea withdrawal
tially in Plerixafor-mobilized samples (Online is associated with an increase in the number of circulating
Supplementary Figure S2 and data not shown). This popula- CD34+ cells in SCD patients;28 and (iii) in various clinical
tion might contain plasmacytoid dendritic cell progenitors settings hydroxyurea has been associated with myelosup-
that are mobilized by Plerixafor, as recently described.16 pression29,30 suggesting BM toxicity and potential impair-
Proof of stemness was further confirmed by transplan- ment of HSC.
tation into conditioned, immunodeficient mice. Human In order to optimize the safety of the mobilization pro-
chimerism in the BM and spleen was similar in recipients cedure in SCD patients, we reduced HbS levels to below
of Filgrastim-mobilized CD34+ cells from healthy donorss 30% via erythrocyte exchanges, and we closely moni-
and Plerixafor-mobilized CD34+ cells from SCD patients tored white blood cell counts and serum levels of inflam-
(Figure 3A,B). Similar counts of lymphoid and myeloid matory cytokines. Furthermore, the use of Plerixafor
subsets were detected in both groups (Figure 3A,B). The avoided the adverse events associated with Filgrastim:
numbers of human CD34+ cells, HSC and multipotent vascular events and splenic rupture after the administra-
progenitors in bone marrow were comparable between tion of this latter have been extensively reported in clinical
the two groups (Online Supplementary Figure S5). After sec- populations and even in healthy stem cell donors.31
ondary transplantation, all recipients of Plerixafor-mobi- Although these events are usually rare, their frequency
lized SCD samples and Filgrastim-mobilized CD34+ cells may be higher in the presence of other vascular risk fac-
from healthy donors displayed engraftment - further tors (such as SCD).31 Thus the benefit/risk ratio of using a
demonstrating the presence of true HSC in HSPC mobi- hematopoietic growth factor such as Filgrastim (especially
lized with Plerixafor in SCD patients (data not shown). at the high doses required in patients without a malignant
blood disease) appears to be unacceptably low. Hence, we
gave our three patients Plerixafor at the standard dose. No
Discussion adverse events occurred, serum levels of inflammatory
cytokines were in the normal ranges (data not shown) and
Successful transplantation of autologous gene-corrected the white blood cell counts did not exceed 40x109/L (i.e. a
cells primarily depends on the collection and effective value often reported in the literature, and far from the 50
genetic modification of a sufficient number of true stem to 75x109/L often observed in healthy donors after 5 days
cells. Thus, poor harvesting of BM or mobilized peripheral of Filgrastim treatment).32
stem cells limits the success of this procedure. To over- The rapid mobilization with Plerixafor alone (compared
come the need for two or more BM aspirates, we initiated with Filgrastim) is also an important advantage. The cur-
a phase I/II clinical trial with the objective of establishing rent guidelines on mobilization in patients with a malig-
whether Plerixafor-induced stem cell mobilization in SCD nant disease recommend initiating apheresis 11 h after
patients can avoid the increased risk of vaso-occlusive Plerixafor administration;33 this contrasts with the 5 to 7
crises observed with Filgrastim mobilization and of vali- days required for collection after Filgrastim administra-
dating the efficiency of HSPC harvesting with this proce- tion. Moreover, rapid, transient stem cell mobilization by

784 haematologica | 2018; 103(5)


Stem cell mobilization in sickle cell patients

Filgrastim+Plerixafor (especially in very poor mobilizers) immunodeficient NSG mice, in order to establish whether
has been observed by various groups (including ours); the an inflammatory expression profile interfered with
rapid decrease in peripheral blood stem cells may cause engraftment and self-renewal capacity. In fact, the cells
collection to fail when apheresis is initiated according to engrafted as well as Filgrastim-mobilized samples in both
conventional guidelines.34 We monitored the egress of primary and secondary transplantation, demonstrating
CD34+ cells into the blood every hour after Plerixafor that Plerixafor-mobilized SCD CD34+ cells contain true
injection. The time course of CD34+ mobilization was stem cells that are able to reconstitute human
remarkably similar in our three patients. Peak counts of hematopoiesis as well as their Filgrastim-mobilized coun-
over 80 CD34+/μL were achieved as early as 2 to 3 h after terparts. Moreover, it is possible to effectively correct
Plerixafor administration; this confirms that HSPC can be Plerixafor-mobilized SCD HSC by gene addition (manu-
harvested immediately in SCD patients. The decrease in script in preparation). Taken as a whole, our results show
CD34+ cell count observed after 6 h might be the conse- that the inflammatory characteristics of HSC do not
quence of a short-term mobilization of HSC by Plerixafor impair self-renewal and engraftment.
and therefore to the reduced egress of CD34+ cells from In conclusion, the present results show that CD34+ cells
BM combined with their return to the BM. Another possi- can be safely mobilized with Plerixafor in SCD patients
bility is that the drop in CD34+ cells is due to the leuka- under well-defined clinical conditions, including a 3-
pheresis procedure. Pantin et al. analyzed the kinetics of month interruption of hydroxyurea treatment, monthly
CD34+ counts in healthy donors treated with Plerixafor transfusions and red blood cell exchanges. The proportion
and not subjected to the apheresis procedure.35 In their of true stem cells in the Plerixafor-mobilized CD34+ popu-
work, CD34+ cell counts peaked at 6 h at lower values lation was significantly higher than the proportion from
than the ones observed in our study and start to decrease any other source, although their egress must be monitored
10 h after Plerixafor administration, with differences in carefully. After harvesting under specific conditions, the
kinetics and range of mobilization probably related to the cells can be successfully immunoselected. In the case that
clinical conditions of healthy donors versus SCD patients. the optimal dose of CD34+ cells required for gene therapy
Overall, the study by Pantin et al. suggests that leuka- (6-9x106/kg) is not reached after one apheresis and in the
pheresis per se does not cause a decrease in CD34+ cell absence of adverse events, a second mobilization by
counts. Additionally, the limited collection efficiency Plerixafor, 24 h after the first one, will be considered. In
(≤30% of the circulating CD34+ cells) (Table 2) does not our small cohort, the high numbers of cells enabled us to
support the hypothesis that the drop is due to the leuka- extend our gene-addition therapy project without having
pheresis procedure. Close monitoring of peripheral blood to perform several low-yield BM harvesting steps.
CD34+ cell counts is therefore a crucial point for efficient Furthermore, the results removed the obstacle of collect-
apheresis in SCD patients mobilized with Plerixafor. ing an appropriate graft for genome-editing purposes.
The leukapheresis product contained significantly more Lastly, our study emphasizes the importance of consider-
HSC than the other stem cell sources used as controls, i.e. ing the specific characteristics of a diseased BM; finding a
8- to 10-fold more than in BM from healthy donors or way to put the patient’s hematopoietic system into a
SCD patients and in Filgrastim- or Plerixafor-mobilized steady-state condition may circumvent a lack of true stem
cells from healthy donors. Accordingly, HSPC from the cells in the harvested product or poor engraftment of
patients’ Plerixafor-mobilized samples showed elevated genetically-modified cells.
transcription of several HSC-associated genes. We do not
have a formal explanation for this result; we can only Acknowledgments
hypothesize that sickling cycles damage the BM stroma The authors would like to thank Jean-Marc Luby for excellent,
and favor the mobilization of HSC. dedicated technical assistance, Valérie Jolaine, Michaela
Genes involved in inflammatory and immune responses Semeraro for the logistics of the clinical trial, Michaela Semeraro
were upregulated in Plerixafor-mobilized SCD samples. for care of patients, and Christine Bole and Olivier Alibeu for the
The inflammation-related characteristics of HSC and their RNA sequencing. We also thank Frédéric Galacteros, Pablo
environment constitute a major obstacle to both allogene- Bartolucci and Susanne Matthes-Martin for revision of the clini-
ic and autologous transplantation. Significant pathological cal protocol.
changes in hematopoiesis have been described by Weisser
et al.36 in a setting of murine and human chronic granulo- Funding
matous disease, an inherited disease characterized by This work was supported by state funding from the Agence
chronic, sterile, granulomatous inflammation). In mice Nationale de la Recherche as part of the Investissements d’Avenir
and in humans with chronic granulomatous disease, BM program (ANR-10-IAHU-01 and ANR-16-CE18-0004), the
and Filgrastim-mobilized grafts contain a low proportion French National Institute of Health and Medical Research
of HSC; in transplanted mice, this feature is associated (INSERM), and Assistance Publique-Hôpitaux de Paris (AP-
with low reconstitution potential.36 Hence, we transplant- HP). This work was also supported by a grant from the European
ed mobilized CD34+ cells from SCD patients into Research Council (ERC 2015-AdG, GENEFORCURE).

References 2. Walters MC, De Castro LM, Sullivan KM, Sickle cell disease: an international survey
et al. Indications and results of HLA-identi- of results of HLA-identical sibling
cal sibling hematopoietic cell transplanta- hematopoietic stem cell transplantation.
1. Ware RE, de Montalembert M, Tshilolo L, tion for sickle cell disease. Biol Blood Blood. 2017;129(11):1548-1556.
Abboud MR. Sickle cell disease. Lancet. Marrow Transplant. 2016;22(2):207-211. 4. Ribeil JA, Hacein-Bey-Abina S, Payen E, et
2017;390(10091):311-323. 3. Gluckman E, Cappelli B, Bernaudin F, et al. al. Gene therapy in a patient with sickle

haematologica | 2018; 103(5) 785


C. Lagresle-Peyrou et al.

cell disease. N Engl J Med. 2017;376(9): CXCR4 antagonist. Blood. 2003;102(8): moratorium? Cytotherapy. 2009;11(4):464-
848-855. 2728-2730. 471.
5. Cavazzana M, Ribeil JA, Lagresle-Peyrou 16. Schroeder MA, Rettig MP, Lopez S, et al. 27. Yannaki E, Papayannopoulou T, Jonlin E, et
C, Andre-Schmutz I. Gene therapy with Mobilization of allogeneic peripheral blood al. Hematopoietic stem cell mobilization
hematopoietic stem cells: the diseased stem cell donors with intravenous plerix- for gene therapy of adult patients with
bone marrow's point of view. Stem Cells afor mobilizes a unique graft. Blood. severe beta-thalassemia: results of clinical
Dev. 2017;26(2):71-76. 2017;129(19):2680-2692. trials using G-CSF or plerixafor in splenec-
6. Wang TF, Chen SH, Yang SH, Su YC, Chu 17. Broxmeyer HE, Orschell CM, Clapp DW, et tomized and nonsplenectomized subjects.
SC, Li DK. Poor harvest of peripheral al. Rapid mobilization of murine and Mol Ther. 2012;20(1):230-238.
blood stem cell in donors with microcytic human hematopoietic stem and progenitor 28. Richard RE, Siritanaratkul N, Jonlin E,
red blood cells. Transfusion. 2013;53 cells with AMD3100, a CXCR4 antagonist. Skarpidi E, Heimfeld S, Blau CA. Collection
(1):91-95. J Exp Med. 2005;201(8):1307-1318. of blood stem cells from patients with sick-
7. Constantinou VC, Bouinta A, Karponi G, et 18. Devine SM, Flomenberg N, Vesole DH, et le cell anemia. Blood Cells Mol Dis.
al. Poor stem cell harvest may not always al. Rapid mobilization of CD34+ cells fol- 2005;35(3):384-388.
be related to poor mobilization: lessons lowing administration of the CXCR4 29. Charache S, Terrin ML, Moore RD, et al.
gained from a mobilization study in antagonist AMD3100 to patients with mul- Effect of hydroxyurea on the frequency of
patients with beta-thalassemia major. tiple myeloma and non-Hodgkin's lym- painful crises in sickle cell anemia.
Transfusion. 2017;57(4):1031-1039. phoma. J Clin Oncol. 2004;22(6):1095- Investigators of the Multicenter Study of
8. Abboud M, Laver J, Blau CA. 1102. Hydroxyurea in Sickle Cell Anemia. N Engl
Granulocytosis causing sickle-cell crisis. 19. Karponi G, Psatha N, Lederer CW, et al. J Med. 1995;332(20):1317-1322.
Lancet. 1998;351(9107):959. Plerixafor+G-CSF-mobilized CD34+ cells 30. Alvarez-Larran A, Perez-Encinas M, Ferrer-
9. Adler BK, Salzman DE, Carabasi MH, represent an optimal graft source for tha- Marin F, et al. Risk of thrombosis according
Vaughan WP, Reddy VV, Prchal JT. Fatal lassemia gene therapy. Blood. 2015;126(5): to need of phlebotomies in patients with
sickle cell crisis after granulocyte colony- 616-619. polycythemia vera treated with hydrox-
stimulating factor administration. Blood. 20. Ferrari G, Cavazzana M, Mavilio F. Gene yurea. Haematologica. 2017;102(1):103-
2001;97(10):3313-3314. therapy approaches to hemoglobinopathies. 109.
10. Grigg AP. Granulocyte colony-stimulating Hematol Oncol Clin North Am. 2017;31(5): 31. Halter J, Kodera Y, Ispizua AU, et al. Severe
factor-induced sickle cell crisis and multior- 835-852. events in donors after allogeneic
gan dysfunction in a patient with com- 21. Lionnet F, Arlet JB, Bartolucci P, et al. hematopoietic stem cell donation.
pound heterozygous sickle cell/beta+ tha- [Guidelines for management of adult sickle Haematologica. 2009;94(1):94-101.
lassemia. Blood. 2001;97(12):3998-3999. cell disease]. Rev Med Interne. 2009;30 32. Lack NA, Green B, Dale DC, et al. A phar-
11. Onitilo AA, Lazarchick J, Brunson CY, Frei- (Suppl 3):S162-223. macokinetic-pharmacodynamic model for
Lahr D, Stuart RK. Autologous bone mar- 22. Lamming CE, Augustin L, Blackstad M, the mobilization of CD34+ hematopoietic
row transplant in a patient with sickle cell Lund TC, Hebbel RP, Verfaillie CM. progenitor cells by AMD3100. Clin
disease and diffuse large B-cell lymphoma. Spontaneous circulation of myeloid-lym- Pharmacol Ther. 2005;77(5):427-436.
Transplant Proc. 2003;35(8):3089-3092. phoid-initiating cells and SCID-repopulat- 33. To LB, Levesque JP, Herbert KE. How I treat
12. Kamble RT, Tin UC, Carrum G. Successful ing cells in sickle cell crisis. J Clin Invest. patients who mobilize hematopoietic stem
mobilization and transplantation of filgras- 2003;111(6):811-819. cells poorly. Blood. 2011;118(17):4530-
tim mobilized hematopoietic stem cells in 23. Doulatov S, Notta F, Eppert K, Nguyen LT, 4540.
sickle cell-hemoglobin C disease. Bone Ohashi PS, Dick JE. Revised map of the 34. Lefrere F, Mauge L, Rea D, et al. A specific
Marrow Transplant. 2006;37(11):1065- human progenitor hierarchy shows the ori- time course for mobilization of peripheral
1066. gin of macrophages and dendritic cells in blood CD34+ cells after plerixafor injection
13. Rosenbaum C, Peace D, Rich E, Van Besien early lymphoid development. Nat in very poor mobilizer patients: impact on
K. Granulocyte colony-stimulating factor- Immunol. 2010;11(7):585-593. the timing of the apheresis procedure.
based stem cell mobilization in patients 24. Six EM, Bonhomme D, Monteiro M, et al. Transfusion. 2013;53(3):564-569.
with sickle cell disease. Biol Blood Marrow A human postnatal lymphoid progenitor 35. Pantin J, Purev E, Tian X, et al. Effect of
Transplant. 2008;14(6):719-723. capable of circulating and seeding the thy- high-dose plerixafor on CD34(+) cell mobi-
14. Tormey CA, Snyder EL, Cooper DL. mus. J Exp Med. 2007;204(13):3085-3093. lization in healthy stem cell donors: results
Mobilization, collection, and transplanta- 25. Handgretinger R, Kuci S. CD133-positive of a randomized crossover trial.
tion of peripheral blood hematopoietic pro- hematopoietic stem cells: from biology to Haematologica. 2017;102(3):600-609.
genitor cells in a patient with multiple medicine. Adv Exp Med Biol. 2013;777:99- 36. Weisser M, Demel UM, Stein S, et al.
myeloma and hemoglobin SC disease. 111. Hyperinflammation in patients with chron-
Transfusion. 2008;48(9):1930-1933. 26. Fitzhugh CD, Hsieh MM, Bolan CD, Saenz ic granulomatous disease leads to impair-
15. Liles WC, Broxmeyer HE, Rodger E, et al. C, Tisdale JF. Granulocyte colony-stimulat- ment of hematopoietic stem cell functions.
Mobilization of hematopoietic progenitor ing factor (G-CSF) administration in indi- J Allergy Clin Immunol. 2016;138(1):219-
cells in healthy volunteers by AMD3100, a viduals with sickle cell disease: time for a 228.e219.

786 haematologica | 2018; 103(5)


Red Cell Biology & its Disorders ARTICLE

RON kinase inhibition reduces renal


endothelial injury in sickle cell disease mice Ferrata Storti
Foundation

Alfia Khaibullina,1 Elena A. Adjei,1,2 Nowah Afangbedji,1 Andrey Ivanov,1


Namita Kumari,1 Luis E.F. Almeida,3 Zenaide M.N. Quezado,3 Sergei Nekhai1,4,5
and Marina Jerebtsova5

1
Center for Sickle Cell Disease, College of Medicine, Howard University, Washington, DC;
2
Departments of Genetics and Human Genetics, College of Medicine, Howard University,
Washington, DC; 3Department of Perioperative Medicine, NIH Clinical Center, National
Institutes of Health, Bethesda, MD; 4Department of Medicine, College of Medicine,
Howard University, Washington, DC and 5Department of Microbiology, College of Haematologica 2018
Medicine, Howard University, Washington, DC, USA Volume 103(5):787-798

ABSTRACT

S
ickle cell disease patients are at increased risk of developing a chron-
ic kidney disease. Endothelial dysfunction and inflammation asso-
ciated with hemolysis lead to vasculopathy and contribute to the
development of renal disease. Here we used a Townes sickle cell disease
mouse model to examine renal endothelial injury. Renal disease in
Townes mice was associated with glomerular hypertrophy, capillary
dilation and congestion, and significant endothelial injury. We also
detected substantial renal macrophage infiltration, and accumulation of
macrophage stimulating protein 1 in glomerular capillary. Treatment of
human cultured macrophages with hemin or red blood cell lysates sig-
nificantly increased expression of macrophage membrane-associated
protease that might cleave and activate circulating macrophage stimulat-
ing protein 1 precursor. Macrophage stimulating protein 1 binds to and
activates RON kinase, a cell surface receptor tyrosine kinase. In cultured
human renal glomerular endothelial cells, macrophage stimulating pro- Correspondence:
tein 1 induced RON downstream signaling, resulting in increased phos- marina.jerebtsova@howard.edu
phorylation of ERK and AKT kinases, expression of Von Willebrand fac-
tor, increased cell motility, and re-organization of F-actin. Specificity of
macrophage stimulating protein 1 function was confirmed by treatment
Received: September 15, 2017.
with RON kinase inhibitor BMS-777607 that significantly reduced
downstream signaling. Moreover, treatment of sickle cell mice with Accepted: March 1, 2018.
BMS-777607 significantly reduced glomerular hypertrophy, capillary Pre-published: March 8, 2018.
dilation and congestion, and endothelial injury. Taken together, our find-
ings demonstrated that RON kinase is involved in the induction of renal
endothelial injury in sickle cell mice. Inhibition of RON kinase activation doi:10.3324/haematol.2017.180992
may provide a novel approach for prevention of the development of
renal disease in sickle cell disease. Check the online version for the most updated
information on this article, online supplements,
and information on authorship & disclosures:
www.haematologica.org/content/103/5/787

Introduction
©2018 Ferrata Storti Foundation
Sickle cell disease (SCD) is the most commonly inherited hematologic disorder Material published in Haematologica is covered by copyright.
caused by a single nucleotide mutation in the β-globin gene (HBB) resulting in HbS All rights are reserved to the Ferrata Storti Foundation. Use of
published material is allowed under the following terms and
hemoglobin. HbS polymerization leads to sickling and hemolysis of red blood cells conditions:
(RBCs), vaso-occlusion and organ damage. SCD patients are at increased risk of https://creativecommons.org/licenses/by-nc/4.0/legalcode.
developing chronic kidney disease (CKD).1,2 Renal involvement in SCD can be pres- Copies of published material are allowed for personal or inter-
ent in childhood, as evidenced in 16-28% of children with clinical manifestations nal use. Sharing published material for non-commercial pur-
poses is subject to the following conditions:
of proteinuria and microalbuminuria.3 Albuminuria and proteinuria are observed in https://creativecommons.org/licenses/by-nc/4.0/legalcode,
more than 50% of adult SCD patients, and renal failure is developed in about sect. 3. Reproducing and sharing published material for com-
30%.4,5 SCD-associated nephropathy is characterized by tubular dysfunction, mercial purposes is not allowed without permission in writing
which is manifested by inability to concentrate urine, and consequent hypos- from the publisher.
thenuria and polyuria, and glomerular damage. Glomerular abnormalities are char-
acterized by glomerular hypertrophy, expansion of mesangium, thrombotic

Haematologica | 2018; 103(5) 787


A. Khaibullina et al.

microangiopathy, focal segmental glomerulosclerosis chain, biologically inactive pro-MSP1. It is activated by


(FSGS), membranoproliferative glomerulonephritis, and proteolytic cleavage of Arg483-Val484 bond by either
albuminuria.5-8 Two major disease mechanisms of chronic serum proteases or proteases expressed on the cell
kidney disease in SCD have been proposed: 1) hemolysis- surface.18 Pro-MSP1 diffuses into local tissues where it is
endothelial dysfunction leading to vasculopathy; and 2) activated by proteolytic cleavage and plays a role in the
inflammation and hyper-viscosity leading to vaso-occlu- tissue injury or repair.18 Activated MSP1 binds to and acti-
sion.9 Intrarenal RBC hemolysis was suggested to be a trig- vates a cell surface receptor tyrosine kinase, Recepteur
ger for both mechanisms. d'Origine Nantais (RON).19 We hypothesize that endocy-
tosis of RBC lysis products by kidney-infiltrating
Spleen, the physiological site of RBC removal from the macrophages stimulates expression of MT-SP1, which
circulation, is abnormal in SCD patients. The functional then locally activates circulating MSP1. We further
asplenia is likely to increase the rates of intravascular hypothesize that MSP1 binds to RON tyrosine kinase
hemolysis. Sickling of RBCs and intra-organ hemolysis receptor and activates glomerular endothelium in SCD.
stimulate infiltration by circulating monocytes and their dif- We test these hypotheses using a humanized mouse
ferentiation into macrophages. Endocytosis of RBC lysate model of SCD (Townes) which recapitulates several
products affects macrophage phenotypes.10,11 Intravascular hematologic manifestations of human SCD, including
RBC hemolysis also releases lysate products that impair renal vascular occlusion, as well as vascular, tubular and
endothelial function leading to chronic vasculopathy. glomerular changes.20,21 Here we showed that glomerular
Vascular endothelium and monocytes are activated in SCD disease in SCD mice was associated with endothelial
patients, and monocyte numbers are increased.12-14 injury, increase in renal macrophage infiltration, and
Activated macrophages express matriptase-1 (MT-SP1) glomerular MSP1 accumulation. In vitro, treatment of cul-
which is one of the proteases that cleavages and activates tured human macrophages with hemin, a breakdown
circulating macrophage stimulating protein 1 (MSP1).15,16 product of hemoglobin, or RBC lysate significantly
MSP1 was shown to accumulate in glomeruli in the rat increased expression of MT-SP1. In cultured human renal
model of anti-Thy1 glomerular disease; its neutralization glomerular endothelial cells, MSP1 treatment induced
by antibodies reduced serum creatinine and proteinuria, phosphorylation of RON downstream signaling ERK and
and protected rats from glomerular injury.17 MSP1 is a plas- AKT kinases, increased expression of von Willebrand fac-
ma protein secreted by liver and circulated as a single- tor (vWF) and cell motility, and induced re-organization of

A E
P=4x10-17

Figure 1. Renal disease in sickle cell


disease (SCD) mice is characterized
C
F by significant glomeruli hypertrophy
and capillary dilation. (A-D)
Representative pictures of hematoxylin
P=5x10-9
and eosin staining (H&E) (A and B),
and periodic acid–Schiff staining (PAS)
(C and D) of renal sections. Squares
show enlarged areas (B and D). Bar
sizes on microphotographs are
100 μm (A and C) and 40 μm. (B and
D D). (E and F) Quantification of
glomeruli size (E) and capillary size per
glomeruli cross section (F) is per-
formed using CellSens Standard soft-
ware. Five mice per group were used
for each staining. For quantification
graphs, means are shown. Each dot
represents a value obtained from one
glomerulus cross-section. Ctrl: control.

788 haematologica | 2018; 103(5)


Renal accumulation of MSP1 in sickle cell disease

F-actin. RON kinase inhibitor (RONi, BMS-777607) signif- Taken together, these data demonstrated that renal
icantly reduced RON signaling. Moreover, injections of glomerular accumulation of MSP1 and the activation of
RONi in young SCD mice prevented the development of RON kinase were involved in the induction of renal
glomerular disease, substantially reducing glomerular endothelial injury in SCD mice. Inhibition of RON kinase
hypertrophy, capillary dilation and congestion, and activation is a novel approach to prevent CKD develop-
endothelial injury. ment in SCD.

E F
P=5x10-16 P=8x10-9

Figure 2. Renal disease in sickle cell disease (SCD) mice is associated with significant endothelial injury. (A-D) Representative pictures of von Willebrand factor
(vWF) (A and B) and CD34 (C and D) immunostaining (red) of renal sections. Squares show enlarged areas (B and D). Non-specific primary antibodies (Abs) were
used as a negative control. Bar sizes on the microphotographs are 100 μm (A and C) and 40 μm. (B and D). (E and F) Quantification of vWF (E) and CD34 (F) expres-
sion in glomeruli cross sections is performed using ImageJ Fiji version. Five mice per group were used for each staining. For quantification graphs, means are shown.
Each dot represents a value obtained from one glomerulus cross-section. Ctrl: control.

haematologica | 2018; 103(5) 789


A. Khaibullina et al.

Methods System. Townes mice, here referred to as SCD mice, were


obtained from the Jackson Laboratory.
Mice and RONi injections Two-month old mice were injected subcutaneously with 10
The animal protocol was approved by the Institutional Animal mg/kg of body weight of RON inhibitor (RONi, BSM-777607,
Care and Use Committee at the Children's National Health Santa Cruz Scientific) in 2% DMSO daily for 14 consecutive days.

E F
P=4x10 -5
P=3x10-9

Figure 3. Glomerular macrophages infiltration and MSP1 accumulation is increased in sickle cell disease (SCD) mice. (A-D) Representative pictures of
macrophages (F4/80) (A and B, red) and macrophage stimulating protein 1 (MSP1) (C and D, brown) immunostaining of renal sections. Squares show enlarged
areas immunostaining (B and D). Non-specific primary antibodies (Abs) were used as a negative control. Bar sizes on the microphotographs are 100 µm (A and C)
and 40 µm. (B and D). (E and F) Quantification of F4/80 positive macrophages per glomeruli cross section (E) and MSP1 accumulation in the glomeruli (F) is per-
formed using ImageJ Fiji version software. Five mice per group were used for each staining. For quantification graphs, means are shown. Each dot represents a
value obtained from one glomerulus cross-section. Ctrl: control.

790 haematologica | 2018; 103(5)


Renal accumulation of MSP1 in sickle cell disease

Six SCD and 4 control mice were injected with RONi. Similar Results
groups were injected with 2% DMSO (vehicle).
SCD mouse kidneys have increased macrophage
Immunohistochemistry infiltration and MSP1 accumulation in the glomeruli
Paraffin embedded tissue sections were used for immunostain- Kidneys were collected from 4-month old SCD and con-
ing with rat anti-mouse F4/80 (AbD Serotec), rabbit anti-vWF trol mice (n=5 per group) and renal injury was evaluated.
(Dako), rat anti-mouse ICAM (BioLegend), rat anti-mouse CD34 SCD mice develop RBC sickling, anemia, leukocytosis,
(Cedarline), and mouse anti-MSP1 (R&D Systems). AEC and DAB behavioral changes, and multi-organ pathology character-
kits were obtained from Vector Laboratory. Images were acquired istic of SCD patients.21,25,26 Glomerular abnormalities in 4-
with an Olympus 1x51 microscope with an Olympus DP 72 cam- month old SCD mice were characterized by: glomerular
era. Quantification of positive staining was performed using hypertrophy [Figure 1A, B, and E, hematoxylin and eosin
ImageJ Fiji version and CellSens Standard (Olympus) software. (H&E) staining]; expansion of mesangium [Figure 1C and
D, periodic acid Schiff (PAS) staining, and Online
Cell culture Supplementary Figure S1, PCNA staining]; capillary dilation,
Human glomerular endothelial cell line (HGEC) was generated and thickening of capillary loops and glomerular basement
as described22 and maintained in DMEM media supplemented membrane (Figure 1C, D and F, PAS staining). Glomerular
with 10% fetal bovine serum (FBS) and antibiotics (all from capillaries, peritubular cortical capillaries, and capillaries in
Thermo-Fisher). THP1 human monocytic cell line was purchased the medulla and papilla were markedly congested, and
from ATCC and maintained in RPMI-1640 media supplemented sickling of RBCs was observed especially in the medulla
with 10% FBS, antibiotics and 2 μM β-mercaptoethanol (Sigma- and papilla (Online Supplementary Figure S2, H&E staining).
Aldrich). THP-1 cells were differentiated into macrophages by Capillary congestion and dilation together with hemolysis
treatment with 10 nM phorbol 12-myristate 13 acetate (PMA, might induce endothelial cell injury and secretion of von
Sigma-Aldrich) for 48 hours (h). Willebrand Factor (vWF) into the surrounding blood and
sub-endothelium.27,28 Indeed, glomerular capillary expres-
Treatment of THP-1 cells with hemin and RBC lysate sion of vWF was significantly increased in SCD mice
Hemin was obtained from Frontier Scientific. Blood samples (Figure 2A, B and E, immunostaining, red). Murine
were collected from healthy control subjects. RBCs were isolated glomerular capillary showed positive immunostaining for
by centrifugation, frozen at -80°C for 15 min, and then thawed for endothelial marker CD3429 that was significantly increased
lysis. Cellular debris was pelleted by centrifugation, and the in SCD mice (Figure 2C, D and F, immunostaining, red).
hemolysates were stored at -80°C. Differentiated THP-1 cells Because CD34 is also expressed on hematopoietic progen-
were treated either with different concentrations of hemin or itors cells which are increased in the circulation in SCD
hemolysates for 18 h. mice30 these cells may also be a source of increased CD34
staining in the congested capillary. We also observed a sig-
Immunofluorescent staining and western blots nificant renal glomerular and interstitial infiltration of acti-
Rabbit MT-SP1 (Calbiochem) antibody was used for immunos- vated macrophages in SCD mice (Figure 3A, B and E,
taining and western blot (WB) of THP1 cells. HGEC were treated F4/80 immunostaining, arrows, and Online Supplementary
with 1 μM of human recombinant MSP1 (R&D Systems) with or Figure S3). Glomerular infiltrated macrophages were nega-
without RON inhibitor (200 nM) for varying times. WB analysis tive for the inducible nitric oxide synthase (iNOS), a mark-
was performed with rabbit anti-p44/p42 MAPK (Erk1/2), rabbit er of M1 pro-inflammatory macrophage (Online
anti-phospho-p44/p42 Erk1/2, rabbit anti-pan-Akt, and rabbit Supplementary Figure S4). MSP1 was accumulated in SCD
anti-phospho-Akt antibodies (all from Cell Signaling Technology). renal glomerular capillary (Figure 3C, D and F, immunohis-
Mouse anti-β-actin antibodies and phalloidin-FITC conjugate tostaining, brown). High levels of MSP1 accumulation
were obtained from Sigma-Aldrich. were found in 46±8% of glomeruli in SCD mice. In con-
trast, less than 20% of glomeruli demonstrated low levels
Wounding migration assay of MSP1 accumulation in control mice. Together, these
The HGEC monolayers were wounded by strokes across the findings show that renal disease in SCD mice is associated
diameter of the well with 2.5-mm-wide pipet and medium with with significant endothelial injury, macrophage infiltra-
MSP1 (1 μM) was added. The width of the wound was visualized tion, and accumulation of MSP1 in the glomerular capillar-
with pictures taken at pre-treatment and 5 h post treatment. A ies.
total of 20 random measurements were analyzed for each wound
at each time point. Hypoxia, hemin, and red blood cell lysate stimulate
expression of matriptase 1 in human macrophages
Isolation of mouse renal glomeruli and glomeruli MSP1 is produced by the liver and secreted into circula-
permeability assay tion where it is activated by proteolytic cleavage.18
Mouse renal glomeruli were isolated from control mice using a Macrophage membrane-bound matriptase 1 (MT-SP1) is
sieving technique.23 Glomerular permeability was measured by a one of the proteases which activate MSP1 protein.16,31 We
determination of albumin permeability, as described previously24 tested whether renal hypoxia and RBC hemolysis
with slight modification (Online Supplementary Methods). increased expression of MT-SP1. Meta-analysis of the
NCBI Geo database demonstrated a 2-fold increase in MT-
Statistical analysis SP1 expression in human monocyte-derived differentiated
Results were expressed as mean±Standard Deviation. macrophages compared to non-differentiated monocytes
Differences between two groups were compared by unpaired (Figure 4A). Meta analysis also showed that MT-SP1
parametric t-test, between multiple groups by one-way ANOVA expression was further increased in macrophages that
(GraphPad software). Differences between groups were consid- were cultured under hypoxia (Figure 4B). Non-differentiat-
ered significant at P<0.05. ed human pro-monocytic cell line (THP-1 cells) expressed

haematologica | 2018; 103(5) 791


A. Khaibullina et al.

a low level of MT-SP1, which was increased after THP-1 endothelial cell line (HGEC) recently generated in our lab-
differentiation into macrophages by PMA treatment oratory.22 Treatment with 1 μM MSP1 did not induce pro-
(Figure 4C and D, compare lanes 1 and 2). Treatment of liferation of HGEC (Figure 5A). In contrast, MSP1 treat-
THP-1-derived macrophages with hemin or RBC lysate ment significantly increased motility of HGEC in a 2D
further increased MT-SP1 expression (Figure 4C-F). wound assay (Figure 5B and C). F-actin plays a central role
Immunofluorescent staining of THP-1-derived in the endothelial cell motility and permeability.32
macrophages demonstrated membrane and cytoplasmic Inflammatory mediators can alter F-actin formation and
distribution of MT-SP1 (Figure 4G, green) which was sig- distribution.32 In confluent HGEC, F-actin formed mostly a
nificantly increased upon treatment with hemin (10 μM) cortical rim with few stress fibers (Figure 5D, green).
or RBC lysate (Figure 4G). Collectively, hemolysis prod- MSP1 stimulated re-organization of F-actin and formation
ucts (tested in this study) and hypoxia (meta-analysis data) of stress fibers. (Figure 5D, green). Treatment cells with
demonstrated increased expression of MT-SP1 in human specific RON kinase inhibitor (200 nM; RONi) inhibited
macrophages. Higher levels of MT-SP1 expression might MSP1-induced formation of stress fibers and vWF expres-
induce local activation and accumulation of circulating sion (Figure 5D, F-actin and vWF staining). MSP1 treat-
MSP1. ment also increased expression of vWF in HGEC (Figure
5D, red), and this increased expression was inhibited by
MSP1 induces endothelial cell motility and activated RONi (Figure 5D). Next, we examined activation of RON
downstream RON signaling of ERK and AKT kinase signaling in HGEC. Binding of MSP1 to RON leads
To assess the impact of MSP1 on the endothelium, we to phosphorylation of the downstream kinases, ERK and
examined its effect on cultured human glomerular AKT.33 Levels of ERK and AKT phosphorylation in HGEC

A B
P=0.008 P=0.002

C D

Figure 4. Hemin, red blood


cell (RBC) count and hypoxia
treatment increase expres-
sion of matriptase MT-SP1 in
human THP1-derived
macrophages. (A and B)
Meta-analysis of membrane
type serine protease 1 (MT-
SP1) expression in human
primary monocyte-derived
macrophages using Geo
E F database NCBI (A) Data Set
GDS3203 and (B) Data Set
GDS2036. For quantification
graphs, mean and Standard
Deviation (SD) are shown. (C
and E) Western blot of MT-
SP1 in human THP-1-derived
macrophages treated with
different concentrations of
hemin (C) or RBC lysate (D).
β-actin was used for normal-
ization. (D and F)
Quantification of MT-SP1 on
Western blot. For quantifica-
tion graphs, mean and SD
G
are shown. *P<0.05. Results
are representative of three
independent experiments.
(G) Immunostaining of MT-
SP1 in THP-1-derived
macrophages treated with
hemin (10 nM) or RBC lysate
(green). DAPI was used for
nuclear staining. Bar size 40
μm. PMA: phorbol 12-myris-
tate 13-acetate.

792 haematologica | 2018; 103(5)


Renal accumulation of MSP1 in sickle cell disease

A B C

P=0.003

E F

G H I J

Figure 5. MSP1 treatment of cultured human glomerular endothelial cell line (HGEC) induces cell motility and vWF expression, F–actin re-organization, and RON
kinase signaling in HGEC. (A) Cell growth measured by MTT assay. Five wells are used for either control or treatment with 1 μM of recombinant MSP1 in each exper-
iment. Results are representative of three independent experiments. (B) Representative picture of wound migration assay of control cells and cells treated with 1
μM of MSP1. Bar size 300 μm. (C) Quantification of wound migration assay. Three wells are used per treatment in each experiment. Results are representative of
three independent experiments. (D) Immunostaining of F-actin (green) and vWF (red) in control and treated MSP1 (1 μM) treated cells with or without RONi (200
nM). DAPI (for F-actin) and Hematoxylin (for vWF) were used for nuclear staining. Non-specific primary antibodies were used for negative control. Bar size 40 μm for
F-actin and 100 μm for vWF. (E and F) Western blots of phosphorylated and non-phosphorylated forms of ERK and AKT kinases in HGEC. (E) Cells were treated with
MSP1 (1 μM) and collected at different time points after treatment. (F) Cells were treated with MSP1 (1 μM) with or without RON inhibitor (RONi, 200 nM) for 30
min. (G-J) Quantification of pERK and pAKT on Western blot. For quantification graphs, mean and SD are shown. *P<0.05. Results are representative of three inde-
pendent experiments. β-actin used for loading normalization.

haematologica | 2018; 103(5) 793


A. Khaibullina et al.

Figure 6. MSP1 increases glomerular perme-


P=1x10-5
ability in whole glomeruli assay. (A)
Representative pictures of glomeruli isolated
from control mice placed in isooncotic and
P=0.008
hypooncotic solutions. Glomeruli were pre-
treated with vehicle (PBS), or with MSP1 (1
μM) with or without RON inhibitor (RONi 200
nM). Bar size 40 μm. (B) Quantification of
glomerular volume change in hypooncotic
solution. CellSens Standard software was
used for measurement of the glomeruli area
and glomeruli volume was calculated. Percent
of volume changes in isooncotic solution is
shown. For quantification, graphs means are
shown. Each dot represents a value obtained
from one glomerulus.

were significantly increased after MSP1 treatment (Figure increased glomerular permeability for albumin, and RONi
5E-G). Phosphorylation of ERK and AKT was significantly reduced MSP1-associated glomerular permeability.
reduced after treatment with RONi (Figure 5H-J). Taken
together, MSP1 activated RON receptor signaling in cul- Treatment of SCD mice with RONi significantly
tured HGEC leading to the re-organization of F-actin and ameliorates glomerular endothelial injury
increasing cell motility and vWF expression. To test whether RONi prevents development of
glomerular endothelial injury in young SCD mice, 2-
MSP1 increases permeability in SCD mouse glomeruli month old SCD mice were injected with either RONi (10
Glomerular hyperfiltration is an early stage manifesta- mg/kg of body weight in 2% DMSO) or vehicle (2%
tion in the development of SCD glomerulopathy.34,35 To DMSO, n=6 per group) subcutaneously for 14 days.
test whether MSP1 increases glomeruli permeability, we Control non-SCD mice were also injected with either
utilized a mouse whole glomeruli permeability assay.24 RONi or vehicle (n=4 per group). We used younger (2-
Renal glomerular permeability measurement was based month old) mice to test whether RONi prevents develop-
on the determination of albumin permeability. In the ment of glomerular disease, because older (4-month old)
absence of capillary filtration, the diffusional loss of albu- mice had already developed profound glomerular disease
min from glomeruli capillary is negligible.24 Placement of (Figures 1 and 2). Only 20% of 2-month old mice devel-
intact glomeruli into the hypooncotic solution increases oped microalbuminuria (1 of 5 mice), and 40% of 12-week
glomeruli volume due to water accumulation inside old non-treated SCD mice developed microalbuminuria (2
glomeruli according to oncotic gradient.24 Treatment of of 5 mice) (data not shown). In contrast, we found that all
glomeruli with permeating agents that increase glomerular non-treated mice had glomerular endothelia cell injury at
filtration significantly increases albumin loss and reduces 12 weeks of age. Because endothelial injury was detected
oncotic gradient, leading to a reduced glomerular volume before the onset of albuminuria, we focused on the role of
in the hypooncotic solution.24 Renal glomeruli were isolat- RON signaling in the development of endothelial injury.
ed from control mice and treated with recombinant MSP1 Administration of RONi in SCD mice did not affect body
(1 μM) in the presence or absence of RONi (200 nM) for weight (Figure 7A), but significantly reduced kidney size
30 min (see Methods for details). PBS was used as a vehicle (Figure 7B and C). Moreover, administration of RONi in
control. Placement of vehicle-treated glomeruli in hypoon- SCD mice significantly reduced glomerular hypertrophy
cotic solution significantly increased glomerular volume and capillary congestion (Figure 8A and E and Online
by more than 3.5-fold (Figure 6A and B). MSP1 treatment Supplementary Figure S5, H&E staining). Capillary dilation
reduces glomerular volume, whereas RONi treatment and thickening of capillary loops and glomerular basement
restored the ability of glomeruli to enlarge in the hypoon- membrane were significantly reduced in SCD mice treat-
cotic solution (Figure 6A and B). Taken together, pre-treat- ed with RONi compared to mice with vehicle injection
ment of mouse glomeruli with MSP1 significantly (Figure 8B and F and Online Supplementary Figure S5, PAS

794 haematologica | 2018; 103(5)


Renal accumulation of MSP1 in sickle cell disease

staining). RONi treatment did not affect mesangial expan- A


sion in SCD mice (Figure 8A and B, H&E and Online
Supplementary Figure S6, PAS staining). However, RONi
administration in SCD mice significantly reduced capillary
expression of vWF (Figure 8C and G and Online
Supplementary Figure S5, immunostaining, red), and
Intercellular Adhesion Molecule 1 (ICAM1) (Figure 8D
and E and Online Supplementary Figure S5, immunostain-
ing, red), which are the markers of endothelial injury.36,37
Thus, inhibiton of RON receptor in SCD mice significant-
ly ameliorated development of endothelial and glomerular
injury. B

Discussion
Renal disease in SCD patients includes a variety of
glomerular and tubular complications, but the mechanism C P=0.04
of their development is not fully understood. The most
important finding in our study is that MSP1 and its recep-
tor RON kinase may play a role in the activation of renal
endothelium and development of glomerular pathology in
SCD mice. Moreover, treatment of mice with RONi, an
inhibitor of RON kinase, significantly ameliorated
glomerular hypertrophy, capillary dilation and congestion,
and endothelial injury. These findings reveal a previously
unknown mechanism which contributes to glomerular
endothelial injury in SCD.
Sickle cell disease mice spontaneously develop FSGS
Figure 7. Treatment of sickle cell disease (SCD) mice with RON inhibitor
reduces kidney hypertrophy. (A) Treatment of SCD mice with RON inhibitor
that may be directly associated with RBC sickling and
chronic hemolysis. The focal nature of glomerulosclerosis (RONi) does not affect body weight (BW). (B) Representative picture of kidneys
in SCD mice and SCD patients apparently excludes the of controls and SCD mice treated either with (RONi) or vehicle (2% DMSO). (C)
effect of global factors, such as hypoxia, cell-free heme, Quantification of kidney weight. Kidney weights (KW) to BW ratios are shown.
iron, and other circulating factors that would lead to a
global and not focal glomerulosclerosis with involvement
of only 50% of glomeruli in SCD mice. Thus, locally-pro-
duced factor(s) are more likely to contribute to the devel-
opment of FSGS. MSP1.15 MSP1 expression has been found in the renal
In agreement with a previous report,21 we demonstrate tubular cells.43,44 In agreement with previous studies, we
here that macrophage infiltration in renal glomeruli was did not observe MSP1 expression or accumulation in
increased in SCD mice. Sickling and adhesion of RBCs, glomeruli of control mice. In contrast, we found that
and accumulation of RBC lysate products within the kid- MSP1 was accumulated in approximately 46±8% of renal
ney might stimulate renal infiltration of monocyte-derived glomerular capillaries in SCD mice. This accumulation
macrophages. Monocyte-derived renal macrophages are rate was similar to the percentage of injured glomeruli in
present in all forms of kidney disease with inflammation, SCD mice. Glomerular accumulation of MSP1 was previ-
and renal capillary macrophage infiltration is a character- ously shown in the rat model of anti-Thy1 glomerular dis-
istic pathology of FSGS.38 In many human biopsy studies, ease, and the neutralization of MSP1 by the injected anti-
number of glomerular or interstitial macrophages correlate bodies reduced serum creatinine and proteinuria, and pro-
with poor outcomes, suggesting their possible role in the tected animals from glomerular injury.17 However, the
disease progression.39,40 However, the role of infiltrating mechanism of MSP1-associated glomerular injury was not
macrophages in the progression of renal disease is not well clarified. RON is expressed in human renal tubular cells
understood. Phagocytosis of senescent sickled RBCs, RBC and glomerular mesangial cells.43,44 MSP1 treatment
exosomes and endocytosis of cell-free hemoglobin induces growth, motility and collagen invasion of mesan-
increases inflammatory response in the cultured human gial cells.43 Expression of functional RON in endothelial
monocytes/macrophages.12 Activation of proteases is a cells is unknown. MSP1 that is accumulated in glomerular
universal inflammatory response in macrophages.41 We capillary of SCD mice may potentially affect endothelial
demonstrate here that products of RBC hemolysis signifi- cells, or leak from capillary to affect mesangial cells or
cantly increased expression of macrophage membrane- podocytes. The increased glomeruli size associated with
bound protease, MT-SP1 in cultured human macrophages. dilated glomerular capillary and mesangial proliferation
Meta-analysis data also showed that hypoxia and mono- was reported both in SCD patients and mouse model of
cyte differentiation increased MT-SP1 expression in pri- SCD.20,21,45 In our study, inhibition of RON in SCD mice
mary human macrophages. MT-SP1 is a type II membrane significantly reduced glomerular hypertrophy, as well as
serine protease that plays important roles in cell migration capillary dilation and congestion without reduction of
and tumor cell metastasis.42 mesangial expansion. It is possible that the short time of
MT-SP1 is one of the proteases that activate circulating treatment was not enough to produce statistically signifi-

haematologica | 2018; 103(5) 795


A. Khaibullina et al.

cant differences in mesangial cell proliferation, or that fac- and kidney epithelial cells.18,46 We demonstrated that
tors other than MSP1 could stimulate mesangial growth MSP1 treatment increased motility of HGECs and induced
in SCD mice. Thus, we hypothesized that MSP1 played a formation of F-actin stress fibers that is essential for motil-
role in the activation of endothelial cells. The effect of ity and permeability of endothelial cells.32 Further studies
MSP1 on renal glomerular endothelial cells is still not are needed to determine whether MSP1/RON signaling
known. induces permeability of cultured endothelial cells.
To test the effect of MSP1 on endothelium, we used cul- Interestingly, MSP1 increased vWF expression levels in
tured HGEC. MSP1 had previously been shown to stimu- HGEC. High levels of vWF were also found in SCD
late motility of murine resident peritoneal macrophages murine glomeruli. RONi treatment reduced endothelial

E F G H

P=3x10-8 P=2x10-7 P=0.002 P=0.04

Figure 8. Treatment of sickle cell disease (SCD) mice with RON inhibitor (RONi) reduces renal endothelial injury. Representative pictures of renal sections of control
and SCD mice treated with either RONi or vehicle (DMSO) are shown. (A) Hematoxylin&Eosin (H&E) staining. (B) Periodic Acid-Schiff (PAS) staining. (C) von Willebrand
factor (vWF) immunostaining (red). (D) Intercellular Adhesion Molecule (ICAM) immunostaining (red). Bar sizes 50µm. (E and F) Quantification of glomeruli size (E)
and capillary size per glomeruli cross section (F) is performed using CellSens Standard software. (G and H) Quantification of vWF (G) and ICAM (H) expression in
glomeruli cross sections is performed using ImageJ Fiji version software. Six mice per group were used for each staining. Each dot represents a value obtained from
one glomerulus cross-section. For quantification graphs, means are shown.

796 haematologica | 2018; 103(5)


Renal accumulation of MSP1 in sickle cell disease

vWF levels in vivo and in vitro. vWF mediates adhesion of in the development of glomerular injury.
sickle RBCs to endothelial cells, inducing oxidant stress, The present study established for the first time that
and increasing expression of ICAM-1, VCAM and E- inhibition of RON kinase significantly ameliorates SCD
selectin.47 Indeed, RONi treatment in SCD mice was asso- renal pathology in mice. Short-term inhibition of RON
ciated with reduced vWF levels in glomerular capillaries, kinase reduced endothelial injury in young SCD mice dur-
and decreased RBC adhesion and ICAM-1 levels. ing the early stage of renal disease before the onset of
Therefore, the mechanism of RON inhibition may be albuminuria. We do not know how long this effect per-
associated with prevention of glomerular capillary conges- sists after RONi withdrawal. Whether short-term inhibi-
tion, leading to improvement of renal hemodynamics. A tion of RON kinase will ameliorate already developed
correlation between an alteration of renal hemodynamics renal disease in older mice is currently under investigation.
and a renal histological injury has been demonstrated in a Future studies will elucidate a role of Ron kinase in renal
model of renal ischemia in SCD.48 In addition, our results disease in SCD patients. These findings also highlight a
demonstrate that MSP1 treatment of glomeruli isolated new potential therapeutic target for CKD in SCD. A
from control mice significantly increased albumin perme- recent pre-clinical study50 and Phase 1 clinical trial (clinical-
ability that was effectively prevented by RON inhibition. trials.gov identifier: 01721148) of BMS-777607 RON
Hyperfiltration is associated with glomerular hypertrophy inhibitor for treatment of human cancers demonstrated its
and glomerulosclerosis.49 We demonstrate a significant safety and good tolerability, providing a proof of principal
reduction in glomerular size in SCD mice after treatment for the potential use of this pharmacological approach.
with RON inhibitors, suggesting a possible change in
hyperfiltration. Reduction of glomerular size after 14-day Funding
treatment with RONi is unlikely to be due to structural This work was supported in part by CHaRM pilot grant
remodeling or reduced proliferation of endothelial cells. awarded to MJ through NIH grant 1P50HL118006 (to SN) and
To the best of our knowledge, this is the first study NIH grants 1R01HL125005 (to SN), 5G12MD007597 (to
demonstrating function of MSP1/RON in the glomerular SN), and R41MD008829-01 (to ZMQ). The content is solely
endothelial cells. Future studies will clarify the mechanism the responsibility of the authors and does not necessarily represent
of MSP1-associated endothelial cell activation and its role the official views of the National Institutes of Health.

References bacterial function. Infect Immun. 1983; chain. J Biol Chem. 1997;272(27):16999-
40(3):917-923. 17004.
1. Hamideh D, Alvarez O. Sickle cell disease 11. Vinchi F, Costa da Silva M, Ingoglia G, et 20. Ryan TM, Ciavatta DJ, Townes TM.
related mortality in the United States al. Hemopexin therapy reverts heme- Knockout-transgenic mouse model of sick-
(1999-2009). Pediatr Blood Cancer. induced proinflammatory phenotypic le cell disease. Science. 1997;278(5339):
2013;60(9):1482-1486. switching of macrophages in a mouse 873-876.
2. Elmariah H, Garrett ME, De Castro LM, et model of sickle cell disease. Blood. 21. Kasztan M, Fox BM, Speed JS, et al. Long-
al. Factors associated with survival in a 2016;127(4):473-486. Term Endothelin-A Receptor Antagonism
contemporary adult sickle cell disease 12. Belcher JD, Marker PH, Weber JP, Hebbel Provides Robust Renal Protection in
cohort. Am J Hematol. 2014;89(5):530-535. RP, Vercellotti GM. Activated monocytes Humanized Sickle Cell Disease Mice. J Am
3. Becton LJ, Kalpatthi RV, Rackoff E, et al. in sickle cell disease: potential role in the Soc Nephrol. 2017;28(8):2443-2458.
Prevalence and clinical correlates of activation of vascular endothelium and 22. Jerebtsova M, Kumari N, Obuhkov Y,
microalbuminuria in children with sickle vaso-occlusion. Blood. 2000;96(7):2451- Nekhai S. Adenoviral E4 gene stimulates
cell disease. Pediatr Nephrol. 2010;25(8): 2459. secretion of pigmental epithelium derived
1505-1511. 13. Klug PP, Kaye N, Jensen WN. Endothelial factor (PEDF) that maintains long-term sur-
4. Saraf SL, Zhang X, Kanias T, et al. cell and vascular damage in the sickle cell vival of human glomerulus-derived
Haemoglobinuria is associated with chron- disorders. Blood Cells. 1982;8(1):175-184. endothelial cells. Mol Cell Proteomics.
ic kidney disease and its progression in 14. Mahoney DH Jr, Fernbach DJ. Monocyte 2012;11(11):1378-1388.
patients with sickle cell anaemia. Br J functions in sickle cell disorders. Am J 23. Misra RP. Isolation of glomeruli from mam-
Haematol. 2014;164(5):729-739. Pediatr Hematol Oncol. 1983;5(4):409-411. malian kidneys by graded sieving. Am J
5. Guasch A, Navarrete J, Nass K, Zayas CF. 15. Bhatt AS, Welm A, Farady CJ, et al. Clin Pathol. 1972;58(2):135-139.
Glomerular involvement in adults with Coordinate expression and functional pro- 24. Savin VJ, Sharma R, Lovell HB, Welling DJ.
sickle cell hemoglobinopathies: Prevalence filing identify an extracellular proteolytic Measurement of albumin reflection coeffi-
and clinical correlates of progressive renal signaling pathway. Proc Natl Acad Sci cient with isolated rat glomeruli. J Am Soc
failure. J Am Soc Nephrol. 2006;17(8):2228- USA. 2007;104(14):5771-5776. Nephrol. 1992;3(6):1260-1269.
2235. 16. Qiu D, Owen K, Gray K, Bass R, Ellis V. 25. Wang L, Almeida LE, de Souza Batista CM,
6. Sharpe CC, Thein SL. Sickle cell nephropa- Roles and regulation of membrane-associ- et al. Cognitive and behavior deficits in
thy - a practical approach. Br J Haematol. ated serine proteases. Biochem Soc Trans. sickle cell mice are associated with pro-
2011;155(3):287-297. 2007;35(Pt 3):583-587. found neuropathologic changes in hip-
7. Scheinman JI. Sickle cell disease and the 17. Rampino T, Soccio G, Gregorini M, et al. pocampus and cerebellum. Neurobiol Dis.
kidney. Nat Clin Pract Nephrol. 2009;5(2): Neutralization of macrophage-stimulating 2015;85:60-72.
78-88. protein ameliorates renal injury in anti-thy 26. Khaibullina A, Almeida LE, Wang L, et al.
8. Tejani A, Phadke K, Adamson O, et al. 1 glomerulonephritis. J Am Soc Nephrol. Rapamycin increases fetal hemoglobin and
Renal lesions in sickle cell nephropathy in 2007;18(5):1486-1496. ameliorates the nociception phenotype in
children. Nephron. 1985;39(4):352-355. 18. Leonard EJ. Biological aspects of sickle cell mice. Blood Cells Mol Dis.
9. Potoka KP, Gladwin MT. Vasculopathy and macrophage-stimulating protein (MSP) and 2015;55(4):363-372.
pulmonary hypertension in sickle cell dis- its receptor. Ciba Found Symp. 1997;212: 27. Sporn LA, Marder VJ, Wagner DD.
ease. Am J Physiol Lung Cell Mol Physiol. 183-191. Inducible secretion of large, biologically
2015;308(4):L314-324. 19. Wang MH, Julian FM, Breathnach R, et al. potent von Willebrand factor multimers.
10. Hand WL, King-Thompson NL. Effect of Macrophage stimulating protein (MSP) Cell. 1986;46(2):185-190.
erythrocyte ingestion on macrophage anti- binds to its receptor via the MSP beta 28. Wagner DD, Bonfanti R. von Willebrand

haematologica | 2018; 103(5) 797


A. Khaibullina et al.

factor and the endothelium. Mayo Clin 1990;70(2):427-451. promote tubular regeneration after acute
Proc. 1991;66(6):621-627. 37. Wertheimer SJ, Myers CL, Wallace RW, injury. J Am Soc Nephrol. 2008;19(10):
29. Kairaitis LK, Wang Y, Gassmann M, Tay Parks TP. Intercellular adhesion molecule-1 1904-1918.
YC, Harris DC. HIF-1alpha expression fol- gene expression in human endothelial cells. 45. Nath KA, Hebbel RP. Sickle cell disease:
lows microvascular loss in advanced Differential regulation by tumor necrosis renal manifestations and mechanisms. Nat
murine adriamycin nephrosis. Am J Physiol factor-alpha and phorbol myristate acetate. Rev Nephrol. 2015;11(3):161-171.
Renal Physiol. 2005;288(1):F198-206. J Biol Chem. 1992;267(17):12030-12035. 46. Zhang K, Yao HP, Wang MH. Activation of
30. Blouin MJ, De Paepe ME, Trudel M. 38. Cao Q, Harris DC, Wang Y. Macrophages RON differentially regulates claudin
Altered hematopoiesis in murine sickle cell in kidney injury, inflammation, and fibro- expression and localization: role of claudin-
disease. Blood. 1999;94(4):1451-1459. sis. Physiology. 2015;30(3):183-194. 1 in RON-mediated epithelial cell motility.
31. Kawaguchi M, Orikawa H, Baba T, 39. Hill GS, Delahousse M, Nochy D, Mandet Carcinogenesis. 2008;29(3):552-559.
Fukushima T, Kataoka H. Hepatocyte C, Bariety J. Proteinuria and tubulointersti- 47. Sultana C, Shen Y, Rattan V, Johnson C,
growth factor activator is a serum activator tial lesions in lupus nephritis. Kidney Int. Kalra VK. Interaction of sickle erythrocytes
of single-chain precursor macrophage-stim- 2001;60(5):1893-1903. with endothelial cells in the presence of
ulating protein. FEBS J. 2009;276(13):3481- 40. Rollino C, Basolo B, Roccatello D, endothelial cell conditioned medium
3490. Menegatti E, Piccoli G. Atypical presence of induces oxidant stress leading to
32. Prasain N, Stevens T. The actin cytoskele- antimyeloperoxidase antibodies in 2 trans- transendothelial migration of monocytes.
ton in endothelial cell phenotypes. planted patients. Nephron. 1993;63(4):480. Blood. 1998;92(10):3924-3935.
Microvasc Res. 2009;77(1):53-63. 41. Tomlinson ML, Garcia-Morales C, Abu- 48. Juncos JP, Grande JP, Croatt AJ, et al. Early
33. Wagh PK, Peace BE, Waltz SE. Met-related Elmagd M, Wheeler GN. Three matrix met- and prominent alterations in hemodynam-
receptor tyrosine kinase Ron in tumor alloproteinases are required in vivo for ics, signaling, and gene expression follow-
growth and metastasis. Adv Cancer Res. macrophage migration during embryonic ing renal ischemia in sickle cell disease. Am
2008;100:1-33. development. Mech Dev. 2008;125(11- J Physiol Renal Physiol. 2010;298(4):F892-
34. Haymann JP, Stankovic K, Levy P, et al. 12):1059-1070. 899.
Glomerular hyperfiltration in adult sickle 42. Darragh MR, Bhatt AS, Craik CS. MT-SP1 49. Brenner BM, Lawler EV, Mackenzie HS.
cell anemia: a frequent hemolysis associat- proteolysis and regulation of cell-microen- The hyperfiltration theory: a paradigm
ed feature. Clin J Am Soc Nephrol. vironment interactions. Front Biosci. shift in nephrology. Kidney Int.
2010;5(5):756-761. 2008;13:528-539. 1996;49(6):1774-1777.
35. Aygun B, Mortier NA, Smeltzer MP, 43. Rampino T, Collesi C, Gregorini M, et al. 50. Bieniasz M, Radhakrishnan P, Faham N, De
Hankins JS, Ware RE. Glomerular hyperfil- Macrophage-stimulating protein is pro- La O JP, Welm AL. Preclinical Efficacy of
tration and albuminuria in children with duced by tubular cells and activates mesan- Ron Kinase Inhibitors Alone and in
sickle cell anemia. Pediatr Nephrol. gial cells. J Am Soc Nephrol. 2002;13(3): Combination with PI3K Inhibitors for
2011;26(8):1285-1290. 649-657. Treatment of sfRon-Expressing Breast
36. Pober JS, Cotran RS. Cytokines and 44. Cantaluppi V, Biancone L, Romanazzi GM, Cancer Patient-Derived Xenografts. Clin
endothelial cell biology. Physiol Rev. et al. Macrophage stimulating protein may Cancer Res. 2015;21(24):5588-5600.

798 haematologica | 2018; 103(5)


Myeloproliferative Disorders ARTICLE

The KIT and PDGFRA switch-control inhibitor


DCC-2618 blocks growth and survival Ferrata Storti
Foundation
of multiple neoplastic cell types in
advanced mastocytosis
Mathias Schneeweiss,1,2 Barbara Peter,1,2 Siham Bibi,3 Gregor Eisenwort,1,2
Dubravka Smiljkovic,1 Katharina Blatt,1,2 Mohamad Jawhar,4 Daniela Berger,2
Gabriele Stefanzl,2 Susanne Herndlhofer,1,2 Georg Greiner,5 Gregor Hoermann,5
Emir Hadzijusufovic,1,2,6 Karoline V. Gleixner,1,2 Peter Bettelheim,7 Klaus Geissler,8
Wolfgang R. Sperr,1,2 Andreas Reiter,4 Michel Arock3 and Peter Valent1,2
Haematologica 2018
Volume 103(5):799-809
1
Ludwig Boltzmann Cluster Oncology, Medical University of Vienna, Austria; 2Department of
Internal Medicine I, Division of Hematology and Hemostaseology, Medical University of
Vienna, Austria; 3Laboratoire de Biologie et Pharmacologie Appliquee, CNRS UMR 8113,
Ecole Normale Superieure de Cachan, France; 4Department of Hematology and Oncology,
University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg
University, Germany; 5Department of Laboratory Medicine, Medical University of Vienna,
Austria; 6Department for Companion Animals and Horses, University Clinic for Small
Animals, Internal Medicine Small Animals, University of Veterinary Medicine Vienna,
Austria; 7Elisabethinen Hospital Linz, Austria and 8Fifth Medical Department, Hospital
Hietzing, Vienna, Austria

ABSTRACT

S
ystemic mastocytosis is a complex disease defined by abnormal
growth and accumulation of neoplastic mast cells in various organs.
Most patients exhibit a D816V-mutated variant of KIT, which con-
fers resistance against imatinib. Clinical problems in systemic mastocyto-
sis arise from mediator-related symptoms and/or organ destruction
caused by malignant expansion of neoplastic mast cells and/or other
myeloid cells in various organ systems. DCC-2618 is a spectrum-selective Correspondence:
pan KIT and PDGFRA inhibitor which blocks KIT D816V and multiple peter.valent@meduniwien.ac.at
other kinase targets relevant to systemic mastocytosis. We found that
DCC-2618 inhibits the proliferation and survival of various human mast
cell lines (HMC-1, ROSA, MCPV-1) as well as primary neoplastic mast
Received: September 1, 2017.
cells obtained from patients with advanced systemic mastocytosis (IC50
<1 μM). Moreover, DCC-2618 decreased growth and survival of primary
Accepted: January 31, 2018.
Pre-published: February 8, 2018.
neoplastic eosinophils obtained from patients with systemic mastocyto-
sis or eosinophilic leukemia, leukemic monocytes obtained from patients
with chronic myelomonocytic leukemia with or without concomitant doi:10.3324/haematol.2017.179895
systemic mastocytosis, and blast cells obtained from patients with acute
myeloid leukemia. Furthermore, DCC-2618 was found to suppress the Check the online version for the most updated
proliferation of endothelial cells, suggesting additional drug effects on information on this article, online supplements,
systemic mastocytosis-related angiogenesis. Finally, DCC-2618 was and information on authorship & disclosures:
www.haematologica.org/content/103/5/799
found to downregulate IgE-mediated histamine release from basophils
and tryptase release from mast cells. Together, DCC-2618 inhibits
growth, survival and activation of multiple cell types relevant to ©2018 Ferrata Storti Foundation
advanced systemic mastocytosis. Whether DCC-2618 is effective in vivo Material published in Haematologica is covered by copyright.
in patients with advanced systemic mastocytosis is currently under inves- All rights are reserved to the Ferrata Storti Foundation. Use of
published material is allowed under the following terms and
tigation in clinical trials. conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode.
Copies of published material are allowed for personal or inter-
nal use. Sharing published material for non-commercial pur-
poses is subject to the following conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
Introduction sect. 3. Reproducing and sharing published material for com-
mercial purposes is not allowed without permission in writing
Systemic mastocytosis (SM) is a hematopoietic neoplasm with complex biology from the publisher.
and pathology, and a variable clinical course.1-7 The disease is characterized by
abnormal expansion and accumulation of neoplastic mast cells (MC) in one or more

Haematologica | 2018; 103(5) 799


M. Schneeweiss et al.

internal organs, including the bone marrow.1-3 Various eosinophils display KIT D816V.33 By contrast, expression of
types of SM have been recognized by the World Health rearranged PDGFR variants is rarely seen in SM, although
Organization (WHO).8-11 The indolent variant of SM is in some patients with a FIP1L1/PDGFRA fusion gene, the
associated with ‘hematologic stability’ and thus with an MC expansion has a histopathological picture indistin-
almost normal life expectancy.12-14 By contrast, the progno- guishable from that of SM.34 Treatment of SM-AHN repre-
sis in patients with advanced SM, including SM with an sents a clinical challenge because the AHN-component is
associated hematologic neoplasm (AHN), aggressive SM often resistant.16,32
(ASM) and MC leukemia (MCL) is unfavorable, with short DCC-2618 is a switch-control type II inhibitor of KIT,
survival times and poor responses to conventional which arrests KIT in an inactive state, regardless of activat-
therapy.1-5,12,13,15 Current research is, therefore, focusing on ing mutations, such as KIT D816V.35 Moreover, several
therapeutic targets and the effects of novel antineoplastic additional oncogenic kinases, including FLT-3, PDGFRA,
drugs on various cell types relevant to advanced SM.16 PDGFRB, KDR, TIE2 and FMS are recognized by DCC-
Since most patients with SM also suffer from mediator- 2618.35 Recently, the first clinical trials with DCC-2618
related symptoms that may sometimes be severe or even (NCT02571036) were started in patients with kinase-dri-
life-threatening, such drugs are often selected based on ven malignancies. In addition, first preclinical studies have
their dual effects on MC growth and MC activation. shown that DCC-2618 may exert antineoplastic effects on
Most patients with SM express the D816V-mutated vari- neoplastic MC.36
ant of the stem cell factor receptor, KIT, which mediates In our current study, we show that DCC-2618 is a potent
ligand-independent activation and autonomous growth inhibitor of growth and survival of neoplastic human MC
and differentiation of MC.17-22 The D816V KIT point muta- expressing various KIT mutations. Furthermore, we show
tion also confers resistance against several tyrosine kinase that DCC-2618 produces growth inhibition and apoptosis
inhibitors, including imatinib.23-26 Novel kinase blockers act- in other cell types that play a role in advanced SM. Finally,
ing on KIT D816V have, therefore, been developed. The we show that DCC-2618 inhibits IgE-dependent histamine
highlighting example is midostaurin (PKC412).27,28 secretion from basophils and tryptase secretion from MC.
However, despite superior clinical efficacy seen in a global All in all, our data suggest that DCC-2618 is a promising,
phase II trial,28 patients with advanced SM often exhibit or novel drug for the treatment of advanced SM.
acquire resistance.28,29 A number of different mechanisms
may underlie resistance against midostaurin. One obvious
problem is that the drug does not suppress all clinically rel- Methods
evant sub-clones and cell-types, especially cells lacking KIT
D816V.28,29 Such sub-clones are often seen in the context of Reagents
advanced SM. Over 50% of these patients have or develop The reagents used in this study are described in the Online
an AHN.30-32 Of these patients with an AHN, approximate- Supplement. DCC-2618 and its active metabolite, DP-5439, were
ly 80-90% have an associated myeloid neoplasm, the most kindly provided by Dr. B. Smith (Deciphera Pharmaceuticals LLC,
frequent ones being chronic myelomonocytic leukemia Lawrence, KS, USA).
(CMML) and acute myeloid leukemia (AML).8-11,30-32 In these
patients, leukemic expansion of monocytes and/or blast Isolation of primary neoplastic cells
cells is typically found. In other patients, an expansion of Primary neoplastic cells were isolated from bone marrow
eosinophils, sometimes resembling chronic eosinophilic samples of 11 patients with SM. The bone marrow cells were
leukemia (SM-CEL), is found. In most of these patients, obtained during routine diagnostic investigations after written

Table 1. Characteristics of patients with systemic mastocytosis and response of neoplastic cells to DCC-2618 and DP-5439.
Patient n. Age m/f Diagnosis, SM KIT Serum tryptase BM MC % MC DCC-2618 DP-5439 PKC412
variant D816V ng/mL infiltration % in MNC IC50 IC50 IC50
#1 68 m ISM + 83.3 n.a. 1 114 nM 414 nM 56 nM
#2 56 m ISM + 103 20 5 240 nM 390 nM 35 nM
#3 49 f ISM - 22.9 10 n.a. 198 nM 554 nM n.a.
#4 66 m ISM-MPN-eo + 170 5 n.a. 394 nM 1481 nM n.a.
#5 82 f SSM + 284 50 n.a. 347 nM 1584 nM n.a.
#6 57 f ASM + 87.9 50 n.a. 331 nM 366 nM n.a.
#7 90 m ASM + 125 20 25 386 nM 679 nM 360 nM
#8 63 m ASM-AML + 33.9 15 8 393 nM 554 nM 66 nM
#9 65 m ASM-CMML + 220 50 30 460 nM 907 nM n.a.
#10 78 m ASM-MPN-eo + 45.9 15 5 83 nM 64 nM 114 nM
#11 91 m MCL + 330 20 50 321 nM 592 nM 65 nM
Diagnoses were established according to WHO criteria. Primary bone marrow cells were incubated with various concentrations of DCC-2618, DP-5439 or midostaurin (PKC412)
at 37°C for 48 h. Proliferation was then determined by measuring uptake of 3H-thymidine and IC50 values were calculated. WHO: World Health Organization; m: male; f: female;
SM: systemic mastocytosis; PB, peripheral blood; BM, bone marrow; MC: mast cells; MNC, mononuclear cells, nM, nanomolar; n.a., not available; IC50, half maximal inhibitory con-
centration; ISM: indolent SM; MPN: myeloproliferative neoplasms; SSM: smoldering SM; ASM: aggressive SM; CMML: chronic monomyelocytic leukemia; MCL: mast cell leukemia.

800 haematologica | 2018; 103(5)


DCC-2618: a new drug against mastocytosis

informed consent had been given. Patients were classified as Results


having indolent SM (ISM; n=3), smoldering SM (SSM; n=1),
ASM (n=2), SM-AHN (n=4) and MCL (n=1) according to WHO DCC-2618 and its metabolite DP-5439 inhibit
criteria.8-11 In addition, neoplastic cells were obtained from ten proliferation of neoplastic mast cells
patients suffering from CMML, ten with AML and three with DCC-2618 and its active metabolite, DP-5439 were
hypereosinophilia. The patients’ characteristics are shown in found to inhibit 3H-thymidine uptake and thus prolifera-
Table 1 (SM patients) and Online Supplementary Table S1 (other tion in a dose-dependent manner in all MC lines tested,
hematologic disorders). Heparinized bone marrow cells were with slightly lower IC50 values obtained in HMC-1.1 cells
layered over Ficoll to isolate mononuclear cells. The study was lacking KIT D816V and ROSAKIT WT cells compared to the
approved by the ethics committee of the Medical University of KIT D816V-positive cell lines HMC-1.2 and ROSAKIT D816V
Vienna. (Figure 1A and Table 2). IC50 values obtained in HMC-1.1
cells with DCC-2618 were also lower than IC50 values
Culture of human cell lines obtained with midostaurin.25,26 In addition, DCC-2618 and
The following human MCL-like cell lines were employed in DP-5439 were found to inhibit proliferation of ROSAKIT K509I
this study: HMC-1.1 and HMC-1.2,37 three ROSA sub-clones cells with lower IC50 values (DCC-2618, IC50: 34±10 nM)
(ROSAKIT WT, ROSAKIT D816V, ROSAKIT K509I)38 and four MCPV-1 sub- compared to ROSAKIT D816V cells (Figure 1A). Unexpectedly,
clones (MCPV-1.1, MCPV-1.2, MCPV-1.3, MCPV-1.4).39 In addi- DCC-2618 and its metabolite also produced growth-inhi-
tion, we examined several AML cell lines, the CEL-related cell bition in the multi-resistant MC lines MCPV-1.1, MCPV-
line EOL-1, the microvascular endothelial cell line HMEC-1, and 1.2, MCPV-1.3 and MCPV-1.4 (Figure 1B and Table 2).
cultured human umbilical vein endothelial cells (HUVEC). A Finally, we were able to show that DCC-2618 and DP-
description of cell lines is provided in the Online Supplement. 5439 induced dose-dependent inhibition of growth of pri-
mary neoplastic bone marrow cells obtained from patients
Evaluation of growth, survival of neoplastic cells suffering from various forms of SM, including ASM and
Drug-exposed cells (cell lines or primary cells) were analyzed MCL (Figure 1C, Table 1). Interestingly, the effects of
for proliferation and survival. The bioassays employed are DCC-2618 on primary neoplastic BM cells and the related
described in the Online Supplementary Methods. IC50 values obtained in different SM variants were compa-
rable (Figure 1C, Table 1).
Western blotting
For evaluation of KIT and BTK signaling, HMC-1.1, HMC-1.2, DCC-2618 inhibits KIT, STAT5, AKT, and ERK activation in
ROSAKIT WT and ROSAKIT D816V cells were incubated in control neoplastic mast cells
medium or in DCC-2618 (0.5-5 μM) for 4 h at 37°C. Western As expected, DCC-2618 was found to suppress phos-
blotting was performed essentially as described elsewhere.26,40 phorylation of KIT in ROSAKIT WT and ROSAKIT D816V cells as
For evaluation of downstream signaling pathways of KIT, HMC- well as in both HMC-1 sub-clones (Figure 2A). In addition,
1.1, HMC-1.2, ROSAKIT WT and ROSAKIT D816V cells were first pre- DCC-2618 was found to decrease the expression of phos-
incubated overnight in Iscove modified Dulbecco medium phosphorylated (p)STAT5, pAKT and pERK1/2 in all cell
devoid of fetal calf serum and of stem cell factor. Cells (106) from lines tested (Figure 2B). As expression levels of pSTAT5 in
each line were then treated with DCC-2618 (0.001-10 μM) for
90 min at 37°C. At the end of the treatment, ROSAKIT WT cells
were stimulated with stem cell factor-containing supernatants
(10%) of Chinese hamster ovary cells transfected with the Table 2. Effects of DCC-2618 and DP-5439 on growth of various
murine scf (kl) gene (CHO-KL) at room temperature for 10 min. human cell types.
Thereafter, Western blotting was performed essentially as Cell line / cell type DCC-2618, IC50 DP-5439, IC50
described previously.26,40 Antibodies against phosphorylated
(p)Kit, STAT5, pSTAT5, ERK1/2, pERK1/2 were purchased from HMC-1.1 12.3±3.7 nM 13±4 nM
Cell Signaling (Danvers, MA, USA), antibodies against pBTK HMC-1.2 123±36 nM 96±23 nM
were bought from NovusBiologicals (Littleton, CO, USA) and ROSA KIT WT 41±5 nM 56±21 nM
antibodies against total KIT and total BTK were from Santa Cruz ROSA KIT D816V 168±65 nM 188±60 nM
Biotechnology (Santa Cruz, CA, USA).
ROSA KIT K509I 34±10 nM 28±13 nM
Measurement of mediator release MCPV-1.1 298±77 nM 357±179 nM
Drug-exposed cells (blood basophils obtained from healthy MCPV-1.2 1000±932 nM 913±378 nM
individuals and HMC-1 cells) were analyzed for histamine- and MCPV-1.3 724±511 nM 756±435 nM
tryptase release as described in the Online Supplementary MCPV-1.4 670±418 nM 697±410 nM
Methods.
MOLM-13 132±95 nM 73±31 nM
Evaluation of apoptosis in basophils MV4-11 147±88 nM 76±24 nM
Drug-exposed blood basophils obtained from healthy donors KG-1 2.7±3.7 μM 5.2±4.4 μM
by dextran sedimentation were analyzed for cell survival by U937 7.8±5.4 μM 2.5±0.3 μM
flow cytometry. Technical details are described in the Online
EOL-1 1.8±1.3 nM 1±0.8 nM
3.7±2.2 μM 2±1.3 μM
Supplementary Methods.
HMEC-1
Statistical analysis HUVEC 707±224 nM 605±222 nM
To determine the level of significance the Student t-test was Cell lines were incubated with various concentrations of DCC-2618 or DP-5439 at 37°C
for 48 h.Then, proliferation was determined by measuring uptake of 3H-thymidine and
applied. Results were considered to be statistically significantly
IC50 values were calculated. Values represent the mean±S.D. from three independent
different when P was <0.05. experiments. IC50: half maximal inhibitory concentration.

haematologica | 2018; 103(5) 801


M. Schneeweiss et al.

HMC-1.1 cells were rather low and difficult to quantify by and Online Supplementary Figure S2A,B). The metabolite
Western blotting, we also performed intracellular flow DP-5439 was found to be equally effective in producing
cytometry-staining experiments using an antibody against apoptosis in MC lines compared to DCC-2618 (Figure
pSTAT5. In these experiments, DCC-2618 was found to 3A,B and Online Supplementary Figure S2A,B). Together,
counteract pSTAT5 expression in HMC-1.1 and HMC-1.2 these data show that DCC-2618 is a novel potent anti-
cells in a dose-dependent manner (Online Supplementary neoplastic compound inducing apoptosis and growth
Figure S1). DCC-2618 did not inhibit phosphorylation of arrest in neoplastic MC.
BTK, another important target of tyrosine kinase inhibitors
expressed by neoplastic MC (Figure 2C). DCC-2618 produces synergistic growth-inhibitory effects
with midostaurin and cladribine (2CdA) in neoplastic
DCC-2618 induces apoptosis in neoplastic mast cells mast cells
To explore the mechanism of drug action, we analyzed In advanced SM, drug combinations may be required to
the effects of DCC-2618 on the survival of neoplastic suppress malignant cell growth. We found that DCC-2618
MC. As assessed by light microscopy, DCC-2618 induced and midostaurin produce clear cooperative (synergistic)
apoptosis in HMC-1.1, HMC-1.2, ROSAKIT WT, ROSAKIT growth-inhibitory effects in HMC-1.1 cells (Online
D816V
and ROSA KIT K509I cells in a dose-dependent manner Supplementary Figure S3A,C). In HMC-1.2 cells, the drug
(Figure 3A). The effects of DCC-2618 on survival were combination also produced cooperative antineoplastic
more pronounced in KIT D816V-negative MC lines than effects, but these effects were additive rather than syner-
in KIT D816V-positive MC lines (Figure 3A). DCC-2618 gistic as defined by Calcusyn software (Online
was also found to produce apoptosis in the multi-resis- Supplementary Figure S3A,C). In addition, we found that
tant MCPV-1 cell lines (Figure 3B). The apoptosis-induc- DCC-2618 and 2CdA induce clear synergistic growth-
ing effect of DCC-2618 on MC was confirmed by com- inhibitory effects on HMC-1.1 and HMC-1.2 cells (Online
bined annexin V/propidium-iodide staining (Figure 3A,B Supplementary Figure S3B,D).

B C

Figures 1. DCC-2618 and its active metabolite DP-5439 inhibit proliferation of neoplastic mast cells. HMC-1, ROSA (A), MCPV-1 (B) and primary neoplastic mast
cells (C) obtained from patients with different variants of systemic mastocytosis (ISM, SSM, ASM, MCL) were incubated in control medium (0 nM) or medium con-
taining various concentrations of DCC-2618 or DP-5439, as indicated, at 37°C for 48 h. Thereafter, 3H-thymidine uptake was measured. Results in (A) and (B) are
expressed as percent of control and represent the mean±S.D. from three independent experiments. Results in (C) are expressed as percent of control and represent
mean±S.D from triplicates. Asterisk (*): P<0.05 compared to control medium.

802 haematologica | 2018; 103(5)


DCC-2618: a new drug against mastocytosis

DCC-2618 inhibits IgE-dependent histamine release DCC-2618 counteracts growth and survival of leukemic
from basophils and spontaneous tryptase release from monocytes and blast cells
neoplastic mast cells We next explored the effects of DCC-2618 on AHN cell-
Since patients with SM often suffer from symptoms types. In a first step, we examined AML cell responses.
caused by mediators released from neoplastic MC and/or DCC-2618 was found to inhibit the proliferation of all
basophils, we evaluated the effect of DCC-2618 on anti- AML cell lines tested, with considerably lower IC50 values
IgE-induced histamine release. We found that DCC-2618 obtained with the FLT3-mutated cell lines MOLM-13
(0.1-1.0 μM) slightly inhibited anti-IgE mediated histamine (132±95 nM) and MV4-11 (147±88 nM) compared to KG-
release from normal human blood basophils (Figure 4A). 1 and U937 cells (Table 2, Figure 5A). Similar effects were
This drug effect was found to be specific in that DCC-2618 seen with DP-5439 (Figure 5A). DCC-2618 was also found
did not inhibit C5a- or calcium ionophore-induced hista- to induce apoptosis in MOLM-13, MV4-11 and KG-1 cells
mine release from basophils (Online Supplementary Figure (Figure 5B and Online Supplementary Figure S5). Finally, we
S4A). As expected, DCC-2618 did not affect the viability of found that DCC-2618 and DP-5439 produced dose-depen-
basophils between 0.1 and 1.0 μM and did not induce his- dent inhibition of growth in primary leukemia cells
tamine secretion within 30 min of incubation (Online obtained from patients with AML or CMML (Online
Supplementary Figure S4B). In consecutive experiments, we Supplementary Table S1 and Figure 5C). In one patient with
also found that DCC-2618 suppresses the spontaneous ASM-CMML, we isolated mononuclear cells and found
(baseline) secretion of tryptase from HMC-1.1 and HMC- that DCC-2618 and DP-5439 induced growth inhibition of
1.2 cells during the entire incubation period (days 1 these cells in the same way as in mononuclear cells
through 6) (Figure 4B). obtained from patients with CMML without SM (Table 1

Figures 2. DCC-2618 inhibits phosphorylation of KIT and other targets in neoplastic mast cells. (A,C) HMC-1 and ROSA cells were incubated in control medium (ROSAKIT
WT
: Iscove modified Dulbecco medium (IMDM) with stem cell factor, SCF; ROSAKIT D816V: IMDM without SCF) or medium containing various concentrations of DCC-2618, as
indicated, at 37°C for 4 h. Thereafter, cells were harvested and Western blotting was performed as described in the text using antibodies against phosphorylated (p)KIT,
total KIT, pBTK and total BTK. (B) HMC-1 and ROSA cells were first pre-incubated overnight in IMDM devoid of fetal calf serum and of SCF. Cells were then treated with
DCC-2618 (0.001-10 μM) for 90 min at 37°C. At the end of the treatment, ROSAKIT WT cells were stimulated with SCF (10% CHO-KL) at room temperature for 10 min.
Thereafter, cells were harvested and Western blotting was performed as described in the text using antibodies against pSTAT5, total STAT5, pAKT, total AKT, pERK1/2,
total ERK1/2. Western blot experiments were performed at least twice. Western blots in this figure show one representative experiment.

haematologica | 2018; 103(5) 803


M. Schneeweiss et al.

and Figure 5C). Together, these data suggest that DCC- survival of the FIP1L1-PDGFRA (F/P) positive EOL-1 cell
2618 counteracts growth of AHN cells, including CMML line. DCC-2618 was found to inhibit proliferation in
monocytes and AML blasts. EOL-1 cells at low nanomolar range of concentrations
(IC50: 1.8±1.3 nM) (Online Supplementary Figure S6A).
DCC-2618 inhibits the proliferation of neoplastic Similar effects were seen with DP-5439 (Online
eosinophils Supplementary Figure S6A). DCC-2618 also induced apop-
Advanced SM is often accompanied by eosinophilia. In tosis in EOL-1 cells (Online Supplementary Figure S6C).
addition PDGFRA is a known target of DCC-2618. We Next, we examined the effects of DCC-2618 on growth
analyzed the effects of DCC-2618 on proliferation and of primary eosinophils. In these experiments, DCC-2618

Figures 3. DCC-2618 and DP-5439 induce apoptosis in neoplastic mast cells. HMC-1, ROSA (A) and MCPV-1 (B) were incubated in control medium (0 μM) or medium
containing various concentrations of DCC-2618 and DP-5439, as indicated, at 37°C for 48 h. Cells were then harvested and the percentage of apoptotic cells was
quantified morphologically on Wright-Giemsa-stained cytospin preparations (left panels) or by flow cytometry (determination of annexinV/PI-positive cells, right pan-
els). Results represent the mean±S.D. of three independent experiments. Asterisk (*): P<0.05 compared to control medium.

804 haematologica | 2018; 103(5)


DCC-2618: a new drug against mastocytosis

was found to inhibit the proliferation of neoplastic bone drugs the prognosis of these patients remains poor with
marrow cells obtained from a patient with ASM (Table 1 short survival times.10,16,28,29 Research is, therefore, seeking
and Online Supplementary Figure S6B). In addition, DCC- new effective drugs and novel treatment concepts. DCC-
2618 was found to block the growth of bone marrow cells 2618 is a novel switch-control type II blocker that exerts
obtained from patients with secondary hypereosinophilic inhibitory effects on KIT D816V, other KIT mutants, and
syndromes (Online Supplementary Table S1 and Online several other critical target kinases, such as FLT3,
Supplementary Figure S6B). PDGFRA and KDR.35 We here describe that DCC-2618
inhibits the proliferation of nine different human MC
DCC-2618 inhibits growth of human endothelial cells lines, with lower IC50 values obtained in HMC-1.1 cells
Increased bone marrow angiogenesis has been impli- and ROSAKIT WT cells than in KIT D816V-positive HMC-1.2
cated in the pathogenesis of SM.41 To investigate potential and ROSAKIT D816V cells. In addition, DCC-2618 was found
effects of DCC-2618 on angiogenesis, we explored drug to block the proliferation of primary neoplastic MC
effects on growth of HUVEC and the microvascular obtained from patients with ASM or MCL. Moreover,
endothelial cell line HMEC-1. As assessed by 3H-thymi- DCC-2618 exerted major antineoplastic effects on AHN
dine uptake, DCC-2618 and its metabolite were found to cells and endothelial cells, all of which may be relevant in
inhibit the proliferation of HUVEC and HMEC-1 cells in the pathogenesis of advanced SM. Based on these obser-
a dose-dependent manner (Online Supplementary Figure vations DCC-2618 is a novel emerging drug candidate for
S7). DCC-2618 exerted stronger effects on HUVEC advanced SM. Indeed, clinical trials with DCC-2618 have
(707±224 nM) than on HMEC-1 cells (3.7±2.2 µM). been started recently.
The multi-kinase inhibitor midostaurin (PKC412) is
effective against the D816V-mutated variant of KIT and
Discussion has shown promising results in patients with advanced
SM in a global phase II trial, with an overall response rate
Due to the poor response to conventional drugs, treat- of 60%.28 In addition, midostaurin was found to suppress
ment of patients with advanced SM is still a major chal- mediator-related symptoms and IgE-dependent histamine
lenge in clinical practice. Despite the availability of new release from basophils.28,42 However, despite clinical effi-

Figures 4. Effects of DCC-2618 on anti-IgE-induced histamine release from normal


basophils. (A) Primary blood basophils from healthy donors were incubated in control medi-
um (0 μM) or in various concentrations of DCC-2618, as indicated, at 37°C for 30 min.
Thereafter, cells were incubated in control buffer or in buffer containing anti-IgE antibody
E-124.2.8 (1 μg/mL) at 37°C for 30 min. After incubation, cells were centrifuged at 4°C,
and cell-free supernatants and cell suspensions recovered and examined for histamine-
content by radioimmunoassay. Histamine release was calculated as percent of total hista-
mine and is expressed as percent of control. Results represent the mean±S.D. of four inde-
pendent experiments. Asterisk (*): P<0.05 compared to control medium. (B) HMC-1 cells
were cultured in the presence or absence of DCC-2618 (HMC-1.1: 25 nM; HMC-1.2: 500
nM) over a 6-day period. Spontaneous release of tryptase from HMC-1.1 and HMC-1.2 cells
was measured by determining tryptase concentrations in cell-free supernatants and
lysates. Tryptase release is expressed as percent of total (intra- and extra-cellular) tryptase.
Results represent the mean±S.D. of three independent experiments. Asterisk (*): P<0.05.

haematologica | 2018; 103(5) 805


M. Schneeweiss et al.

Figures 5. Effects of DCC-2618 and DP-5439 on proliferation and survival of acute myeloid leukemia (AML) and chronic myelomonocytic leukemia (CMML). (A,C)
MOLM-13, MV4-11, KG-1, U937 and primary leukemic cells were incubated in control medium (0 μM) or medium containing various concentrations of DCC-2618 or
DP-5439, as indicated at 37°C for 48 h. Thereafter, 3H-thymidine uptake was determined. Results in (A) are expressed as percent of control and represent the
mean±S.D. from three independent experiments. Asterisk (*): P<0.05 compared to control medium. Results of (C) are expressed as percent of control and represent
the mean±S.D. from triplicates. (B) MOLM-13, MV4-11, KG-1 and U937 cells were incubated with control medium (0 nM) or various concentrations of DCC-2618 and
DP-5439, as indicated, for 48 h. Thereafter cells were harvested and the percentage of apoptotic cells was quantified morphologically on Wright-Giemsa-stained
cytospin preparations (left panels) or by flow cytometry (determination of annexinV/PI-positive cells, right panels). Results represent the mean±S.D. of three inde-
pendent experiments. Asterisk (*): P<0.05 compared to control medium.

806 haematologica | 2018; 103(5)


DCC-2618: a new drug against mastocytosis

cacy, midostaurin is unable to produce long-lasting com- inhibit growth of primary eosinophils obtained from
plete remission in all patients.28 Therefore, new drugs and patients with KIT D816V-positive SM or reactive hypere-
drug-combinations are currently being tested in the con- osinophilia. Together, these data suggest that DCC-2618
text of advanced SM. DCC-2618 might be a promising inhibits multiple AHN-related cell types, which may be
candidate for several reasons. First, DCC-2618 exhibits a relevant clinically as progression of SM is often accompa-
broad target profile and is able to block growth of various nied by multilineage expansion of various sub-clones,
neoplastic cells.36 In the current study, DCC-2618 was including cells harboring or lacking KIT D816V.15,28,29
found to block growth of neoplastic cells obtained from A number of different pro-oncogenic pathways and tar-
patients with ASM and MCL. In addition, the drug pro- gets may be involved in KIT D816V-dependent expansion
duced growth inhibition in all MCL-like cell lines tested, and accumulation of MC in advanced SM.25,40,44-50 Several
including KIT-mutated cells and cell lines in which other of these target pathways may be sensitive to therapy
oncogenic pathways (such as the RAS pathway) trigger with tyrosine kinase inhibitors. We studied whether key
malignant cell growth. Moreover, unlike other KIT-target- target pathways in neoplastic MC can be disrupted by
ing drugs, DCC-2618 is able to suppress the growth and DCC-2618. As assessed by Western blotting, DCC-2618
survival of other cell types relevant to advanced SM and was found to block the phosphorylation and thus activa-
AHN, including monocytes, blast cells, neoplastic tion of wild-type KIT and KIT D816V. In addition, we
eosinophils and endothelial cells. The concentrations were able to show that DCC-2618 blocks the activation
required to mediate these cellular inhibitory effects are of AKT, ERK and STAT5, suggesting that multiple target
readily achievable based on the recent report of clinical pathways are accessible to this drug. By contrast, howev-
exposure of 5 μM or higher in patients with gastrointesti- er, the drug did not disrupt activation of BTK, another
nal stroma tumors.43 important target displayed by neoplastic MC.46
After intake, DCC-2618 is considered to be converted Since the target spectrum of midostaurin (PKC412) and
to one active metabolite, DP-5439. We therefore investi- DCC-2618 is not identical, we were also interested to
gated whether DP-5439 is also able to counteract growth learn whether DCC-2618 and midostaurin can produce
and survival of neoplastic cells. In these experiments, we synergistic antineoplastic effects on neoplastic MC.
were able to show that DP-5439 is able to suppress Indeed, we found that both drugs induce cooperative or
growth and survival of neoplastic MC and of other even synergistic growth-inhibitory effects on HMC-1.1
leukemic (non-MC-lineage) cells in the same way (and and HMC-1.2 cells.
with comparable IC50 values) as DCC-2618. These data Specific alterations in the microenvironment, including
suggest that DCC-2618 treatment should be effective increased angiogenesis, are frequently detectable in
even if the DP-5439 metabolite may accumulate over advanced bone marrow neoplasms and are often consid-
time. ered to play an important role in disease progression. A
It is well known that about one-third of all patients typical finding in the affected bone marrow of patients
with advanced SM have an AHN at diagnosis. Of these with advanced SM is increased microvessel density.41 We
patients, most have a myeloid neoplasm, often in the found that DCC-2618 inhibits the proliferation of human
form of CMML or AML.2-8,13,32 The treatment of these SM- endothelial cells, including HUVEC and a microvascular
AHN patients is a clinical challenge because the AHN is endothelial cell line, HMEC-1. These data suggest that
often drug-resistant. In fact patients with SM-AHN still DCC-2618 also acts as an anti-angiogenic agent.
have a poor prognosis with an overall survival time of Interestingly, the IC50 values obtained for HMEC-1 cells
about 24 months.13,32 Because of its broad activity profile, were higher than those for HUVEC, which may be
we asked whether DCC-2618 might be a promising agent explained by the fact that HMEC-1 is a cell line, whereas
for patients with SM-AHN. In a first step, we found that HUVEC are primary cells. An alternative explanation
DCC-2618 is a potent inhibitor of proliferation and sur- would be the lack of key targets in HMEC-1 cells.
vival of the FLT3-mutated AML cell lines MOLM-13 and Indeed, it is well known, that KDR, a key target of DCC-
MV4-11. DCC-2618 also inhibits the growth of other 2618, is only expressed in HUVEC but not in HMEC-1
AML cell lines examined (KG-1 and U937), but at IC50 val- cells.
ues considerably higher than those for MOLM-13 or Patients with SM frequently suffer from symptoms
MV4-11 cells. We also found that DCC-2618 counteracts produced by MC-derived mediators.4-6,16 These mediators
proliferation of primary leukemic cells obtained from are released on IgE-dependent activation of MC and may
patients with SM-AHN, AML or CMML (Table 1 and cause severe problems or even lead to life-threatening
Online Supplementary Table S1). These findings suggest anaphylaxis. Concomitant (IgE-dependent) allergies are,
that DCC-2618 may be a promising agent for SM-AHN. therefore, relevant comorbidities in the context of SM.
In SM patients, disease progression is often accompa- We found that DCC-2618 counteracts IgE-dependent
nied by expansion of neoplastic eosinophils, sometimes secretion of histamine from basophils obtained from
even resembling (chronic) eosinophilic leukemia. In most healthy donors. In addition, we were able to show that
cases the eosinophils are of clonal origin as they express DCC-2618 blocks IgE-independent, spontaneous release
KIT D816V.33 In rare cases, neoplastic eosinophils display of tryptase from HMC-1.1 and HMC-1.2 cells in vitro.
the FIP1L1/PDGFRA fusion gene.34 However, this fusion These results suggest that, apart from its antineoplastic
gene is usually detectable only in eosinophilic neoplasms, effects, DCC-2618 might also have an impact on media-
such as CEL. Since DCC-2618 is known to exert inhibito- tor release (and probably on the resulting symptoms) in
ry effects against PDGFRA35 we examined its effects on patients with SM with concomitant allergies. Whether
EOL-1 cells harboring FIP1L1-PDGFRA. DCC-2618 was these data can be reproduced in vivo and whether the drug
found to exert strong anti-proliferative and apoptosis- is able to suppress mediator symptoms in patients with
inducing effects in EOL-1 cells, with IC50 values in the low advanced SM or SM with concomitant allergies remains
nanomolar range. In addition, DCC-2618 was found to to be determined. In fact, whereas the concentrations of

haematologica | 2018; 103(5) 807


M. Schneeweiss et al.

DCC-2618 required to block tryptase secretion in MC growth of neoplastic MC in patients with advanced SM is
were rather low (<1.0 μM), the concentrations required to currently being explored in a clinical trial
block IgE-dependent histamine release were rather high (NCT02571036).
(1 μM).
Collectively, our data indicate that DCC-2618 is a novel Acknowledgments
promising agent that counteracts growth and survival of This study was supported by a grant from Deciphera
various cell types relevant to the pathogenesis of Pharmaceuticals LLC. We like to thank Dr. Dan Flynn, Dr.
advanced SM. Whether DCC-2618 is able to block Bryan Smith and Dr. Oliver Rosen for helpful discussion.

References systemic mastocytosis and related mast cell (2CdA) and evaluation of cooperative drug
disorders: current treatment options and effects. Blood. 2006;107(2):752-759.
proposed response criteria. Leuk Res. 27. Fabbro D, Ruetz S, Bodis S, et al. PKC412 —
1. Metcalfe DD. Classification and diagnosis 2003;27(7):635-641. a protein kinase inhibitor with a broad ther-
of mastocytosis: current status. J Invest 16. Arock M, Akin C, Hermine O, et al. Current apeutic potential. Anticancer Drug Des.
Dermatol. 1991;96(3):2S-4S. treatment options in patients with mastocy- 2000;15(1):17-28.
2. Valent P. Biology, classification and treat- tosis: status in 2015 and future perspectives. 28. Gotlib J, Kluin-Nelemans HC, George TI, et
ment of human mastocytosis. Wien Klin Eur J Haematol. 2015;94(6):474-490. al. Efficacy and safety of midostaurin in
Wochenschr. 1996;108(13):385-397. 17. Furitsu T, Tsujimura T, Tono T, et al. advanced systemic mastocytosis. N Engl J
3. Horny HP, Valent P. Diagnosis of mastocy- Identification of mutations in the coding Med. 2016;374(26):2530-2541.
tosis: general histopathological aspects, sequence of the proto-oncogene c-kit in a 29. Gotlib J, Berubé C, Growney JD, et al.
morphological criteria, and immunohisto- human mast cell leukemia cell line causing Activity of the tyrosine kinase inhibitor
chemical findings. Leuk Res. 2001;25(7): ligand-independent activation of the c-kit PKC412 in a patient with mast cell leukemia
543-551. product. J Clin Invest. 1993;92(4):1736- with the D816V KIT mutation. Blood.
4. Escribano L, Akin C, Castells M, et al. 1744. 2005;106(8):2865-2870.
Mastocytosis: current concepts in diagnosis 18. Nagata H, Worobec AS, Oh CK, et al. 30. Sperr WR, Horny HP, Valent P. Spectrum of
and treatment. Ann Hematol. 2002;81(12): Identification of a point mutation in the cat- associated clonal hematologic non-mast cell
677-690. alytic domain of the protooncogene c-kit in lineage disorders occurring in patients with
5. Valent P, Akin C, Sperr WR, et al. Diagnosis peripheral blood mononuclear cells of systemic mastocytosis. Int Arch Allergy
and treatment of systemic mastocytosis: patients who have mastocytosis with an Immunol. 2002;127(2):140-142.
state of the art. Br J Haematol. 2003;122(5): associated hematologic disorder. Proc Natl 31. Horny HP, Sotlar K, Sperr WR, et al.
695-717. Acad Sci. 1995;92(23):10560-10564. Systemic mastocytosis with associated
6. Akin C, Metcalfe DD. Systemic mastocyto- 19. Feger F, Ribadeau Dumas A, et al. Kit and c- clonal haematological non-mast cell lineage
sis. Annu Rev Med. 2004;55:419-432. kit mutations in mastocytosis: a short diseases: a histopathological challenge. J
7. Arock M, Valent P. Pathogenesis, classifica- overview with special reference to novel Clin Pathol. 2004;57(6):604-608.
tion and treatment of mastocytosis: state of molecular and diagnostic concepts. Int Arch 32. Pardanani A. Systemic mastocytosis in
the art in 2010 and future perspectives. Allergy Immunol. 2002;127(2):110-114. adults: 2017 update on diagnosis, risk strat-
Expert Rev Hematol. 2010;3(4):497-516. 20. Longley BJ, Tyrrell L, Lu SZ, et al. Somatic c- ification and management. Am J Hematol.
8. Valent P, Horny H-P, Escribano L, et al. kit activating mutation in urticaria pigmen- 2016;91(11):1146-1159.
Diagnostic criteria and classification of mas- tosa and aggressive mastocytosis: establish- 33. Pardanani A, Reeder T, Li CY, et al.
tocytosis: a consensus proposal. Leuk Res. ment of clonality in a human mast cell neo- Eosinophils are derived from the neoplastic
2001;25(27):603-625. plasm. Nat Genet. 1996;12(3):312-314. clone in patients with systemic mastocyto-
9. Valent P, Horny H-P, Li CY, et al. 21. Fritsche-Polanz R, Jordan JH, Feix A, et al. sis and eosinophilia. Leuk Res. 2003;27(10):
Mastocytosis (mast cell disease). World Mutation analysis of C-KIT in patients with 883-885.
Health Organization (WHO) Classification myelodysplastic syndromes without mas- 34. Pardanani A, Ketterling RP, Brockman SR, et
of Tumours. Pathology & Genetics. tocytosis and cases of systemic mastocyto- al. CHIC2 deletion, a surrogate for FIP1L1-
Tumours of Haematopoietic and Lymphoid sis. Br J Haematol. 2001;113(2):357-364. PDGFRA fusion, occurs in systemic masto-
Tissues. Eds: Jaffe ES, Harris NL, Stein H, 22. Arock M, Sotlar K, Akin C, et al. KIT muta- cytosis associated with eosinophilia and
Vardiman JW 2001;1:291-302. tion analysis in mast cell neoplasms: recom- predicts response to imatinib mesylate ther-
10. Valent P, Akin C, Metcalfe DD. mendations of the European Competence apy. Blood. 2003;102(9):3093-3096.
Mastocytosis: 2016 updated WHO classifi- Network on Mastocytosis. Leukemia. 35. Smith BD, Hood MM, Wise SC, et al. DCC-
cation and novel emerging treatment con- 2015;29(6):1223-1232. 2618 is a potent inhibitor of wild-type and
cepts. Blood. 2017;129(11):1420-1427. 23. Shah NP, Lee FY, Luo R, et al. Dasatinib mutant KIT, including refractory exon 17
11. Horny HP, Akin C, Arber D, et al. (BMS-354825) inhibits KITD816V, an ima- D816 KIT mutations, and exhibits efficacy
Mastocytosis. In: WHO Classification of tinib-resistant activating mutation that trig- in refractory GIST and AML xenograft
Tumours of Haematopoietic and Lymphoid gers neoplastic growth in most patients models. Cancer Res. 2015;75(15): abstract
Tissues. Eds: Swerdlow SH, Campo E, with systemic mastocytosis. Blood. 2690.
Harris NL et al. IARC Press Lyon, France, 2006;108(1):286-291. 36. Bai Y, Bandara G, Ching Chan E, et al,
vol 3, pp 62-69, 2017(2):62-69. 24. Akin C, Brockow K, D'Ambrosio C, et al. Targeting the KIT activating switch control
12. Lim KH, Tefferi A, Lasho TL, et al. Systemic Effects of tyrosine kinase inhibitor STI571 pocket: a novel mechanism to inhibit neo-
mastocytosis in 342 consecutive adults: sur- on human mast cells bearing wild-type or plastic mast cell proliferation and mast cell
vival studies and prognostic factors. Blood. mutated c-kit. Exp Hematol. activation. Leukemia. 2013; 27(2):278-285.
2009;113(23):5727-5736. 2003;31(8):686-692. 37. Butterfield JH, Weiler D, Dewald G, Gleich
13. Pardanani A, Lim KH, Lasho TL, et al. 25. Gleixner KV, Mayerhofer M, Sonneck K, et GJ. Establishment of an immature mast cell
Prognostically relevant breakdown of 123 al. Synergistic growth-inhibitory effects of line from a patient with mast cell leukemia.
patients with systemic mastocytosis associ- two tyrosine kinase inhibitors, dasatinib Leuk Res. 1988;12(4):345-355.
ated with other myeloid malignancies. and PKC412, on neoplastic mast cells 38. Saleh R, Wedeh G, Herrmann H, et al., A
Blood. 2009;114(18):3769-3772. expressing the D816V-mutated oncogenic new human mast cell line expressing a func-
14. Escribano L, Alvarez-Twose I, Sánchez- variant of KIT. Haematologica. tional IgE receptor converts to factor-inde-
Muñoz L, et al. Prognosis in adult indolent 2007;92(11):1451-1459. pendence and tumorigenicity by KIT
systemic mastocytosis: a long-term study of 26. Gleixner KV, Mayerhofer M, Aichberger KJ, D816V-transfection. Blood. 2014;124(1):
the Spanish Network on Mastocytosis in a et al. PKC412 inhibits in vitro growth of 111-120.
series of 145 patients. J Allergy Clin neoplastic human mast cells expressing the 39. Hoermann G, Blatt K, Greiner G, et al.,
Immunol. 2009;124(3):514-521. D816V-mutated variant of KIT: comparison CD52 is a molecular target in advanced sys-
15. Valent P, Akin C, Sperr WR, et al. Aggressive with AMN107, imatinib, and cladribine temic mastocytosis. FASEB J. 2014;28(8):

808 haematologica | 2018; 103(5)


DCC-2618: a new drug against mastocytosis

3540-3551. inhibitor, in patients with gastrointestinal Blood. 2011;118(7):1885-1898.


40. Peter B, Gleixner KV, Cerny-Reiterer S, et al. stroma tumor (GIST) and other solid 47. Wilson TM, Maric I, Simakova O, et al.
Polo-like kinase-1 as novel target in neoplas- tumors. J Clin Oncol. 2017;35:15(S)2515. Clonal analysis of NRAS activating muta-
tic mast cells: demonstration of growth- 44. Ustun C, Arock M, Kluin-Nelemans HC, et tions in KIT-D816V systemic mastocytosis.
inhibitory effects of siRNA and the Polo- al. Advanced systemic mastocytosis: from Haematologica. 2011;96(3):459-463.
like kinase-1 targeting drug BI 2536. molecular and genetic progress to clinical 48. Schwaab J, Schnittger S, Sotlar K, et al.
Haematologica. 2011;96(5):672-680. practice. Haematologica. 2016;101(10): Comprehensive mutational profiling in
41. Wimazal F, Jordan JH, Sperr WR, et al. 1133-1143. advanced systemic mastocytosis. Blood.
Increased angiogenesis in the bone marrow 45. Jawhar M, Schwaab J, Schnittger S, et al. 2013;122(14):2460-2466.
of patients with systemic mastocytosis. Am Additional mutations in SRSF2, ASXL1 49. Bibi S, Langenfeld F, Jeanningros S, et al.
J Pathol. 2002;160(5):1639-1645. and/or RUNX1 identify a high-risk group of Molecular defects in mastocytosis: KIT and
42. Peter B, Winter GE, Blatt K, et al. Target inter- patients with KIT D816V(+) advanced sys- beyond KIT. Immunol Allergy Clin North
action profiling of midostaurin and its temic mastocytosis. Leukemia. 2016;30(1): Am. 2014;34(2):239-262.
metabolites in neoplastic mast cells predicts 136-143. 50. Damaj G, Joris M, Chandesris O, et al.,
distinct effects on activation and growth. 46. Gleixner KV, Mayerhofer M, Cerny-Reiterer ASXL1 but not TET2 mutations adversely
Leukemia. 2016;30(2):464-472. S, et al. KIT-D816V-independent oncogenic impact overall survival of patients suffering
43. Janku F, Razak ARA, Gordon MS, et al. signaling in neoplastic cells in systemic mas- systemic mastocytosis with associated
Pharmacokinetic-driven phase I study of tocytosis: role of Lyn and BTK activation clonal hematologic non-mast-cell diseases.
DCC-2618, a pan-KIT and PDGFRA and disruption by dasatinib and bosutinib. PLoS One. 2014;9(1):e85362.

haematologica | 2018; 103(5) 809


ARTICLE Acute Myeloid Leukemia

Distinct protein signatures of acute myeloid


Ferrata Storti
Foundation leukemia bone marrow-derived stromal cells
are prognostic for patient survival
Steven M. Kornblau,1* Peter P. Ruvolo,1* Rui-Yu Wang,1 V. Lokesh Battula,1
Elizabeth J. Shpall,2 Vivian R. Ruvolo,1 Teresa McQueen,1 YiHua Qui,1
Zhihong Zeng,1 Sherry Pierce,1 Rodrigo Jacamo,1 Suk-Young Yoo,3
Phuong M. Le,1 Jeffrey Sun,1 Numsen Hail Jr,1 Marina Konopleva1 and Michael
Andreeff1

1
Section of Molecular Hematology and Therapy, Department of Leukemia; 2Department
of Stem Cell Transplantation and 3Bioinformatics and Computational Biology, The
Haematologica 2018
Volume 103(5):810-821 University of Texas M.D. Anderson Cancer Center, Houston, Texas, TX, USA
*SMK and PPR contributed equally to this work.

ABSTRACT

M
esenchymal stromal cells (MSC) support acute myeloid
leukemia (AML) cell survival in the bone marrow (BM)
microenvironment. Protein expression profiles of AML-derived
MSC are unknown. Reverse phase protein array analysis was performed
to compare expression of 151 proteins from AML-MSC (n=106) with
MSC from healthy donors (n=71). Protein expression differed signifi-
cantly between the two groups with 19 proteins over-expressed in
leukemia stromal cells and 9 over-expressed in normal stromal cells.
Unbiased hierarchical clustering analysis of the samples using these 28
proteins revealed three protein constellations whose variation in expres-
sion defined four MSC protein expression signatures: Class 1, Class 2,
Class 3, and Class 4. These cell populations appear to have clinical rele-
vance. Specifically, patients with Class 3 cells have longer survival and
remission duration compared to other groups. Comparison of leukemia
Correspondence: MSC at first diagnosis with those obtained at salvage (i.e. relapse/refrac-
skornblau@mdanderson.org or
tory) showed differential expression of 9 proteins reflecting a shift
mandreef@mdanderson.org toward osteogenic differentiation. Leukemia MSC are more senescent
compared to their normal counterparts, possibly due to the over-
expressed p53/p21 axis as confirmed by high β-galactosidase staining. In
Received: May 9, 2017. addition, overexpression of BCL-XL in leukemia MSC might give sur-
Accepted: February 1, 2018. vival advantage under conditions of senescence or stress and over-
expressed galectin-3 exerts profound immunosuppression. Together, our
Pre-published: March 15, 2018. findings suggest that the identification of specific populations of MSC in
AML patients may be an important determinant of therapeutic response.
doi:10.3324/haematol.2017.172429

Check the online version for the most updated Introduction


information on this article, online supplements,
and information on authorship & disclosures: There is growing evidence to support the importance of the leukemia bone mar-
www.haematologica.org/content/103/5/810 row (BM) niche in the process of acute myeloid leukemia (AML) chemoresis-
tance.1,2 Hence, optimal therapeutic strategies should also address neighboring cells
in the tumor microenvironment. The critical support cells in the leukemia BM
©2018 Ferrata Storti Foundation microenvironment are mesenchymal stromal cells (MSC).3-8 Depending on the
Material published in Haematologica is covered by copyright. type, MSC can act either to support or suppress tumors.4,8-15 Our group and others
All rights are reserved to the Ferrata Storti Foundation. Use of have found that MSC support leukemia cell survival by diverse mechanisms that
published material is allowed under the following terms and
conditions:
include secretion of cytokines and chemokines, activation of survival signaling in
https://creativecommons.org/licenses/by-nc/4.0/legalcode. tumor cells, and blocking immune surveillance by suppressing natural killer (NK)
Copies of published material are allowed for personal or inter- and T cells.2-5,13
nal use. Sharing published material for non-commercial pur- Mesenchymal stromal cells are essential for human hematopoiesis, particularly
poses is subject to the following conditions:
as a source of SDF-1, which regulates homing, proliferation, and differentia-
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- tion.6,9,10,16-18 Moreover, studies from our group and others have demonstrated that
mercial purposes is not allowed without permission in writing MSC protect leukemia cells from chemotherapy.6,19-23 We have recently found that
from the publisher. there is reciprocal activation of NFkB signaling between MSC and AML and acute
lymphoblastic leukemia (ALL) cells that likely contribute to the effectiveness of the
microenvironment to protect malignant cells.7 Medyouf et al. recently demonstrat-

810 haematologica | 2018; 103(5)


Proteomic profiling reveals complexity of AML MSC

ed that blast cells from myelodysplastic syndrome (MDS) Cell senescence assessment
patients induce changes in MSC reflecting reprograming Microscopy assessment of β-galactosidase staining was used to
of the stromal cells.24 MSC may also influence hematopoi- detect cell senescence using a detection kit from Cell Signal
etic precursors to promote leukemogenesis as evidenced Technology (Boston, MA, USA). Early passage cells (passage 2)
by the development of AML and MDS in mice where the were imaged using a Nikon Coolpix 950 camera attached to a
MSC osteo-progenitors were engineered to lack Dicer, a Nikon TMS light microscope (Nikon Instruments Inc.). AML-MSC
key regulator of microRNA (miR) processing.2 (n=4) and NL-MSC (n=5) were lysed in kit buffer. Measurement of
Furthermore, a recent study from Zhao et al. reported that β-galactosidase was performed using an in vitro fluorometric assay
p21 could be critical for inducing senescence in MSC from with fluorescein di-β-D-galactopyranoside (FDG) as substrate.
MDS patients with concomitant induction of interleukin- Incubation time was 2 hours (h). Fluorescence was measured
6 (IL6) and transforming growth factor β (TGF-β).25 This using an Optima Fluorometer (Durham, NC, USA). Activity is pre-
study is consistent with findings that support the role of sented as fluorescence units/1000 cells/minute.
Dicer in regulating MSC biology and also establish a pos-
sible mechanism of aberrant survival functions in malig- Pathway analysis
nant MSC that may be associated with p21 and senes- String software (String 10.1; available from: http://string-db.org)33
cence. The ability of malignant MSC to withstand senes- was used to determine protein associations. Pathway analysis to
cence may depend on the expression of the anti-apoptotic identify canonical pathways, upstream regulators, and protein net-
molecule BCL-XL.26,27 works was performed using Ingenuity Pathway software
The cellular composition of stromal cells in a cancer (Qiagen).
microenvironment, such as the leukemic BM niche, is like-
ly markedly different from that of the normal BM. We, Results
therefore, set out to study the protein expression and acti-
vation in leukemic MSC (AML-MSC) and compared and Proteins are differentially expressed in AML versus
contracted these to normal MSC (NL-MSC) to determine healthy MSC
if and how they are functionally different. Reverse phase We have routinely utilized RPPA to analyze protein
protein array analysis (RPPA, pioneered in our laborato- expression from clinical samples from many hematologic
ry)28-32 was used to examine expression of 151 proteins in malignancies.28-32 We examined protein expression in blasts
MSC derived from AML BM (n=106) with those derived from newly diagnosed AML patients (n=85), CD34+ cells
from healthy donors (n=71). The results presented here from normal donors (n=10), MSC from healthy donors
identify 28 that were differentially expressed between the (n=71), and MSC from newly diagnosed AML patients
two. Importantly, the 28 proteins identified as differential- (n=54). Both normal MSC and AML-MSC expressed MSC
ly expressed in the AML versus normal MSC could be defining lineage markers CD73, CD90, and CD 105 as
grouped into four protein constellation (PC) expression determined by flow cytometry (Online Supplementary
signatures with different biological properties and clinical Figure S1). MSC from salvage samples (i.e. relapse/refrac-
implications regarding patient response to therapy. tory) were also studied (n=46). The RPPA was probed
with 151 antibodies targeting 119 different proteins (114
targeting total protein with 32 paired antibodies targeting
Methods phosphoepitopes on 26 proteins, and 5 with only a phos-
phoepitope but not total protein epitope) covering a wide
Patients’ samples variety of cellular functions and pathways (Online
Bone marrow was obtained from AML patients (n=106) under- Supplementary Table S1). Protein expression in AML-MSC,
going diagnostic BM aspiration and from healthy donors (n=71) NL-MSC, AML blasts and normal CD34+ cells was com-
who were undergoing BM harvest for use in allogeneic BM trans- pared using principle component analysis (Online
plantation. Samples were acquired in accordance with the regula- Supplementary Figure S1) and unbiased hierarchical cluster-
tions and protocols approved by the Investigational Review Board ing (Online Supplementary Figure S2B). NL-MSC and AML-
of MD Anderson Cancer Center. Informed consent was obtained MSC formed a cluster distinct from AML blasts and NL-
in accordance with the Declaration of Helsinki. Samples were ana- CD34+ cells with the vast majority of the 151 proteins test-
lyzed under an Institutional Review Board-approved laboratory ed showing statistically significant differential expression
protocol. Patients' characteristics are presented in Table 1. Details (143 of 151, P=0.01; 124 of 151, P≤10-6) between MSC and
of isolation of MSC are available in the Online Supplementary the blast/CD34+ cells. This unsurprising observation is
Methods. consistent with a previous report that gene expression
profiles are distinct between blood cells and MSC.34
RPPA Principal component analysis (PCA) also shows that pro-
Proteomic profiling was carried out on MSC samples from tein expression in NL-CD34+ cells is distinct from that of
patients with AML and healthy donors using RPPA. The RPPA AML blasts. These findings were identical to those
method and sample validation technique are described fully else- observed when the analysis was restricted to samples
where.28-32 Antibodies against 151 proteins were used for analysis. from newly diagnosed patients alone.
(A list and the source of the antibodies and the concentrations uti- Next we investigated whether protein expression in
lized is provided in the Online Supplementary Table S1). The sources AML-MSC was different from that of NL-MSC. In PCA,
of antibodies have been been reported previously.30 An IgG sub- the NL-MSC occupied a distinct space from that of the
type-specific secondary antibody was used to amplify the signal, AML-MSC (Figure 1A). Unbiased hierarchical clustering
and finally a stable dye was precipitated. The stained slides were comparing AML-MSC and NL-MSC revealed differential
analyzed using the Microvigene software (version 3.0, Vigene expression of 28 of those proteins (P<0.001; Q=0.0059).
Tech, Carlise, MA, USA) to produce quantified data. Statistical The Q-value, a measure of the false discovery rate deter-
analyses are described in the Online Supplementary Methods. mined by a β-uniform mixture model highlights that these

haematologica | 2018; 103(5) 811


S.M. Kornblau et al.

differences are very unlikely to be random and suggest The proteins clustered into three PCs (from top to bottom
that there are significant differences in the protein expres- in Figure 1B). Nineteen of these proteins had generally
sion patterns of AML-MSC relative to those of NL-MSC. higher expression in AML-MSC, including P1 [STAT1, p-

Table 1. Patients’ demographics (see Figures 2A and 3A).


Variable Category Total Class 2 Class 1 Class 3 Class 4 P
Cohort size Number 101 6 13 18 64
Percentage 100.00% 5.90% 12.90% 17.80% 63.40%
Status New 53.50% 66.70% 53.80% 66.70% 48.40% 0.55
Salvage 45.50% 33.30% 46.20% 33.30% 50.00%
Sex M 61.40% 33.30% 30.80% 72.20% 67.20% 0.03
F 38.60% 66.70% 69.20% 27.80% 32.80%
Race Asian 6.90% 0.00% 7.70% 11.10% 6.30% 0.5
Black 7.90% 16.70% 15.40% 0.00% 7.80%
White 73.30% 50.00% 69.20% 83.30% 73.40%
Hispanic 9.90% 33.30% 7.70% 5.60% 9.40%
Unknown 3.00% 0.00% 7.70% 0.00% 3.10%
FAB M0 4.00% 0.00% 0.00% 5.60% 4.70% 0.21
M1 16.80% 16.70% 38.50% 5.60% 15.60%
M2 24.80% 16.70% 23.10% 22.20% 26.60%
M4 26.70% 66.70% 23.10% 27.80% 23.40%
M5 12.90% 0.00% 7.70% 16.70% 14.10%
M6 3.00% 0.00% 0.00% 11.10% 1.60%
RAEB-T 4.00% 0.00% 0.00% 0.00% 6.30%
Unk 7.90% 0.00% 7.70% 11.10% 7.80%
PS Asymptomatic 15.80% 16.70% 15.40% 11.10% 17.20% 0.44
Symptoms 62.40% 50.00% 46.20% 77.80% 62.50%
In bed <50 13.90% 33.30% 23.10% 5.60% 12.50%
In bed >50 2.00% 0.00% 7.70% 0.00% 1.60%
100% bedridden 1.00% 0.00% 0.00% 5.60% 0.00%
AHD N 75.20% 50.00% 84.60% 83.30% 73.40% 0.33
Y 24.80% 50.00% 15.40% 16.70% 26.60%
Cyto Favorable 2.00% 16.70% 7.70% 0.00% 0.00% 0.026
Intermediate 54.50% 83.30% 53.80% 50.00% 53.10%
Unfavorable 43.60% 0.00% 38.50% 50.00% 46.90%
FLT3 Negative 65.30% 33.30% 69.20% 66.70% 64.10% 0.0979
Positive 20.80% 16.70% 23.10% 16.70% 21.90%
Not Done 8.90% 50.00% 7.70% 16.70% 14.10%
D835 Negative 79.20% 50.00% 92.30% 77.80% 76.60% 0.005
Positive 6.90% 0.00% 0.00% 5.60% 9.40%
Not Done 8.90% 50.00% 7.70% 16.70% 14.10%
NPM1 Negative 46.50% 50.00% 69.20% 72.20% 45.30% 0.08
Positive 10.90% 0.00% 7.70% 0.00% 17.20%
Not Done 5.00% 50.00% 23.10% 27.80% 37.50%
Response CR 58.80% 66.70% 71.40% 63.60% 53.30% 0.8
PR 29.70% 0.00% 0.00% 0.00% 3.30%
HI 1.00% 0.00% 0.00% 9.10% 6.70%
No response 3.00% 33.30% 14.30% 9.10% 13.30%
FAIL 6.90% 0.00% 14.30% 18.20% 23.30%
Relapse Yes 70.00% 100.00% 80.00% 28.60% 81.30% 0.076
Vital Alive 21.60% 0.00% 28.60% 36.40% 16.70% 0.41
Dead 78.40% 100.00% 71.40% 63.60% 83.30%
M: male; F: female; FAB: French-American-British Classification; PS: propensity score; AHD: antecedent hematologic disease; N: no; Y: yes; Cyto: cytogenetic profile; CR: complete
response; PR: partial response; HI: hematologic improvement;*Statistically significant (P) sets are in bold.

812 haematologica | 2018; 103(5)


Proteomic profiling reveals complexity of AML MSC

PDK1 (S241)], CCND1, CDKN1A (p21), ITGA2, PARP1, Although expression of EGFR and ERBB2 expression could
PPP2R2A/B/C/D, the PP2A B regulatory subunit family not be confirmed by western blot analysis, this may reflect
B55, BAK1, CSNK2A1, CDK4, GSK3A/B) and PC2 the enhanced sensitivity of RPPA over standard
[STAT5A/B, BCL2L1 (BCL-XL)], DIABLO, TP53 (p53), immunoblot technology. The remaining nine proteins in
NOTCH 1 (cleaved 1744), SPP1, p-EGFR (Y992), and PC3 were elevated in healthy donor MSC compared to
ERBB2). Expression of 17 of the 19 proteins was validated AML-MSC: SMAD1, CREB1.p133 STMN1, SIRT1, CREB1
by immunoblot analysis (Online Supplementary Figure S3). SMAD4, p-Foxo1/3 (S32), HSP90AA1/B1, and EIF2S1.

Figure 1. Mesenchymal stromal cell (MSC) protein expression signatures. Protein expression is distinct between normal MSC and acute myeloid leukemia (AML)-
MSC. (A) Principal component analysis (PCA) of 151 proteins examined in Class 1 (yellow), Class 2 (light blue), Class 3 (orange) and Class 4 (dark blue). (B) Unbiased
hierarchical clustering identifies 3 protein signature groups: Group 1 (11 members), Group 2 (8 members) and Group 3 (9 members) in Class 1 (yellow), Class 2
(light blue), Class 3 (orange) and Class 4 (dark blue) groups, identified in top row as “MSC protein type”. MSC derived from normal donor (light blue) or AML patient
(dark blue) is shown in the second row marked “cell type”.

haematologica | 2018; 103(5) 813


S.M. Kornblau et al.

Table 2. List of proteins with reverse correlation to one or more other proteins in acute myeloid leukemia (AML) mesenchymal stromal cells (MSC)
versus normal MSC.
Protein AML-MSC negative AML-MSC positive
normal MSC positive normal MSC negative
AKT1S1 STAT3
AKT3 BCL2L11, ERG, SFN, SRCp416
ARC STAT3 p727 CTNNB1
ATF3 BCL2, KIT, SFN, TP53 p15
BCL2 ATF3, CTNNB1
BCL2L11 ATF3, CTNNB1
BECN1 PSMD9, YWHAE
BID GAPDH BIRC5, GAB2 p452
BIRC5 MS4A1, FN1, SRC p416 BID, EIF2S1 p51, IRS1 p1101, RPS6KB1
CAV1 PTK2
CCNB1 MAPK1.3 p202.p204
CTNNB1 BCL2, BCL2L11, FOXO1.3 p24.p32, FOXO3, KIT, ITGAL, ARC, HSP90AA1.B1, MAPK9, PTGS2, YWHAZ
PSMD9 p10, RPS6KB1 p389, SFN, SRC, SRC p416
CDK1 CREB1
CDK4 GAPDH, MAPK1.3 p202.p204, YWHAE
CREB1 CDK1, JUN p73 DIABLO, PECAM1, SFN
CREB2 p133 MAPK9 TP53
DIABLO HSPB1 CREB1
EGLN1 FOXO1.3 p24.p32, FOXO3, MS4A1, SRC p416, TCF4,
YWHAE
EIF2S1 p51 BIRC5, TGM2
ELK1 p383 SMAD1
ERBB2 p1248 TCF4
ERG AKT3
FN1 BIRC5
FOXO1.3/FOXO3 p24.p32 CTNNB1 EGLN1
FOXO3 CTNNB1, IRS1 p1101 EGLN1
GAB2 p452 BID
GAPDH BID, CDK4, GSK3A.B
GSK3A.B GAPDH, STK11
HDAC3 PSMD9
HNRNPK STAT5A.B
HSP90AA1.B1 CTNNB1
HSPB1 DIABLO
IRS1 p1101 FOXO3 BIRC5
ITGAL CTNNB1
JMJD6 STAT3 p727
JUN p73 CREB1 NRP1
KIT ATF3, CTNNB1
LEF1 PTK2
MAP2K1 XIAP
MAPK1.3 p202.p204 CCNB1, CDK4 SRC p527
MAPK9 CREB2 p133 CTNNB1
MAPK14 STAT1
MS4A1 BIRC5 EGLN1
NPM1 SMAD1
NR4A1 PECAM1
NRP1 JUN p73
PECAM1 NR4A1 CREB1
continued on the next page

814 haematologica | 2018; 103(5)


Proteomic profiling reveals complexity of AML MSC

continued from the previous page

PPARG SMAD1
PRKAA1.2 p172 SRC p527
PSMD9 p10 CTNNB1
PSMD9 BECN1, HDAC3
PTGS2 CTNNB1
PTK2 LEF1 CAV1
RPS6KB1 p389 CTNNB1
RPS6KB1 BIRC5
SFN AKT3, ATF3, CTNNB1 CREB1, STMN1
SMAD1 ELK1 p383 NPM1, PPARG,
SRC CTNNB1
SRC p416 AKT3, BIRC5, CTNNB1, XIAP EGLN1
SRC p527 PRKAA1.2 p172, MAPK1.3 p202.p204
STAT1 MAPK14, STAT5A.B
STAT3 p727 AKT1S1
STAT3 p727 ARC, JMJD6
STAT5A.B STAT1 HNRNPK
STK11 GSK3A.B
STMN1 BCL2L11, SFN
TCF4 ERBB2 p1248 EGLN1
TGM2 EIF2S1 p51
TP53 CREB2 p133
TP53 p15 ATF3
XIAP SRC p416 MAP2K1
YWHAE BECN1, CDK4 EGLN1
YWHAZ CTNNB1

Based on various combinations of proteins expressed Clinical correlation


within these three groups, we observed that normal or Among the AML cases, there were very few clinical or
leukemic MSC samples clustered into four distinct PCs. laboratory features that showed a statistically significant
The first (aqua in Figure 1A; top row of annotations above correlation with the four MSC PCs. Specifically there was
the heat map in Figure 1B), was comprised predominantly no association for continuous variables: age, percent BM
of NL-MSC (42 of 46 members), characterized by lower or blood blasts, hemoglobin, platelet count, serum lactate
expression of proteins in constellations 1 and 2, and higher dehydrogenase, albumin, creatinine, bilirubin fibrinogen,
expression of constellation 3 proteins. This was consid- CD7, CD10, CD13, CD14, CD20, CD33 or CD34, or dis-
ered to represent the protein expression pattern of NL- crete variables disease status (new vs. salvage), race,
MSC. AML cases in this signature are hereafter called French-American-British (FAB) Classification, perform-
Class 2. ance status, history of an antecedent hematologic disor-
A second signature (blue in Figure 1A and B) was com- der, presence of an FLT3-ITD or presence of an NPM1
prised predominantly of AML-MSC (62 of 72 members), mutation (Table 1). However, a few notable associations
and was characterized by the opposite protein expres- between MSC protein signature and clinical features were
sion of the NL-MSC signature, with high expression of observed. There was a statistical difference in presence of
PC1 and 2 proteins and lower expression of PC3 pro- FLT3 D835 mutation (P=0.05). While no Class 1 or Class 2
teins. This signature is called Class 4. Two groups with samples contained FLT3 D835 mutation, 9.4% of Class 4
expression signatures between that of the Class 2 MSC MSC and 5.6% of Class 3 MSC had the mutation (Table
and Class 4 MSC were also recognized. One signature 1). Interestingly, there was a statistical difference in MSC
(yellow in Figure 1A and top row of Figure 1B), com- population types between men and women (P=0.03)
prised of 12 AML-MSC and 11 NL-MSC, mirrored the (Table 1). Men had a higher percentage of Class 4 and
expression of Class 2 for protein constellation 1, but that Class 3 MSC while women had a higher percentage of
of Class 4 for PC2 and 3. AML cases in this group are Class 1 and Class 2 MSC. At present it is unclear if sex
hereafter referred to as Class 1. The final signature influences MSC biology in the leukemic niche. Sex-specif-
(orange in Figure 1A and top row of Figure 1B), consist- ic effects in the microenvironment in leukemia is not
ing of 16 AML-MSC and 8 NL-MSC, mirrored the unprecedented as integrin-mediated adhesion-triggered
expression of Class 4 for PC1 and 3 but had low expres- activation of GSK3B occurs exclusively in leukemia pro-
sion of PC2, similar to Class 2. AML cases from this sig- genitor cells derived from female AML patients.35 Total
nature are referred to as Class 3. GSK3 and ITGA2 are present in protein constellation 1

haematologica | 2018; 103(5) 815


S.M. Kornblau et al.

and the proteins in this constellation are elevated in Class These findings suggest that the disease state dictates
3 and Class 4 while they are lower in Class 1 and Class 2. expression of at least some of the identified proteins in
Perhaps the integrin/GSK3 axis is also important in AML- MSC.
MSC.
Finally, there were statistically significant biases with Evidence for dysregulated signaling in AML-MSC
different cytogenetic groups associating with different The observed differences in protein expression between
MSC protein signatures (P=0.026) (Table 1). Specifically, AML-MSC and NL-MSC suggest that pathway utilization
although few patients had favorable cytogenetics in this may be dysregulated or non-canonical in the AML-MSC.
dataset, these were found exclusively in the Class 1 and To look for evidence of abnormal utilization, we searched
Class 2 signatures. Conversely, patients with unfavor- for statistically significant (R>0.2; P< 0.0001) protein-pro-
able cytogenetics were not observed in the Class 2 sig- tein correlations that were reversed in the direction of cor-
nature, but they were found in relatively equal propor- relation in the AML-MSC compared to the pattern in the
tions in the three AML specific signatures (i.e. Classes 1, NL-MSC. A representative analysis of STAT5 expression
3, and 4). with the other proteins in NL-MSC revealed that this tran-
Having determined that leukemic MSC divide AML scription factor is negatively correlated with hnRPK and
patients into discrete protein signatures, we next exam- positively correlated with STAT1 (Online Supplementary
ined whether signature membership affected outcomes. Figure S5B). However, in AML-MSC the correlations are
There was no significant difference in median overall sur- reversed (Online Supplementary Figure S5B).
vival between the four MSC populations, although the A list of all protein correlations that are reversed in
median survival of 105 weeks (wk) in the Class 3 signature AML-MSC compared to NL-MSC is provided in Table 2.
was longer than the other three groups at 25, 48 and 58 Of note, there are significant changes in protein correla-
wk (Figure 2A); this suggested that patients with Class 3 tions involving β-catenin. As shown in Table 2, BCL2,
MSC may have a better disease prognosis. Despite the BCL2L11, FOXO1.3 p24.p32, FOXO3, KIT, ITGAL,
small sample size, relapse rates among the newly diag- PSMD9 p10, RPS6KB1 p389, SFN, SRC, and SRC p416
nosed cases were different, with 4 of 5 Class 1 MSC are negatively correlated with β-catenin in NL-MSC but
patients and 13 of 16 Class 4 MSC patients relapsing com-
pared to only 2 of 7 Class 3 MSC cases (P=0.076).
Furthermore the median remission duration was different
between patients with Class 3 MSC and the three other A
signatures (101 wk for Class 3 MSC vs. 42.2 wk for Class
4 MSC; P=0.05) (Figure 2B). Other comparisons were not
made due to the small sample size. These findings suggest
that MSC can influence patient survival, although at pres- P=0.26
ent the mechanism is unknown.

Effect of age on protein expression in NL-MSC and P=0.6


AML-MSC
As AML is a disease of the elderly, many of the AML-
MSC samples were obtained from individuals aged over
60 years (y). Of the 106 AML-MSC samples, 50 samples
were from individuals under 60 y. Of the 71 MSC samples
from healthy donors, 68 samples were from individuals
under 60 y. To assess the impact of age on the differences
in protein expression between the AML-MSC and NL-
MSC groups, the protein expression of 24 of the differen- B
tially-expressed proteins was reassessed in age-matched
sets. The three groups were: a) under 30 y (AML n=13; NL
n=37); b) 30-49 y (AML n=17; NL n=25); and c) 50 to 59 y
(AML n=20; NL n=6). As shown in Online Supplementary
Table S2 and Online Supplementary Figure S4A, of the 19
proteins elevated in AML-MSC compared to NL-MSC,
only two (i.e. BCL-XL and PPP2R2A/B/C/D) were signifi-
cantly higher in all age groups. This suggests that
increased expression of these 2 proteins is dependent on P=0.05
the disease state and not on age. Six other proteins includ-
ing p53 and CDKN1A (p21) were also elevated in two of
the three age groups suggesting expression of these pro-
teins is also dependent on the disease state rather than on P=0.12
age (Online Supplementary Table S2).
Analysis of p53 and CDKN1A (p21) protein expression
was performed by western blot analysis on age-matched
AML and NL samples (age 40-49 y; n=3 for both groups).
Protein expression of both p53 and CDKN1A (p21) was at Figure 2. Survival and remission duration differ in patients based on mesenchy-
least 2- to 3-fold higher in AML-MSC compared to the mal stromal cell (MSC) population. Kaplan-Meir curves showing overall survival
(A) and remission duration (B). N: number.
healthy donor samples (Online Supplementary Figure 4B).

816 haematologica | 2018; 103(5)


Proteomic profiling reveals complexity of AML MSC

are positively correlated with the protein in AML-MSC. Proteins differentially expressed in AML-MSC share
ARC (NOL3), HSP90AA1/B1, MAPK9, PTGS2, and interactomes
YWHAZ were positively correlated with β-catenin in To assess the relationship among the proteins identified
NL-MSC but are negatively correlated with the protein in the RPPA analysis, protein association network analysis
in AML-MSC. Ingenuity Pathway Analysis (IPA) was was performed using STRING 10.533 on proteins identi-
performed using software on the proteins identified as fied as significantly different in the AML-MSC and NL-
differentially correlated with β-catenin in the MSC sets MSC (Figure 1B). Blalock et al. used a previous version of
and β-catenin itself (i.e. CTNNB1; BCL2; BCL2L11; String software to map the nuclear interactome.36 In cases
FOXO1; FOXO3; KIT; ITGAL; PSMD9; RPS6KB1; SFN; where a family of proteins was identified (e.g. PPP2R2 set
SRC; NOL3; HSP90AA1; HSP90B1; MAPK9; PTGS2; and HSP90 set), a representative member was included in
YWHAZ). IPA revealed these proteins were highly asso- the analysis. With the exception of PDK-1 all proteins are
ciated with PI3K/AKT signaling (top canonical pathway; interconnected at least through one association (Figure 3).
P=7.35E-19) (Online Supplementary Figure S5). IPA also This finding suggests that there is an interconnection
identified the top upstream regulator of this set of pro- between the various proteins that are distinctly expressed
teins as p53 (P=1.04E-11). between the NL-MSC and AML-MSC groups.

Activation
Inhibition
Binding
Phenotype
Catalysis
Post-translational regulation
Reaction
Transcriptional regulation

Figure 3. Proteins differentially expressed in acute myeloid leukemia (AML) and normal mesenchymal stromal cell (MSC) are highly interactive. (A) String analysis
was performed by String 10.0 using interactions based on “action”; available from: http://string-db.org. (B) Model of involvement of Group 1 and 2 proteins in AKT
signaling. Red: proteins are members of Group 1 or 2. Yellow; proteins are non-members but possible links.

haematologica | 2018; 103(5) 817


S.M. Kornblau et al.

Pathway analysis suggests NL-MSC have prominent was identified as one of the top three upstream regulators
adipogenic signaling while all AML-MSC populations (P=1.22E-14), suggesting that signaling mediated by this
have prominent PI3K/AKT signaling survival kinase is prominent in AML-MSC.
Pathway analysis was performed using IPA. Two sepa-
rate data sets were created: one with proteins from PC3 AML-MSC are senescent compared to NL-MSC
(elevated in normal MSC) and one with proteins from PC1 The p21 protein appears to be critical for senescence in
and 2 (elevated in AML-MSC). Proteins in group 3 are myelodysplastic syndrome (MDS)-MSC22 and AML-
associated with adipogenesis (ninth top canonical path- MSC similar to MDS-MSC have elevated p21 (Figure 1B).
way; P=2.19E-05) (Online Supplementy Figure S6). PC3 pro- This finding suggests that AML-MSC might also be more
teins are elevated in MSC that were presumably normal senescent than NL-MSC, as was the case in MDS-MSC.25
but reduced in AML-MSC suggesting differences in differ- Senescence was observed in normal donor MSC- and
entiation potential of MSC between NL-MSC and AML- AML-derived MSC using β-galactosidase staining. AML-
MSC. Of the 7 proteins identified, SIRT1, FOXO1, and MSC were more senescent than MSC derived from
SMAD1 each can activate PPARβ which is a critical regu- healthy donors in this representative example (Figure 4A).
lator of adipocyte differentiation.37-39 PC3 also displayed To account for age effects on senescence, MSC were taken
the strongest association with AMPK signaling (P=6.84E- from donors under 58 y. Average age of the AML patients
07). The top upstream regulator identified in the set of (n=4) was 52 y and the average age of normal donors (n=5)
PC3 proteins was PDGFB (P=1.01E-06). PI3K/AKT path- was 47 y. Also, MSC of similar cell passage (passage 2 or
way was highly associated with PC1 and 2 proteins, 3) were used, so effects of cell passage were unlikely. As
which are elevated in AML-MSC (top canonical path- shown in Figure 4B, β-galactosidase activity was signifi-
ways; P=7.16E-17) (Online Supplementary Figure S7). AKT cantly higher (almost 2-fold) in AML-MSC compared to

Figure 4. Acute myeloid leukemia (AML) mesenchymal


stromal cell (AML-MSC) are more senescent than nor-
mal MSC (NL-MSC). (A) Representative microscopy of an
AML-MSC and a normal MSC with two different slide
areas. (B). Level of β-galactosidase was assessed by
enzymatic assay in normal MSC (n=5) and AML-MSC
(n=4). Statistical significance determined by Student
t-test; *P=0.027.

818 haematologica | 2018; 103(5)


Proteomic profiling reveals complexity of AML MSC

NL-MSC (P=0.032). These findings suggest that AML- covery that AML-MSC have significantly different protein
MSC tend toward senescence. expression patterns compared to normal MSC, with 28 of
151 analyzed proteins being highly significantly different
Therapy alters proteins expression in AML-MSC (FDR<0.006). These changes assumed four signatures in
Protein expression in AML-MSC might change between AML, 81% of which were very different from that of nor-
diagnosis and relapse, perhaps as a result of relapse, or in mal MSC, while 6% had an identical signature to NL-
response to acquired changes in AML blasts. To determine MSC, and another 13% were more like the normal signa-
if AML-MSC protein expression was changing between ture than the leukemia patterns. Signature membership
diagnosis and relapse we compared expression between showed an association with cytogenetics, with 'favorable'
the samples obtained at first diagnosis (n=53) to those of cytogenetics being limited to the more NL-MSC-like pat-
AML-MSC collected from primary refractory or relapsed tern and 'unfavorable' cytogenetics not occurring in AML
patients (n=54). Nine proteins are differentially expressed with an NL-MSC-like pattern. There was a difference in
between the two MSC groups (Figure 5). the distribution of the MSC population between men and
Phosphorylated β-catenin, phosphorylated RPS6, and women. The significance of these differences is not clear,
galectin-3 are expressed at higher levels in MSC in the sal- but women tended to have higher percentages of Class 1
vage set. SMAD6, TCF4, LYN, integrin-β3, phosphorylat- and Class 2 MSC compared to men. In leukemia progeni-
ed EIF4BP1, and phosphorylated ELK1 are higher in MSC tor cells, GSK3B is activated via an integrin-mediated
at first diagnosis compared to MSC from salvage AML mechanism in response to adhesion to a stromal cell
patients. IPA reveals that, for canonical pathways, a set exclusively in women patients.35 As ITGA2 and GSK3 are
associated with osteoblast differentiation was found to be members of a protein constellation (i.e. constellation 1)
the pathway most associated with the proteins differen- that is differentially expressed in Class 1 and Class 2
tially expressed at diagnosis compared to salvage (Online (lower levels) compared to Class 3 and Class 4 (higher lev-
Supplementary Figure S8). els), it is tempting to speculate that integrin/GSK3 axis
may contribute to sex-specific effects in MSC.
Furthermore, these signatures influence outcomes
Discussion including response rates, remission duration and, perhaps,
survival. Patients with Class 3 MSC fare much better than
This study presents the first systematic study of protein patients with Class 4 MSC as demonstrated by significant-
expression differences between NL-MSC and AML-MSC. ly longer remission duration and a trend toward longer
There were several notable observations. There were clear OS. Changes in protein expression were often character-
differences between the protein expression of MSC ized by protein-protein correlations that were reversed
(whether from healthy donor or AML patient) and AML from those seen in normal MSC, providing insight into the
blast cells. This result was not surprising as one would nature of this dysregulation and potentially providing
predict different proteins would be prominent in mes- therapeutic targets. In NL-MSC, the signature proteins were
enchymal cells and cells of hematopoietic/myeloid line- associated with adipocyte differentiation. That normal
age. The major observation of this research was the dis- MSC, but not AML-MSC, possess protein pathways impor-

Figure 5. A distinct set of proteins is associated with acute myeloid leukemia (AML) patient salvage status. (A) Reverse phase protein arrays (RPPA) reveals protein
expression in AML mesenchymal stromal cells (MSC) differs between diagnosis and the salvage setting for 9 proteins (P=0.05; false discovery rate=0.68).

haematologica | 2018; 103(5) 819


S.M. Kornblau et al.

tant in adipogenesis is consistent with other studies from results in differentiation to adipocytes via a mechanism
our group and another group that found AML-MSC are involving loss of β-catenin.48 As pathway analysis of pro-
primed toward osteoblastic differentiation and not teins associated with normal MSC (i.e. Constellation 3)
adipocytic differentiation.9,40,41 We reported that AML-MSC pathways identifies adipogenesis as a key pathway, it will
express higher levels of osteogenic markers, including be important to determine if AML-MSC would be less
Tissue Non-specific Alkaline Phosphatase (TNAP), RUNX2, likely to skew toward adipocyte differentiation because of
Osterix, and Ostepontin, compared to MSC from age- a PP2A B55 a subunit/β-catenin axis.
matched heathy donors.40 In addition, in that study we BCL-XL is necessary for MSC survival during differenti-
found that AML-MSC readily differentiate along the ation.27 AML-MSC expressed higher BCL-XL and Cyclin
osteogenic lineage pathway but are unable to differentiate D1 protein. These findings may be attributed to STAT5 as
into adipocytes. This differentiation potential of the MSC this transcription factor is a regulator of both BCL-XL and
may be influenced by the leukemia cells themselves, as Cyclin D1.49 We found by qRT-PCR that gene expression
exposure of healthy donor MSC to AML cell lines such as of both BCL-XL and Cyclin D1 was higher in AML-MSC
OCI-AML3 induces gene expression of RUNX2, TNAP, and (n=9) compared to normal MSC (n=10) (Online
other osteogenic genes, and induces osteogenic differentia- Supplementary Figure S9). These findings suggest that ele-
tion of the stromal cells.40 The protein networks prevalent in vation of BCL-XL and Cyclin D1 protein in AML-MSC can
NL-MSC that we identify here are consistent with signaling be attributed at least in part to a transcriptional mecha-
that skews toward adipocytic differentiation in the normal nism possibly involving STAT5.
cells. Diaz de la Guardia et al. found that MSC from a high- Two prominent proteins identified as elevated in the
risk AML group failed to differentiate into adipcocytes.41 AML-MSC group are p21 and p53 (Figure 1B). Elevated
The IPA data on canonical pathways in the AML-MSC expression of p21 and increased senescence of MSC is
compared at first diagnosis with refractory and relapse sam- consistent with the study on MDS-MSC that showed a
ples suggests the importance of MSC of the osteoblastic lin- role for p21 in IL-6 and TGF-β production.25 However, a
eage in the AML niche, as many of these proteins are asso- recent study from Desbourdes et al. found p21 and p53
ciated with osteoblast survival and differentiation. levels were similar between AML-MSC and healthy
PI3K/AKT is very prevalent in group 1 proteins which donor-derived MSC.50 The reason for the difference
are associated with Class 4 MSC by IPA. We have previ- between our results and their results is not clear. It should
ously reported that leukemia cells in co-culture with MCS be noted that the Desbourdes et al. study used less than 5
induce activation of AKT and other survival kinases.42 It is samples each of AML-derived MSC and healthy donor-
plausible that the observed activation of AKT signaling in derived MSC to determine p21 and p53 levels, so perhaps
AML-MSC is due to the presence of leukemia cells in the Class 1 or Class 2 MSC (which would have lower levels of
niche or at least that the malignant cells may contribute to p21 and p53) are over-represented in their samples.50 In
activation. In MSC, AKT has been implicated in positive addition, the average age of the AML patients in the
regulation of Cyclin D1 (CCND1) and CDK4.43,44 The pres- Desbourdes et al. study was 49 y while the average age of
ence of the PP2A B55 a subunit in group 1 suggests that the healthy donors was near 60 y, so perhaps the p21 and
either the protein phosphatase is not active against AKT in p53 levels in the healthy donor MSC are skewed higher as
those cells or that AKT is less active in the AML-Like MSC the donors are older than the patients. For p21 and p53
compared to Normal-Like MSC. Despite its tumor sup- expression, an age match comparison of p21 and p53 in
pressor role in suppression of AKT signaling, the PP2A the AML-MSC and NL-MSC shows levels of these pro-
subunit does support β-catenin expression by dephospho- teins are higher in the AML-MSC in at least 2 ages for each
rylating serine and threonine residues that are required for (Online Supplementary Table S2).
destruction of the transcription factor (e.g. serine 33, thre- In conclusion, proteomic analysis identified a distinct
onine 41).45-47 It is interesting that when expression of the set of proteins that distinguish normal MSC from AML-
151 proteins surveyed by RPPA are correlated in the nor- MSC. Our RPPA studies identified four major signatures
mal MSC and in the AML-MSC, β-catenin exhibits the of MSC in AML patients that may impact their function in
greatest difference in correlation pattern between normal the tumor microenvironment.
MSC and AML-MSC compared to the other proteins. It is
plausible that the PP2A B55 a subunit may be a factor in Funding
this phenomenon, though further investigation will be This work was supported in part by the research funding from
required to verify this potential mechanism. It is interest- the National Institutes of Health (P01CA49639 and MD
ing to note that suppression of PP2A (albeit via the catalyt- Anderson Cancer Center Support Grant CA016672) and the
ic core subunit Ca) in MSC cell line or pre-adipocyte cells Paul and Mary Haas Chair in Genetics (to MA).

3. Barcellos-de-Souza P, Gori V, Bambi F, MJ. The life and fate of mesenchymal stem
References Chiarugi P. Tumor microenvironment: bone cells. Front Immunol. 2014;5:148.
1. Konopleva M, Tabe Y, Zeng Z, Andreeff M. marrow-mesenchymal stem cells as key 6. Greenbaum A, Hsu YM, Day RB, et al.
Therapeutic targeting of microenvironmen- players. Biochim Biophys Acta. CXCL12 in early mesenchymal progenitors
tal interactions in leukemia: mechanisms 2013;1836(2):321-335. is required for haematopoietic stem-cell
and approaches. Drug Resist Updat. 4. da Silva Meirelles L, Chagastelles PC, Nardi maintenance. Nature. 2013;495(7440):227-
2009;12(4-5):103-113. NB. Mesenchymal stem cells reside in virtu- 230.
2. Tabe Y, Konopleva M. Advances in under- ally all post-natal organs and tissues. J Cell 7. Jacamo R, Chen Y, Wang Z, et al. Reciprocal
standing the leukaemia microenvironment. Sci. 2006;119(Pt 11):2204-2213. leukemia-stroma VCAM-1/VLA-4-depen-
Br J Haematol. 2014;164(6):767-778. 5. Eggenhofer E, Luk F, Dahlke MH, Hoogduijn dent activation of NF-kappaB mediates

820 haematologica | 2018; 103(5)


Proteomic profiling reveals complexity of AML MSC

chemoresistance. Blood. 2014;123(17):2691- CLL: implications for rituximab resistance in Identification of the PKR nuclear interac-
2702. CLL. Br J Haematol. 2015;169(2):211-218. tome reveals roles in ribosome biogenesis,
8. Studeny M, Marini FC, Dembinski JL, et al. 22. Xia B, Tian C, Guo S, et al. c-Myc plays part mRNA processing and cell division. J Cell
Mesenchymal stem cells: potential precur- in drug resistance mediated by bone mar- Physiol. 2014;229(8):1047-1060.
sors for tumor stroma and targeted-delivery row stromal cells in acute myeloid leukemia. 37. Ali AT, Hochfeld WE, Myburgh R, Pepper
vehicles for anticancer agents. J Natl Cancer Leuk Res. 2015;39(1):92-99. MS. Adipocyte and adipogenesis. Eur J Cell
Inst. 2004;96(21):1593-1603. 23. Zeng Z, Shi YX, Samudio IJ, et al. Targeting Biol. 2013;92(6-7):229-236.
9. Battula VL, Chen Y, Cabreira Mda G, et al. the leukemia microenvironment by CXCR4 38. Jin W, Takagi T, Kanesashi SN, et al.
Connective tissue growth factor regulates inhibition overcomes resistance to kinase Schnurri-2 controls BMP-dependent adipo-
adipocyte differentiation of mesenchymal inhibitors and chemotherapy in AML. genesis via interaction with Smad proteins.
stromal cells and facilitates leukemia bone Blood. 2009;113(24):6215-6224. Dev Cell. 2006;10(4):461-471.
marrow engraftment. Blood. 2013; 122(3): 24. Medyouf H, Mossner M, Jann JC, et al. 39. Kauppinen A, Suuronen T, Ojala J,
357-366. Myelodysplastic cells in patients reprogram Kaarniranta K, Salminen A. Antagonistic
10. Chen Y, Jacamo R, Shi YX, et al. Human mesenchymal stromal cells to establish a crosstalk between NF-kappaB and SIRT1 in
extramedullary bone marrow in mice: a transplantable stem cell niche disease unit. the regulation of inflammation and metabol-
novel in vivo model of genetically controlled Cell Stem Cell. 2014;14(6):824-837. ic disorders. Cell Signal. 2013;25 (10):1939-
hematopoietic microenvironment. Blood. 25. Zhao Y, Wu D, Fei C, et al. Down-regulation 1948.
2012;119(21):4971-4980. of Dicer1 promotes cellular senescence and 40. Battula VL, Le PM, Sun JC, et al. AML-
11. Kidd S, Spaeth E, Watson K, et al. Origins of decreases the differentiation and stem cell- induced osteogenic differentiation in mes-
the tumor microenvironment: quantitative supporting capacities of mesenchymal stro- enchymal stromal cells supports leukemia
assessment of adipose-derived and bone mal cells in patients with myelodysplastic growth. JCI Insight. 2017;2(13).
marrow-derived stroma. PLoS One. syndrome. Haematologica. 2015; 41. Diaz de la Guardia R, Lopez-Millan B,
2012;7(2):e30563. 100(2):194-204. Lavoie JR, et al. Detailed Characterization of
12. Ling X, Marini F, Konopleva M, et al. 26. Oliver L, Hue E, Priault M, Vallette FM. Basal Mesenchymal Stem/Stromal Cells from a
Mesenchymal Stem Cells Overexpressing autophagy decreased during the differentia- Large Cohort of AML Patients
IFN-beta Inhibit Breast Cancer Growth and tion of human adult mesenchymal stem Demonstrates a Definitive Link to
Metastases through Stat3 Signaling in a cells. Stem Cells Dev. 2012; 21(15):2779- Treatment Outcomes. Stem Cell Reports.
Syngeneic Tumor Model. Cancer 2788. 2017;8(6):1573-1586.
Microenviron. 2010;3(1):83-95. 27. Oliver L, Hue E, Rossignol J, et al. Distinct 42. Tabe Y, Jin L, Tsutsumi-Ishii Y, et al.
13. Poggi A, Giuliani M. Mesenchymal Stromal roles of Bcl-2 and Bcl-Xl in the apoptosis of Activation of integrin-linked kinase is a crit-
Cells Can Regulate the Immune Response in human bone marrow mesenchymal stem ical prosurvival pathway induced in
the Tumor Microenvironment. Vaccines cells during differentiation. PLoS One. leukemic cells by bone marrow-derived
(Basel). 2016;4(4). 2011;6(5):e19820. stromal cells. Cancer Res. 2007;67(2):684-
14. Spaeth E, Klopp A, Dembinski J, Andreeff 28. Kornblau SM, Singh N, Qiu Y, Chen W, 694.
M, Marini F. Inflammation and tumor Zhang N, Coombes KR. Highly phosphory- 43. Gharibi B, Ghuman MS, Hughes FJ. Akt-
microenvironments: defining the migratory lated FOXO3A is an adverse prognostic fac- and Erk-mediated regulation of proliferation
itinerary of mesenchymal stem cells. Gene tor in acute myeloid leukemia. Clin Cancer and differentiation during PDGFRbeta-
Ther. 2008;15(10):730-738. Res. 2010;16(6):1865-1874. induced MSC self-renewal. J Cell Mol Med.
15. Spaeth EL, Labaff AM, Toole BP, Klopp A, 29. Kornblau SM, Womble M, Qiu YH, et al. 2012;16(11):2789-2801.
Andreeff M, Marini FC. Mesenchymal Simultaneous activation of multiple signal 44. Lai VK, Ashraf M, Jiang S, Haider K.
CD44 expression contributes to the acquisi- transduction pathways confers poor prog- MicroRNA-143 is a critical regulator of cell
tion of an activated fibroblast phenotype via nosis in acute myelogenous leukemia. cycle activity in stem cells with co-overex-
TWIST activation in the tumor microenvi- Blood. 2006;108(7):2358-2365. pression of Akt and angiopoietin-1 via tran-
ronment. Cancer Res. 2013; 73(17):5347- 30. Ruvolo PP, Qiu Y, Coombes KR, et al. scriptional regulation of Erk5/cyclin D1 sig-
5359. Phosphorylation of GSK3alpha/beta corre- naling. Cell Cycle. 2012;11(4):767-777.
16. Kim JA, Shim JS, Lee GY, et al. lates with activation of AKT and is prognos- 45. Hein AL, Seshacharyulu P, Rachagani S, et al.
Microenvironmental remodeling as a tic for poor overall survival in acute myeloid PR55alpha Subunit of Protein Phosphatase
parameter and prognostic factor of heteroge- leukemia patients. BBA Clin. 2015;4:59-68. 2A Supports the Tumorigenic and
neous leukemogenesis in acute myeloge- 31. Ruvolo PP, Qui YH, Coombes KR, et al. Low Metastatic Potential of Pancreatic Cancer
nous leukemia. Cancer Res. 2015; 75(11): expression of PP2A regulatory subunit Cells by Sustaining Hyperactive Oncogenic
2222-2231. B55alpha is associated with T308 phospho- Signaling. Cancer Res. 2016; 76(8):2243-
17. Raaijmakers MH, Mukherjee S, Guo S, et al. rylation of AKT and shorter complete remis- 2253.
Bone progenitor dysfunction induces sion duration in acute myeloid leukemia 46. Stamos J, Weis W. The β-catenin destruction
myelodysplasia and secondary leukaemia. patients. Leukemia. 2011; 25(11):1711-1717. complex. Cold Spring Harb Perspect Biol
Nature. 2010;464(7290):852-857. 32. Tibes R, Qiu Y, Lu Y, et al. Reverse phase 2013:5: a007898.
18. Shi C. Recent progress toward understand- protein array: validation of a novel proteom- 47. Zhang W, Yang J, Liu Y, et al. PR55 alpha, a
ing the physiological function of bone mar- ic technology and utility for analysis of pri- regulatory subunit of PP2A, specifically reg-
row mesenchymal stem cells. Immunology. mary leukemia specimens and hematopoiet- ulates PP2A-mediated beta-catenin dephos-
2012;136(2):133-138. ic stem cells. Mol Cancer Ther. phorylation. J Biol Chem. 2009; 284(34):
19. Balakrishnan K, Burger JA, Quiroga MP, et 2006;5(10):2512-2521. 22649-22656.
al. Influence of bone marrow stromal 33. Szklarczyk D, Franceschini A, Wyder S, et 48. Okamura H, Yang D, Yoshida K, Teramachi
microenvironment on forodesine-induced al. STRING v10: protein-protein interaction J, Haneji T. Reduction of PP2A Calpha stim-
responses in CLL primary cells. Blood. networks, integrated over the tree of life. ulates adipogenesis by regulating the
2010;116(7):1083-1091. Nucleic Acids Res. 2015;43(Database Wnt/GSK-3beta/beta-catenin pathway and
20. Kojima K, McQueen T, Chen Y, et al. p53 issue):D447-452. PPARgamma expression. Biochim Biophys
activation of mesenchymal stromal cells par- 34. Huang JC, Basu SK, Zhao X, et al. Acta. 2014;1843(11):2376-2384.
tially abrogates microenvironment-mediat- Mesenchymal stromal cells derived from 49. Grad JM, Zeng XR, Boise LH. Regulation of
ed resistance to FLT3 inhibition in AML acute myeloid leukemia bone marrow Bcl-xL: a little bit of this and a little bit of
through HIF-1alpha-mediated down-regula- exhibit aberrant cytogenetics and cytokine STAT. Curr Opin Oncol. 2000;12(6):543-549.
tion of CXCL12. Blood. 2011; 118(16):4431- elaboration. Blood Cancer J. 2015;5:e302. 50. Desbourdes L, Javary J, Charbonnier T, et al.
4439. 35. Bertrand J, Despeaux M, Joly S, et al. Sex dif- Alteration Analysis of Bone Marrow
21. Marquez ME, Hernandez-Uzcategui O, ferences in the GSK3beta-mediated survival Mesenchymal Stromal Cells from De Novo
Cornejo A, Vargas P, Da Costa O. Bone mar- of adherent leukemic progenitors. Acute Myeloid Leukemia Patients at
row stromal mesenchymal cells induce Oncogene. 2012;31(6):694-705. Diagnosis. Stem Cells Dev. 2017;26(10):
down regulation of CD20 expression on B- 36. Blalock WL, Piazzi M, Bavelloni A, et al. 709-722.

haematologica | 2018; 103(5) 821


ARTICLE Acute Myeloid Leukemia

Clinical relevance of IDH1/2 mutant allele


Ferrata Storti
Foundation burden during follow-up in acute myeloid
leukemia. A study by the French ALFA group
Yann Ferret,1,2,3 Nicolas Boissel,4,5 Nathalie Helevaut,1 Jordan Madic,6 Olivier
Nibourel,1,2,3 Alice Marceau-Renaut,1,2,3 Maxime Bucci,1 Sandrine Geffroy,1 Karine
Celli-Lebras,7 Sylvie Castaigne,8 Xavier Thomas,9 Christine Terré,10 Hervé
Dombret,4,5 Claude Preudhomme1,2,3 and Aline Renneville1,2,3
1
Hematology Laboratory, Biology and Pathology Center, CHRU of Lille; 2INSERM, UMR-S
1172, Cancer Research Institute of Lille; 3University of Lille, F-59000; 4Hematology
Haematologica 2018
Department, Saint-Louis Hospital, AP-HP, Paris; 5EA3518, Institut Universitaire
Volume 103(5):822-829
d’Hématologie (IUH), University 7 Paris Diderot; 6Circulating Biomarkers Laboratory, Curie
Institute, Paris; 7Acute Leukemia French Association (ALFA) coordination, Saint-Louis
Hospital, AP-HP, Paris; 8Hematology Department, Versailles Hospital, Le Chesnay;
9
Hematology Department, Lyon Sud Hospital, Pierre Benite and 10Cytogenetic Laboratory,
Versailles Hospital, Le Chesnay, France

ABSTRACT

A
ssessment of minimal residual disease has emerged as a powerful
prognostic factor in acute myeloid leukemia. In this study, we
investigated the potential of IDH1/2 mutations as targets for min-
imal residual disease assessment in acute myeloid leukemia, since these
mutations collectively occur in 15-20% of cases of acute myeloid
leukemia and now represent druggable targets. We employed droplet
digital polymerase chain reaction assays to quantify IDH1R132,
IDH2R140, and IDH2R172 mutations on genomic DNA in 322 samples
from 103 adult patients with primary IDH1/2 mutant acute myeloid
leukemia and enrolled on Acute Leukemia French Association (ALFA) -
Correspondence: 0701 or -0702 clinical trials. The median IDH1/2 mutant allele fraction in
aline.renneville@gmail.com bone marrow samples was 42.3% (range, 8.2 - 49.9%) at diagnosis of
acute myeloid leukemia, and below the detection limit of 0.2% (range,
<0.2 - 39.3%) in complete remission after induction therapy. In univariate
Received: October 29, 2017. analysis, the presence of a normal karyotype, a NPM1 mutation, and an
Accepted: February 16, 2018. IDH1/2 mutant allele fraction <0.2% in bone marrow after induction
Pre-published: February 22, 2018.
therapy were statistically significant predictors of longer disease-free sur-
vival. In multivariate analysis, these three variables remained significantly
predictive of disease-free survival. In 7/103 (7%) patients, IDH1/2 muta-
doi:10.3324/haematol.2017.183525 tions persisted at high levels in complete remission, consistent with the
presence of an IDH1/2 mutation in pre-leukemic hematopoietic stem
Check the online version for the most updated cells. Five out of these seven patients subsequently relapsed or progressed
information on this article, online supplements, toward myelodysplastic syndrome, suggesting that patients carrying the
and information on authorship & disclosures:
www.haematologica.org/content/103/5/822 IDH1/2 mutation in a pre-leukemic clone may be at high risk of hemato-
logic evolution.

©2018 Ferrata Storti Foundation


Material published in Haematologica is covered by copyright.
All rights are reserved to the Ferrata Storti Foundation. Use of Introduction
published material is allowed under the following terms and
conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode. Assessment of minimal residual disease (MRD) has emerged as a powerful prog-
Copies of published material are allowed for personal or inter- nostic factor in acute myeloid leukemia (AML).1–6 Many studies have shown that
nal use. Sharing published material for non-commercial pur- MRD detection using multiparameter flow cytometry or real-time quantitative
poses is subject to the following conditions:
polymerase chain reaction (qPCR) provides powerful independent prognostic infor-
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- mation in AML.7,8 Chimeric fusion genes, such as PML-RARA, CBFB-MYH11 or
mercial purposes is not allowed without permission in writing RUNX1-RUNX1T1, NPM1 mutations, as well as WT1 expression are well-estab-
from the publisher. lished molecular markers for MRD monitoring in AML. However, sensitive and
leukemia-specific MRD markers are lacking in approximately 40% of AML
patients. This prompted us to investigate the potential of other recurrent molecular

822 haematologica | 2018; 103(5)


IDH1/2 mutant allele burden in AML

abnormalities as targets for MRD assessment in AML, such 36; ClinicalTrials.gov NCT00927498 or ALFA-0702 (Eudra-CT
as mutations in isocitrate dehydrogenase (IDH) 1 and 2. 2008-000668-18; ClinicalTrials.gov NCT00932412) trials.
IDH1/2 mutations affecting IDH1R132, IDH2R140, and Treatment schemes have been previously reported for both tri-
IDH2R172 residues are single-nucleotide mutations that als.13,14 These studies were approved by the ethics committee of
collectively occur in 15-20% of AML and represent driver Saint-Germain en Laye and Sud Est IV, France, respectively, and
mutations in leukemogenesis.9 Mutant IDH1/2 enzymes the institutional review board of the French Regulatory Agency.
have neomorphic activity and catalyze the reduction of a- Bone marrow or peripheral blood samples collected at the time of
ketoglutarate to an oncometabolite, the R-enantiomer of diagnosis of AML and during follow-up were obtained from the
2-hydroxyglutarate (2-HG), which promotes DNA and tissue bank Tumorothèque du Centre de Référence Régional en
histone hypermethylation, altered gene expression, and Cancérologie de Lille (CRRC)” and approval for this study was
impaired hematopoietic differentiation.10,11 Quantification obtained from the institutional review board of CHRU of Lille
of single-nucleotide mutations by qPCR can be challeng- (CSTMT089). All patients provided written informed consent to
ing because of problems with background amplification both treatment and genetic analysis before inclusion in the study,
from the wild-type allele. Recently, the development of in accordance with the declaration of Helsinki. Among all patients
digital PCR has enabled absolute quantification of various included in the ALFA-0701 (n=278) or ALFA-0702 (n=704) trials,
genomic targets with high precision and sensitivity and we selected patients meeting the following criteria: (i) the pres-
has, therefore, turned out to be a promising technique for ence of an IDH1R132 or an IDH2R140/R172 mutation at AML
MRD monitoring, especially for gene mutations.7,12 diagnosis (n=160), (ii) achievement of complete remission after
The clinical significance of residual IDH1/2 mutations in induction therapy (n=130), and (iii) one or more bone marrow fol-
bone marrow in complete remission after chemotherapy low-up sample available for IDH1/2 variant allele fraction
is currently unknown. In this study, we employed digital (IDH1/2-VAF) assessment (n=103) (Figure 1).
PCR assays to quantify IDH1/2 mutant allele fraction at
AML diagnosis and during follow-up in a large cohort of Molecular analysis
AML patients intensively treated in the Acute French Droplet DigitalTM PCR (ddPCR) assays were used to quantify
Leukemia Association (ALFA) trials to investigate whether the IDH1/2 mutant allele and its wild-type counterpart in diagnos-
IDH1/2 mutations are suitable MRD markers that could tic and follow-up samples. During complete remission, only bone
predict clinical outcome in AML patients and provide fur- marrow samples were analyzed for IDH1/2-VAF assessment.
ther information for risk-adapted therapy. IDH1/2-VAF was quantified on genomic DNA using Bio-RadTM
reagents, primers and probes (HEX-labeled wild-type allele; FAM-
labeled mutant alleles). All samples were tested in duplicate wells,
Methods using 90 ng of DNA per well. The PCR product from each well
was then subjected to the QX100 droplet reader (Bio-RadTM),
Patients and treatment which measures the fluorescence of each droplet individually
This study was performed in 103 adult patients (18-70 years) using a two-color detection system. Raw data were analyzed
with previously untreated primary IDH1/2 mutated AML and using QuantaSoft software, version 1.7.4.0917 (Bio-RadTM).
enrolled on the prospective ALFA-0701 (Eudra-CT 2007-002933- Representative two-dimensional plots of droplet fluorescence for

Figure 1. Patient flow chart. VAF,


variant allele fraction.

haematologica | 2018; 103(5) 823


Y. Ferret et al.

IDH1/2 wild-type controls and IDH1/2 mutant samples are shown mutations in AML.9,17 In our cohort, IDH2R172K muta-
in Online Supplementary Figure S1. The mutant allele frequency was tions were mutually exclusive with NPM1 and FLT3 muta-
then estimated using a Poisson distribution model as the fraction tions, but co-occurred with DNMT3A mutations. An iso-
of positive droplets divided by total droplets containing a target. lated trisomy 11 was identified in 5/21 (24%) patients
The limit of detection was defined for each mutation as the mean with the IDH2R172K mutation, while this cytogenetic
value of IDH1/2 wild-type controls plus three standard deviations abnormality was not found in any patient with other
(Online Supplementary Table S1). The upper detection limit of these types of IDH1/2 mutations (24% versus 0%; P<0.001)
ddPCR assays (rounded to 0.2% of mutant allele frequency) was (Figure 2). In the subgroup of IDH2R172K mutant AML
further considered as the threshold for statistical analysis. An (n=21), single-nucleotide polymorphism array analysis
IDH1/2-VAF level below 0.2% was hereafter considered as nega- revealed an additional genomic lesion involving chromo-
tive MRD. Gene mutation analysis and next-generation sequenc- some 11, consisting of a 11p11.2-q12.1 uniparental dis-
ing assays are described in the Online Supplementary Methods and omy, in one patient with normal karyotype AML. No MLL
Online Supplementary Tables S2-S4. partial tandem duplication, known to be strongly associat-
ed with trisomy 11,18 was found by reverse transcriptase
Statistical analysis
Group comparison for categorical and continuous variables was
performed with the Fisher exact and Mann-Whitney test, respec-
Table 1. Baseline characteristics of the patients and acute myeloid
tively. Overall survival was calculated from the date of AML diag- leukemias.
nosis to the last follow-up date by censoring patients alive at that
date. Disease-free survival was calculated from the date of com- Number of patients (%)
plete remission to the date of relapse or death, censoring patients
ALFA-0701 ALFA-0702 Total
alive without an event at the last follow-up date. In some analyses, Gender
data were censored at the time of allogeneic stem cell transplanta- Male 10 38 48 (47)
tion. Univariate and multivariate analyses assessing the impact of Female 16 39 55 (53)
categorical and continuous variables were performed with a Cox Median age (range), years 62 (51-70) 50 (22-60) 54 (22-70)
model.15 The proportional-hazards assumption was checked before Median white blood cell 18 (1-157) 5 (1-377) 7 (1-377)
conducting multivariate analyses.16 Covariates with a P-value <0.1 count (range), x 109/L
in univariate analysis were included in the multivariable models.
Cytogenetics
Statistical analyses were performed with STATA software (STATA
Normal 21 50 71 (69)
12.0 Corporation, College Station, TX, USA). P-values were two-
Abnormal 4 23 27 (26)
sided, with P<0.05 denoting statistical significance. Failure 1 4 5 (5)
IDH1/2 mutation
IDH1 p.R132H/C/G 10 26 36 (35)
Results IDH2 p.R140Q 10 36 46 (45)
IDH2 p.R172K 6 15 21 (20)
Baseline characteristics of the patients and acute
myeloid leukemias Other gene mutations
The patients’ median age was 54 years (range, 22-70). NPM1 mutation 16 34 50 (48)
FLT3 internal tandem duplication 3 16 19 (19)
The median follow-up was 2.7 years (95% CI: 2.3-3.0).
FLT3-tyrosine kinase domain mutation2 7 9 (9)
Results of conventional cytogenetic studies were available DNMT3A mutation 6 23 29 (35)
for 98/103 (95%) patients, of whom 72% had normal TET2 mutation 2 2 4 (4)
karyotype AML. A concomitant NPM1 mutation was CEBPA mutation 1 (1 sm) 3 (2 sm, 1 dm) 4 (4)
found in 50/103 (48%) patients. Only 4/103 (4%) patients
European LeukemiaNet 2008 risk-group
harbored a concomitant TET2 mutation (Table 1), in accor-
Favorable 13 20 33 (32)
dance with the fact that IDH1/2 and TET2 mutations tend Non-favorable 12 52 64 (62)
to be mutually exclusive.10 As opposed to IDH1R132 and Not defined 1 5 6 (6)
IDH2R140 mutations, IDH2R172 mutations are less likely
sm: single mutation; dm: double mutation.
to be accompanied by additional frequently recurring

Figure 2. Barcoding representing the co-occurrence of gene mutations and cytogenetic alterations in our cohort of 103 patients with IDH1/2 mutant acute myeloid
leukemia. ITD: internal tandem duplication; TKD: tyrosine kinase domain.

824 haematologica | 2018; 103(5)


IDH1/2 mutant allele burden in AML

PCR in this subgroup (data not shown). The association with an IDH2R140 mutation and three with an IDH1R132
between IDH2R172 mutation and trisomy 11 observed in mutation, but none with an IDH2R172 mutation. The
our cohort is consistent with results from a previous main characteristics of these seven patients are summa-
study,19 and suggests a potential cooperation between rized in Table 2 and their IDH1/2-VAF profiles are shown
these two genetic alterations in leukemogenesis. in Figure 3B. The only common characteristic identified in
these patients was age over 50 years. In this subgroup, the
IDH1/2 mutation level at diagnosis of acute myeloid median IDH1/2-VAF was 8% (range, 0.8-28.5%) after
leukemia and during follow-up induction and 40% (range, 26-43.5%) after consolidation
At AML diagnosis, IDH1/2-VAF could be assessed by therapy. Of these seven patients, only one is still alive in
next-generation sequencing in 80/103 patients (Online first complete remission, one died from transplant-related
Supplementary Table S5). The median IDH1/2-VAF value mortality, three relapsed, and two developed overt
was 41% (range, 16-53%) in bone marrow and 39.5% myelodysplastic syndrome. Altogether, 5/7 (71%) patients
(range, 6-50%) in peripheral blood samples. In the subset with persistent clonal hematopoiesis with IDH1/2 muta-
of NPM1-mutated AML, IDH1/2-VAF was systematically tions relapsed or progressed toward myelodysplastic syn-
higher than NPM1-VAF, except in one patient with similar drome within 1 to 4 years after AML diagnosis.
VAF for both mutations [n=34 comparisons; median dif-
ference IDH1/2-VAF - NPM1-VAF, 10.5% (range, 0-25%); Univariate and multivariate prognostic analyses
P<0.001] (Online Supplementary Figure S2). This finding The prognostic impact of IDH1/2 mutations in AML
supports the notion that IDH1/2 mutations were present remains controversial.9 In the present cohort composed
in pre-existing clones that subsequently acquired NPM1 exclusively of IDH1/2-mutated AML, the presence of an
mutations. IDH2R172 mutation was associated with a shorter dis-
We also performed ddPCR assays in diagnostic and fol- ease-free survival compared to other IDH1/2 mutation
low-up samples to quantify the IDH1/2-VAF. A total of types, but without the difference reaching statistical sig-
322 samples from 103 patients with IDH1/2 mutations nificance (P=0.088). No difference according to the type of
were analyzed by ddPCR at diagnosis (n=97, of which 69 IDH1/2 mutation was observed regarding overall survival
were bone marrow and 28 peripheral blood samples), dur- (Table 3; Figure 4).
ing hematologic remission (n=211 bone marrow samples), The prognostic impact of IDH1/2-VAF was evaluated in
and at relapse (n=14 bone marrow samples). At AML diag- complete remission after induction therapy in a subset of
nosis, the median IDH1/2-VAF assessed by ddPCR was 95 patients for whom a post-induction bone marrow sam-
42.3% (range, 8.2-49.9%) in bone marrow and 40.6% ple was available for IDH1/2-VAF assessment (Figure 1).
(range, 5.5-53%) in peripheral blood samples, consistent We were not able to perform statistical analysis at later
with our next-generation sequencing data. After induction follow-up time-points, such as post-consolidation,
therapy, the IDH1/2 mutant allele fraction in bone marrow because of the lack of available DNA samples for many
samples decreased significantly compared to the pretreat- patients. Variables considered for univariate and multivari-
ment levels (P<0.001) with a median value below 0.2% ate analyses were age, white blood cell count, cytogenet-
(range, <0.2-39.3%). At AML relapse, the median IDH1/2- ics, mutational status of five genes, and IDH1/2-VAF after
VAF was 21.3% (range, 0.2-38.5%). Among the 14 induction therapy. In univariate analysis for disease-free
patients for whom a bone marrow sample was available survival, the presence of a normal karyotype, a NPM1
for molecular analysis at AML relapse, only one lost the mutation, and a IDH1/2-VAF <0.2% were significantly
mutation during disease evolution (Figure 3A). associated with a longer disease-free survival. In multivari-
ate analysis, these three variables remained significantly
Persistent clonal hematopoiesis with IDH1/2 predictive of disease-free survival. Factors significantly
mutations associated with overall survival were age, the presence of
IDH1/2 mutations persisted at high levels during hema- a normal karyotype, the presence of a NPM1 mutation or
tologic remission in 7/103 (7%) patients, including four a TET2 mutation. Other molecular abnormalities studied,

A B

Figure 3. IDH1/2 mutant allele fraction assessed by droplet digital polymerase chain reaction at diagnosis of acute myeloid leukemia and during follow-up (A)
in the whole cohort and (B) for the seven patients with persistent clonal hematopoiesis with IDH1/2 mutations. The plain lines in the dot plot indicate the median
values.

haematologica | 2018; 103(5) 825


Y. Ferret et al.

as well as IDH1/2-VAF, had no impact on overall survival comitant mutations, such as NPM1 or DNMT3A muta-
(Table 3; Figure 5). tions.17,22 The present study, which only included patients
with IDH1/2 mutations, was not designed to explore the
Discussion prognostic significance of IDH1/2 mutations.
The role of MRD in the management of AML patients is
In this study including 103 adult patients with primary growing. Because of the marked heterogeneity of AML,
IDH1/2 mutant AML who were intensively treated, we no single MRD marker can be applied to all patients.
showed the feasibility of IDH1/2-VAF monitoring using Additionally, the optimal method for measuring clearance
ddPCR and its prognostic relevance after induction thera- of leukemia cells after chemotherapy remains to be deter-
py, independently of pretreatment risk factors. Our find- mined. Here, we focused on IDH1/2 mutations because
ings also suggest that patients with persistent IDH1/2- they are recurrent genetic events in AML, mostly in nor-
mutated clonal hematopoiesis may be at high risk of dis- mal karyotype AML, and now represent druggable targets.
mal hematologic evolution. The digital PCR technique had been previously shown to
The prognostic value of IDH1/2 mutations is still a mat- allow absolute quantification of a nucleic acid target with
ter of debate9 and may be influenced by the type of muta- high precision and sensitivity.12 Our data provide evidence
tions, as we previously reported,20,21 or the profile of con- that measurement of IDH1/2-VAF by ddPCR is feasible.

Table 2. Clinical and biological characteristics of the seven patients with persistent clonal hematopoiesis with IDH1/2 mutations.
UPN 1 UPN 2 UPN 3 UPN 4 UPN 5 UPN 6 UPN 7
Age (years) 50 55 55 50 68 60 63
Gender F M M M F F F
9
WBC count, x 10 /L 28 2.4 4.7 43 34 100 3.2
Cytogenetics Normal Trisomy 8 Normal Normal Failure Normal Normal
NPM1 mutation Pos. Neg. Pos. Neg. Pos. Pos. Neg.
FLT3-ITD Pos. Neg. Neg. Pos. Pos. Neg. Neg.
FLT3-TKD mutation Pos. Neg. Neg. Neg. Neg. Neg. Neg.
CEBPA mutation Neg. NA Neg. Neg. Neg. Neg. Neg.
DNMT3A mutation NA p.R882H (VAF 26%) NA p.R882H (VAF 48%) Neg. Neg. Neg.
TET2 mutation NA Neg. NA Neg. Neg. Neg. Neg.
IDH1/2 mutation IDH2 p.R140Q IDH2 p.R140Q IDH2 p.R140Q IDH2 p.R140Q IDH1 p.R132G IDH1 p.R132C IDH1 p.R132C
(VAF at diagnosis) (44%) (43%) (39%) (47%) (44%) (48%) (43%)
IDH1/2-VAF in CR 28.5% 0.76% 4.87% 28.1% NA 8.2% NA
after induction
IDH1/2-VAF in CR 28.1% 7.2% 42.9% 39.9% 43.5% NA 27.1%
after consolidation
Clinical outcome Alive in CR1 2 years Relapse 4 years MDS 1 year after Relapse 1.5 year Death after Relapse 1.5 year MDS 2.5 years
after AML after AML AML diagnosis after AML allo-SCT after AML after AML
diagnosis diagnosis diagnosis diagnosis diagnosis
UPN: unique patient number; F: female; M: male; WBC: white blood cell; Pos.: positive; Neg.: negative; ITD: internal tandem duplication; TKD: tyrosine kinase domain; NA: not available;
VAF: variant allele fraction; CR: complete remission; AML: acute myeloid leukemia; MDS: myelodysplastic syndrome; allo-SCT: allogeneic stem cell transplantation.

Table 3. Prognostic analysis for disease-free survival and overall survival.


Disease-free survival Overall survival
Univariate Multivariate Univariate
Variable HR 95% CI P HR 95% CI P HR 95% CI P
Age* 1.04 0.98 1.10 0.202 - - - - 1.13 1.00 1.27 0.047
Log10 (white blood cell count)* 1.00 0.99 1.01 0.537 - - - - 1.00 0.99 1.01 0.698
NPM1 mutation 0.23 0.11 0.50 <0.001 0.32 0.12 0.88 0.027 0.19 0.05 0.72 0.014
Normal karyotype 0.26 0.12 0.59 0.001 0.41 0.17 0.99 0.046 0.24 0.07 0.76 0.016
FLT3 internal tandem duplication 1.11 0.33 3.70 0.865 - - - - 1.00 0.12 8.08 1.000
FLT3 tyrosine kinase domain mutation 0.20 0.03 1.48 0.115 - - - - - - - 0.078
DNMT3A mutation 1.42 0.61 3.31 0.413 - - - - 2.42 0.64 9.15 0.192
TET2 mutation 2.66 0.58 12.30 0.209 - - - - 12.59 1.68 94.62 0.014
IDH2 p.R172K mutation 2.04 0.90 4.61 0.088 0.85 0.31 2.32 0.751 1.44 0.39 5.34 0.586
IDH1/2-VAF after induction <0.2% 0.32 0.15 0.69 0.004 0.40 0.18 0.90 0.026 0.46 0.14 1.54 0.208
HR: hazard ratio; CI: confidence interval; VAF: variant allele fraction; *: continuous variable.

826 haematologica | 2018; 103(5)


IDH1/2 mutant allele burden in AML

However, despite technical optimizations, we were not practice, IDH1/2-VAF assessment during and after treat-
able to reach the 0.01% or even 0.1% threshold that we ment could be especially valuable in AML patients with-
would expect as the quantitative detection limit with the out recurrent fusion genes or NPM1 mutations, which are
specific ddPCR assays. This problem was due to a relative- both leukemia-specific and more sensitive MRD markers.
ly high background observed in negative controls, which Keeping in mind the caveat that IDH1/2 mutations can be
always consisted of double-positive (actually false-posi- present in the pre-leukemic clone in some cases, one could
tive) droplets. Polymerase errors occurring during the PCR argue that these mutations could be good MRD markers
amplification step seem to be responsible for the genera- for those patients in whom MRD becomes undetectable
tion of these false-positive signals. after induction or at early follow-up time-points.
The present study is the first to quantify IDH1/2 muta- However, sequential monitoring of IDH1/2-VAF after con-
tion levels in a large cohort of AML patients. Previous solidation therapy or allogeneic stem cell transplantation
studies using Sanger sequencing,23 qPCR,24 or next-genera- could still help to detect disease persistence and guide pre-
tion sequencing technology25 suggested that the presence emptive therapy to prevent hematologic relapse, as sug-
or the level of IDH1/2 mutations was correlated to disease gested in a recent study.26 An alternative approach to MRD
status in most patients with AML, but the small number monitoring in IDH1/2-mutated patients is to quantify the
of IDH1/2-mutated patients included in these studies pre- oncometabolite 2-HG.27,28 A previous study from the ALFA
cluded statistical analysis. Our study revealed that a posi- group showed that total 2-HG serum levels <2 μmol/L
tive IDH1/2-VAF after induction chemotherapy was asso- after induction were associated with better disease-free
ciated with a shorter disease-free survival. Whether survival and overall survival.29 We were not able to corre-
patients with residual IDH1/2 mutations in complete late IDH1/2-VAF and 2-HG levels in this study because of
remission may benefit from allogeneic stem cell transplan- the lack of serum samples.
tation remains to be addressed by future studies. In clinical We found that 7/103 (7%) patients had an IDH1/2

A B

Figure 4. Kaplan-Meier estimates of (A) disease-free survival and (B) overall survival according to the type of IDH1/2 mutation.

A B

Figure 5. Prognostic analysis according to post-induction IDH1/2 mutant allele fraction. Kaplan-Meier estimates of (A) disease-free survival and (B) overall survival
according to IDH1/2 variant allele fraction (IDH1/2-VAF). MRD+ denotes IDH1/2-VAF ≥0.2% and MRD- denotes IDH1/2-VAF <0.2%.

haematologica | 2018; 103(5) 827


Y. Ferret et al.

mutation that persisted at high levels in hematologic molecules might also be considered in patients with per-
remission, consistent with the presence of this mutation sistence of clonal hematopoiesis with IDH1/2 mutations,
in pre-leukemic hematopoietic stem cells. Unlike AML although clearance of the clone carrying the drug targets
blasts, these hematopoietic stem cells survive chemother- seems to occur only in a small subset of treated patients,
apy and persist in remission bone marrow, providing a even with the most potent inhibitors.38 Additionally, pre-
potential reservoir for leukemic progression.30 In our study, clinical and clinical data indicate that IDH1/2 mutations
5/7 (71%) patients with persistent clonal hematopoiesis may identify patients likely to respond to pharmacological
with IDH1/2 mutations relapsed or progressed toward BCL-2 inhibition.39,40 The use of IDH1/2-VAF monitoring in
myelodysplastic syndrome, suggesting that these patients patients treated with an IDH1/2 or BCL-2 inhibitor, such as
may be at high risk of hematologic evolution and should venetoclax, could therefore contribute to the evaluation of
probably be monitored more closely. Klco et al. showed treatment efficacy.
that initiating mutations, such as DNMT3A, TET2, and In conclusion, our study is the first to show that IDH1/2
IDH1/2 mutations, are less likely to be cleared after mutant allele fraction in complete remission after induc-
chemotherapy than cooperating mutations,31 in accor- tion therapy significantly correlates with disease-free sur-
dance with our own and previous data.25,32 Furthermore, vival, independently of pretreatment prognostic factors.
the prognostic value of persisting somatic mutations in However, this difference did not translate into distinct
complete remission appears to vary depending on the overall survival rates in our cohort. Our data provide evi-
gene involved. Recent studies suggested that the presence dence that IDH1/2 mutant allele fraction has the potential
of persistent mutations in DNMT3A, TET2 or ASXL1 lacks to become a useful tool for the management of AML
prognostic impact in terms of AML relapse or survival,33,34 patients as a biomarker of treatment response, in addition
in contrast with what we observed for IDH1/2 mutations. to being a molecular predictor of response to targeted ther-
Patients with IDH1/2 mutations are candidates for tar- apies. Further studies based on larger cohorts of patients
geted therapies. Small-molecule inhibitors of mutant IDH1 are required to confirm and extend our findings, and to
such as ivosidenib or IDH2 such as the recently approved address the question of whether the residual level of
enasidenib are currently under clinical investigation and, IDH1/2 mutation may help to refine the assignment into
when used as single agents, have shown promising results distinct risk groups and guide the decision of whether to
in patients with AML or myelodysplastic syndrome as a perform allogeneic stem cell transplantation or give target-
first-line treatment or in relapsed or refractory diseases.9 ed therapies.
These molecules have been shown to induce differentia-
tion of primary leukemic cells in vitro35,36 and in vivo37 to pro- Acknowledgments
mote clinical responses. Future studies should determine This work was supported by the Association Laurette Fugain,
whether patients with high levels of IDH1/2-VAF after the Ligue Contre le Cancer (North Center), the SIRIC
induction therapy could benefit from a consolidation or ONCOLille, the North-West Canceropole (GIRCI AAP-AE
maintenance therapy including IDH1/2 inhibitors. 2015_53), and the Institut National du Cancer - Direction
Ultimately, one could imagine that the use of these small Generale de l’Offre de Soin (INCA-DGOS_9967).

References HOVON/SAKK AML 42A study. J Clin number. Anal Chem. 2011;83(22):8604–
Oncol. 2013;31(31):3889–3897. 8610.
1. Schnittger S, Weisser M, Schoch C, 6. Krönke J, Schlenk RF, Jensen K-O, et al. 13. Castaigne S, Pautas C, Terré C, et al. Effect
Hiddemann W, Haferlach T, Kern W. New Monitoring of minimal residual disease in of gemtuzumab ozogamicin on survival of
score predicting for prognosis in PML- NPM1-mutated acute myeloid leukemia: a adult patients with de-novo acute myeloid
RARA+, AML1-ETO+, or CBFBMYH11+ study from the German-Austrian acute leukaemia (ALFA-0701): a randomised,
acute myeloid leukemia based on quantifi- myeloid leukemia study group. J Clin open-label, phase 3 study. Lancet.
cation of fusion transcripts. Blood. Oncol. 2011;29(19):2709–2716. 2012;379(9825):1508–1516.
2003;102(8):2746–2755. 7. Grimwade D, Ivey A, Huntly BJP. 14. Thomas X, de Botton S, Chevret S, et al.
2. Stentoft J, Hokland P, Ostergaard M, Hasle Molecular landscape of acute myeloid Randomized phase II study of clofarabine-
H, Nyvold CG. Minimal residual core bind- leukemia in younger adults and its clinical based consolidation for younger adults
ing factor AMLs by real time quantitative relevance. Blood. 2016;127(1):29–41. with acute myeloid leukemia in first remis-
PCR--initial response to chemotherapy pre- 8. Ommen HB. Monitoring minimal residual sion. J Clin Oncol. 2017;35(11):1223-1230.
dicts event free survival and close monitor- disease in acute myeloid leukaemia: a 15. Cox D. Regression models and life tables. J
ing of peripheral blood unravels the kinet- review of the current evolving strategies. R Stat Soc B. 1972;34:187-220.
ics of relapse. Leuk Res. 2006;30(4):389– Ther Adv Hematol. 2016;7(1):3–16. 16. Grambsch PM, Therneau TM. Proportional
395. 9. Medeiros BC, Fathi AT, DiNardo CD, hazards tests and diagnostics based on
3. Cilloni D, Renneville A, Hermitte F, et al. Pollyea DA, Chan SM, Swords R. Isocitrate weighted residuals. Biometrika. 1994;81(3):
Real-time quantitative polymerase chain dehydrogenase mutations in myeloid malig- 515–526.
reaction detection of minimal residual dis- nancies. Leukemia. 2017;31(2):272–281. 17. Papaemmanuil E, Gerstung M, Bullinger L,
ease by standardized WT1 assay to 10. Figueroa ME, Abdel-Wahab O, Lu C, et al. et al. Genomic classification and prognosis
enhance risk stratification in acute myeloid Leukemic IDH1 and IDH2 mutations result in acute myeloid leukemia. N Engl J Med.
leukemia: a European LeukemiaNet study. J in a hypermethylation phenotype, disrupt 2016;374(23):2209–2221.
Clin Oncol. 2009;27(31):5195–5201. TET2 function, and impair hematopoietic 18. Caligiuri MA, Strout MP, Schichman SA, et
4. Freeman SD, Virgo P, Couzens S, et al. differentiation. Cancer Cell. 2010;18(6): al. Partial tandem duplication of ALL1 as a
Prognostic relevance of treatment response 553–567. recurrent molecular defect in acute myeloid
measured by flow cytometric residual dis- 11. Losman J-A, Looper RE, Koivunen P, et al. leukemia with trisomy 11. Cancer Res.
ease detection in older patients with acute (R)-2-hydroxyglutarate is sufficient to pro- 1996;56(6):1418–1425.
myeloid leukemia. J Clin Oncol. mote leukemogenesis and its effects are 19. Eisfeld A-K, Kohlschmidt J, Mrózek K, et al.
2013;31(32):4123–4131. reversible. Science. 2013;339(6127):1621– Adult acute myeloid leukemia with tri-
5. Terwijn M, van Putten WLJ, Kelder A, et al. 1625. somy 11 as the sole abnormality is charac-
High prognostic impact of flow cytometric 12. Hindson BJ, Ness KD, Masquelier DA, et al. terized by the presence of five distinct gene
minimal residual disease detection in acute High-throughput droplet digital PCR sys- mutations: MLL-PTD, DNMT3A, U2AF1,
myeloid leukemia: data from the tem for absolute quantitation of DNA copy FLT3-ITD and IDH2. Leukemia.

828 haematologica | 2018; 103(5)


IDH1/2 mutant allele burden in AML

2016;30(11):2254–2258. improve early detection of acute myeloid DNMT3A mutant transcript levels persist
20. Boissel N, Nibourel O, Renneville A, et al. leukemia relapse after allogeneic in remission and do not predict outcome in
Prognostic impact of isocitrate dehydroge- hematopoietic stem cell transplantation. patients with acute myeloid leukemia.
nase enzyme isoforms 1 and 2 mutations in Haematologica. 2016;101(4):e157-161. Leukemia. 2018;32(1):30-37.
acute myeloid leukemia: a study by the 27. Ward PS, Patel J, Wise DR, et al. The com- 34. Jongen-Lavrencic M, Grob T, Kavelaars FG,
Acute Leukemia French Association group. mon feature of leukemia-associated IDH1 et al. Prospective molecular MRD detection
J Clin Oncol. 2010;28(23):3717–3723. and IDH2 mutations is a neomorphic by NGS: a powerful independent predictor
21. Boissel N, Nibourel O, Renneville A, enzyme activity converting a-ketoglutarate for relapse and survival in adults with
Huchette P, Dombret H, Preudhomme C. to 2-hydroxyglutarate. Cancer Cell. newly diagnosed AML. Blood. 2017;130
Differential prognosis impact of IDH2 2010;17(3):225–234. (Suppl 1):LBA-5.
mutations in cytogenetically normal acute 28. Pollyea DA, Kohrt HE, Zhang B, et al. 2- 35. Chaturvedi A, Araujo Cruz MM, Jyotsana
myeloid leukemia. Blood. 2011;117(13): hydroxyglutarate in IDH mutant acute N, et al. Mutant IDH1 promotes leukemo-
3696–3697. myeloid leukemia: predicting patient genesis in vivo and can be specifically tar-
22. Patel JP, Gönen M, Figueroa ME, et al. responses, minimal residual disease and geted in human AML. Blood. 2013;122(16):
Prognostic relevance of integrated genetic correlations with methylcytosine and 2877–2887.
profiling in acute myeloid leukemia. N Engl hydroxymethylcytosine levels. Leuk 36. Wang F, Travins J, DeLaBarre B, et al.
J Med. 2012;366(12):1079–1089. Lymphoma. 2013;54(2):408–410. Targeted inhibition of mutant IDH2 in
23. Chou W-C, Peng K-Y, Lei W-C, et al. 29. Janin M, Mylonas E, Saada V, et al. Serum leukemia cells induces cellular differentia-
Persistence of mutant isocitrate dehydroge- 2-hydroxyglutarate production in IDH1- tion. Science. 2013;340(6132):622–626.
nase in patients with acute myeloid and IDH2-mutated de novo acute myeloid 37. Amatangelo MD, Quek L, Shih A, et al.
leukemia in remission. Leukemia. leukemia: a study by the Acute Leukemia Enasidenib induces acute myeloid leukemia
2012;26(3):527–529. French Association group. J Clin Oncol. cell differentiation to promote clinical
24. Jeziskova I, Razga F, Toskova M, et al. 2014;32(4):297–305. response. Blood. 2017;130(6):732–741.
Quantitative detection of IDH2 mutation for 30. Shlush LI, Zandi S, Mitchell A, et al. 38. Perl AE. The role of targeted therapy in the
minimal residual disease monitoring in Identification of pre-leukaemic management of patients with AML.
patients with acute myeloid leukemia and haematopoietic stem cells in acute Hematol Am Soc Hematol Educ Program.
its comparison with mutations in NPM1 leukaemia. Nature. 2014;506(7488):328– 2017;2017(1):54–65.
gene. Leuk Lymphoma. 2013;54(4):867–870. 333. 39. Chan SM, Thomas D, Corces-Zimmerman
25. Debarri H, Lebon D, Roumier C, et al. 31. Klco JM, Miller CA, Griffith M, et al. MR, et al. Isocitrate dehydrogenase 1 and 2
IDH1/2 but not DNMT3A mutations are Association between mutation clearance mutations induce BCL-2 dependence in
suitable targets for minimal residual disease after induction therapy and outcomes in acute myeloid leukemia. Nat Med.
monitoring in acute myeloid leukemia acute myeloid leukemia. JAMA. 2015;21(2):178–184.
patients: a study by the Acute Leukemia 2015;314(8):811–822. 40. Konopleva M, Pollyea DA, Potluri J, et al.
French Association. Oncotarget. 2015;6 32. Pløen GG, Nederby L, Guldberg P, et al. Efficacy and biological correlates of
(39):42345–42353. Persistence of DNMT3A mutations at long- response in a phase II study of venetoclax
26. Brambati C, Galbiati S, Xue E, et al. Droplet term remission in adult patients with AML. monotherapy in patients with acute myel-
digital polymerase chain reaction for Br J Haematol. 2014;167(4):478–486. ogenous leukemia. Cancer Discov. 2016;6
DNMT3A and IDH1/2 mutations to 33. Gaidzik VI, Weber D, Paschka P, et al. (10):1106–1117.

haematologica | 2018; 103(5) 829


ARTICLE Acute Lymphoblastic Leukemia

MSH6 haploinsufficiency at relapse


Ferrata Storti
Foundation contributes to the development of thiopurine
resistance in pediatric B-lymphoblastic
leukemia
Nikki A. Evensen,1 P. Pallavi Madhusoodhan,1 Julia Meyer,2 Jason Saliba,1
Ashfiyah Chowdhury,1 David J. Araten,3 Jacob Nersting,4 Teena Bhatla,1
Tiffaney L. Vincent,5 David Teachey,5 Stephen P. Hunger,5 Jun Yang,6
Haematologica 2018 Kjeld Schmiegelow4 and William L. Carroll1

Departments of Pediatrics and Pathology, Perlmutter Cancer Center, NYU-Langone


Volume 103(5):830-839
1

Medical Center, New York, NY, USA; 2Huntsman Cancer Institute, University of Utah
Medical Center, Salt Lake City, USA; 3Department of Medicine, Perlmutter Cancer
Center, NYU-Langone Medical Center, New York NY, USA; 4Department of Pediatrics and
Adolescent Medicine, The University Hospital Rigshospitalet, Copenhagen, Denmark;
5
Department of Pediatrics and the Center for Childhood Cancer Research, Children’s
Hospital of Philadelphia and The Perelman School of Medicine at The University of
Pennsylvania, Philadelphia, PA, USA and 6St. Jude Children’s Research Hospital,
Memphis, TN, USA
ABSTRACT

S
urvival of children with relapsed acute lymphoblastic leukemia is
poor, and understanding mechanisms underlying resistance is
essential to developing new therapy. Relapse-specific heterozygous
deletions in MSH6, a crucial part of DNA mismatch repair, are frequent-
ly detected. Our aim was to determine whether MSH6 deletion results
in a hypermutator phenotype associated with generation of secondary
mutations involved in drug resistance, or if it leads to a failure to initiate
apoptosis directly in response to chemotherapeutic agents. We knocked
down MSH6 in mismatch repair proficient cell lines (697 and UOCB1)
Correspondence: and showed significant increases in IC50s to 6-thioguanine and 6-mer-
captopurine (697: 26- and 9-fold; UOCB1: 5- and 8-fold) in vitro, as well
william.carroll@nyumc.org
as increased resistance to 6-mercaptopurine treatment in vivo. No shift in
IC50 was observed in deficient cells (Reh and RS4;11). 697 MSH6 knock-
down resulted in increased DNA thioguanine nucleotide levels com-
Received: July 13, 2017. pared to non-targeted cells (3070 vs. 1722 fmol/μg DNA) with no differ-
Accepted: February 7, 2018. ence observed in mismatch repair deficient cells. Loss of MSH6 did not
give rise to microsatellite instability in cell lines or clinical samples, nor
Pre-published: February 15, 2018.
did it significantly increase mutation rate, but rather resulted in a defect
in cell cycle arrest upon thiopurine exposure. MSH6 knockdown cells
doi:10.3324/haematol.2017.176362 showed minimal activation of checkpoint regulator CHK1, γH2AX
(DNA damage marker) and p53 levels upon treatment with thiopurines,
Check the online version for the most updated consistent with intrinsic chemoresistance due to failure to recognize
information on this article, online supplements, thioguanine nucleotide mismatching and initiate mismatch repair.
and information on authorship & disclosures: Aberrant MSH6 adds to the list of alterations/mutations associated with
www.haematologica.org/content/103/5/830 acquired resistance to purine analogs emphasizing the importance of
thiopurine therapy.
©2018 Ferrata Storti Foundation
Material published in Haematologica is covered by copyright.
All rights are reserved to the Ferrata Storti Foundation. Use of Introduction
published material is allowed under the following terms and
conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode. Relapsed B-precursor acute lymphoblastic leukemia (B-ALL) is a leading cause of
Copies of published material are allowed for personal or inter- cancer mortality amongst children. Development of chemoresistance is a crucial
nal use. Sharing published material for non-commercial pur- factor contributing to relapse, therefore understanding the biological mechanisms
poses is subject to the following conditions:
underlying this resistance is imperative for discovering innovative treatment strate-
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- gies.1 Recent work has begun to highlight the direct role of relapse
mercial purposes is not allowed without permission in writing specific/enriched genetic alterations in the emergence of clones that have gained a
from the publisher. selective advantage under the pressure of specific chemotherapeutics, such as
NT5C2, TBL1XR1, PRPS1, and CREBBP.1-5 Many of these mutations cause resist-
ance specifically to thiopurines, which are the backbone of maintenance therapy

830 haematologica | 2018; 103(5)


MSH6 haploinsufficiency contributes to resistance

and have proven vital for achieving cures.6 Our analysis of RPMI1640 medium. HEK293T (ATCC) cells were grown in
copy number alterations (CNAs) in diagnosis/relapse pairs DMEM medium. All media were supplemented with 10% FBS,
revealed a relapse specific hemizygous deletion on chro- 1% penicillin/streptomycin under 5% CO2 at 37°C.
mosome 2p16.3 involving MSH6 in 4-10% of patients.7,8
MutS homolog 6 (MSH6) is a major component of the Drug preparation, viral preparation, immunoblotting,
mismatch repair (MMR) system, which is a highly con- apoptosis assays, and cell cycle
served biological process that recognizes and repairs Standard protocols were followed and have been previously
errors in nascent DNA strands during replication to main- described.3,21 More detailed information is provided in the Online
tain genomic integrity. Initial recognition of replicative Supplementary Appendix.
errors is carried out by protein heterodimers consisting of
either MSH6 and MSH2 (hMutSa), or MSH3 and MSH2 Patients’ samples
(hMutSβ). Upon recognition of a mismatch, hMutSa Cryopreserved pediatric B-ALL specimens were obtained from
recruits MutLa (MLH1-PMS2) which engages down- the Children's Oncology Group (COG) ALL cell bank. All patients
stream proteins and enzymes involved in DNA repair.9,10 were treated on COG protocols for newly diagnosed ALL. All sub-
Constitutional defects in MMR, including monoallelic jects provided consent for banking and future research use of these
mutations in Lynch syndrome and biallelic loss in consti- specimens in accordance with the regulations of the institutional
tutional mismatch repair deficiency (CMMRD), are review boards of all participating institutions.
strongly linked to carcinogenesis, where loss of MMR
functionality causes increased mutability and predisposi- Microsatellite instability analysis
tion to malignancy.11-15 Previous work has linked defects Microsatellite instability (MSI) analysis was performed using
in MMR to drug resistance, including thiopurines, in var- MSI Analysis System, v.1.2 (Promega, Madison, WI, USA) follow-
ious cancers.16-19 However, it is uncertain if resistance in ing the manufacturer’s protocol. Detailed information is provided
MMR defective clones occurs through the acquisition of in the Online Supplementary Appendix.
secondary mutations as a consequence of mutagenic ther-
apy, or the outgrowth of clones that have intrinsic drug Measurement of mutation rate
resistance. Our lab previously demonstrated that lower Spontaneous mutation rate was measured using a flow cytom-
expression of MSH6 in patient samples was associated etry assay previously described by Araten et al.22 that detects the
with increased ex vivo resistance to 6-mercaptopurine and presence of numerous glycosylphosphatidylinositol-linked (GPI)
prednisone,7 highlighting the clinical importance of under- membrane proteins (see Online Supplementary Appendix). Briefly,
standing the role of this genetic alteration in B-ALL. GPI(+) isolated clones from the NT and MSH6-KD cell lines were
The mechanism of action of thiopurines is based upon expanded either untreated or treated with 6-TG (0.040 μg/mL and
the insertion of a false nucleotide, namely a thioguanine 0.100 μg/mL, respectively, based on IC50 values determined for
(TGN), into DNA that when thiomethylated pairs with a clones). Cells were then stained for GPI-dependent markers
thymine instead of a cytosine.18 Cytotoxicity is thought to including FLAER-Alexa 488 (Pinewood), CD48, CD52, and CD59
be dependent on the MMR machinery recognizing the (Serotec),23 and analyzed by flow cytometry. The mutant frequen-
mismatch and attempting to match the TGN on the cy (f) was calculated as the number of GPI(-) events divided by the
parental strand with an appropriate base on the daughter total number of live events, and mutation rate (μ) was calculated
strand.19,20 Whether the DNA damage induced by the as f divided by cell divisions.22
repetitive, futile cycles of DNA excision and repair, or
simply the recognition of mismatches by hMutSa is Thioguanine quantification assay
enough to initiate a signaling cascade culminating in cell Cells were treated with 6-thioguanine (6TG) and collected every
cycle arrest and apoptosis is not entirely understood. day for four days. DNA was extracted using Puregene Core Kit A
We sought to delineate whether reduced expression of (QIAGEN). DNA TGN levels were measured using liquid chro-
MSH6 could give rise to chemoresistance in B-precursor matography-tandem mass spectrometry as described previously.24
ALL and elucidate the mechanism responsible for the
resistance. Our data here support the view that reduced In vivo mouse model of chemoresistance
MSH6 directly results in an increased tolerance to incor- All experiments were conducted on protocols approved by
porated TGN and subsequent mismatches through a fail- the Institutional Animal Care and Use Committee and
ure to initiate MMR, thus allowing cells to proliferate and Institutional Review Board of the Children's Hospital of
survive under thiopurine treatment both in vitro and in Philadelphia. Briefly, 1 million UOCB1 NT GFP-CBG or MSH6
vivo. We demonstrate that ALL cell lines with a functional shRNA1 GFP-CBR cells were injected into NSG mice via tail vein
MMR trigger a CHK1-mediated cell cycle arrest in on day 0 (total 20 mice; 10 per cell line). On day 6 leukemic bur-
response to thiopurines that is followed by DNA damage den was confirmed via bioluminescence imaging (BLI) (IVIS
and apoptosis. In contrast, upon reduction of MSH6, the Spectrum imaging system, Perkin Elmer) and animals were ran-
MMR signaling cascade is not fully activated and cells do domized to treatment groups [PBS vehicle or Purixan (50 mg/kg)
not undergo apoptosis. diluted in PBS]. Mice were treated on day 7 by gavage (0.2
mL/mouse). For BLI, 3 mg of luciferin was injected intraperi-
toneally and mice were imaged ten minutes post injection.
Methods Quantification of total flux was determined by analyzing the BLI
images using Living Image Software (Perkin Elmer) (see Online
Cells and reagents Supplementary Appendix).
The B-lineage leukemia cell lines RS4;11 (ATCC, Manassas, VA,
USA), Reh (ATCC), 697 (DSMZ, Braunschweig, Germany), and Statistical analysis
UOCB1 (a kind gift from Dr. Terzah Horton at Texas Children’s Statistical significance was calculated using unpaired t-test for
Cancer Center/Baylor College of Medicine) were grown in IC50s, paired t-test for mutation rates, one-way ANOVA for cell

haematologica | 2018; 103(5) 831


N.A. Evensen et al.

Figure 1. Knockdown of MSH6 in mismatch repair (MMR) proficient cells lead to decreased sensitivity to thiopurines. (A-C, left) Western blot analysis of whole cell
lysates from 697 (A), UOCB1 (B), and Reh and RS4;11 (C). (A-C, right) Apoptotic cells measured by Annexin V and 7AAD staining followed by flow cytometry after 5
days of treatment. Graphs represent 3 experiments each performed with duplicates. Bars indicate mean+Standard Deviation.

832 haematologica | 2018; 103(5)


MSH6 haploinsufficiency contributes to resistance

cycle analysis, and two-way ANOVA for mutation rates with and S2). However, no significant differences were observed
without treatment as well as in vivo studies. when cells were treated with prednisolone (Pred), doxoru-
bicin (Doxo), cytarabine (Ara-c), or methotrexate (MTX)
(Online Supplementary Figure S3A). Interestingly, we found
Results that knockdown of MSH6 also resulted in decreased sen-
sitivity to temozolomide (TMZ), an alkylating agent used
Previously we noted relapse-specific heterozygous dele- to treat glioblastomas, as reported previously (Online
tions in MSH6 in 4 out of 76 patients that were near iden- Supplementary Figure S3B).27,28
tical and deleted MSH6 only for 3 patients while one har- To further support the role of MSH6 in chemoresis-
bored a larger deletion involving more genes within the tance, we knocked down expression in UOCB1 cells,
region (Online Supplementary Figure S1). To begin to eluci- another B-ALL cell line that expresses all four MMR pro-
date the impact of MSH6 deletion on the development of teins (Figure 1B). Similar to the effect observed in 697 cells,
relapsed disease, we knocked down expression of MSH6 depletion of MSH6 with either shRNA significantly
using shRNA in 697 cells, a B-ALL cell line that expresses reduced the induction of apoptosis upon treatment with
all four MMR proteins (Figure 1A) and is MMR profi- thiopurines (Figure 1B) [fold increase in IC50 as compared
cient,25 and tested for changes in chemosensitivity. We to NT with 6TG: 4.8 for shRNA1 (P=0.007) and 3 for
observed approximately 80-90% (shRNA1) and 50-60% shRNA2 (P<0.001); 6MP: 8.3 for shRNA1 (P<0.001) and
(shRNA2) knockdown of MSH6 expression, as well as 9.2 shRNA2 (P<0.001)] (Online Supplementary Figure S4).
decreased expression of MSH2, compared to non-target- Additionally, a similar impact on TMZ resistance was
ing (NT) control cells (Figure 1A), which is consistent with observed with UOCB1 MSH6 shRNA1 expressing cells
literature on the loss of protein stability of MSH2 and compared to NT control cells, although shRNA2 did not
MSH6 when not dimerized.17,26 Knockdown of MSH6 show the same effect, possibly due to less depletion by
with both shRNA1 and shRNA2 leads to a significant shRNA2 (Online Supplementary Figure S3B).
decrease in apoptotic cells when treated with thiopurines To determine the specificity of the phenotype
for five days (Figure 1A). A 26-fold increase in IC50 with observed for MSH6 depletion versus defects in other
6-TG (NT: 0.027 vs. shRNA1: 0.716 μg/mL; P=0.007) and MMR proteins, we assessed the effect of MSH6 knock-
8.5-fold increase for 6-MP (NT: 0.340 vs. shRNA1: 2.89 down in MMR deficient B-ALL cell lines Reh and
μg/mL; P=0.006) was observed for shRNA1 (Online RS4;11.25,29 Both Reh and RS4;11 have minimal to no
Supplementary Figure S2). A 1.7-fold (NT: 0.015 vs. expression of MLH1 and PMS2 (Figure 1C). Knockdown
shRNA2: 0.025 μg/mL; P=0.015) and a 2.6-fold (NT: 0.143 of MSH6 expression had no effect on the sensitivity of
vs. shRNA2: 0.373 μg/mL; P=0.032) increase in IC50 for 6- either Reh or RS4;11 to 6-TG or 6-MP (Figure 1C and
TG and 6-MP, respectively, were observed for shRNA2 Online Supplementary Figure S5).
cells compared to NT cells (Online Supplementary Figure To begin to elucidate the mechanism of resistance, we

A B

Figure 2. Knockdown of MSH6 leads to increased


tolerance of incorporated thioguanine (TGN). (A and
B) TGN incorporation into DNA was measured over
time after treatment with 0.1 μg/mL of 6-thiogua-
nine (6-TG) in 697 (A) and Reh (B) cells using liquid
chromatography-tandem mass spectrometry. A rep-
resentative graph from 3 independent experiments
is shown. (C) Combined ratio of TGN fmole/μg of DNA
in MSH6 shRNA1 knockdown (KD) cells compared to
non-targeting (NT) cells from 3 experiments. Bars
indicate mean+Standard Deviation.

haematologica | 2018; 103(5) 833


N.A. Evensen et al.

measured the level of TGN incorporation into DNA upon respond to and survive thiopurine exposure. Thus, MMR
treatment with 6-TG. 697 MSH6 shRNA1 cells accumulat- proficient cells with high TGN succumb to the damage
ed more TGN/μg DNA over time than NT cells (NT 1722 and therefore display less TGN/μg DNA over time, mean-
and KD 3070 fmol/μg DNA) (Figure 2A and C). In con- while deficient cells tolerate higher levels of TGN.
trast, no difference in TGN levels was observed in Reh We next tested whether or not a change occurs in cell
cells (Figure 2B and C). Additionally, Reh cells had approx- cycle progression upon treatment. 697 NT cells slowed
imately 10-fold higher TGN levels compared to 697 cells their growth and had a significantly higher proportion of
(Figure 2A and B), highlighting the difference between cells in S phase and less cells in G1 beginning at 96 hours
MMR deficient and proficient cells in their ability to (h) (6-TG, P=0.014; 6-MP, P=0.051) and progressing

Figure 3. Thiopurine treatment resulted in an S phase arrest, which was abrogated upon knockdown of MSH6. 697 (A) and UOCB1 (B) NT and MSH6 shRNA1 and
2 expressing cells were treated with indicated drug for 5 days. Cells were fixed with 70% ethanol, treated with RNAse, and then stained with propidium iodide. DNA
content was analyzed by flow cytometry. Representative images from 3 individual experiments are shown. A one-way ANOVA was performed to determine statistical
significance of the increase in % of cells in S phase at each time point.

834 haematologica | 2018; 103(5)


MSH6 haploinsufficiency contributes to resistance

through 120 h of thiopurine treatment (6-TG, P=0.013; 6- sis marker p53. There was a very modest level of γH2AX
MP, P=0.001) compared to the MSH6 shRNA lines by one- starting at 72 h that increased to a higher level at 96 h after
way ANOVA (Figure 3A and Online Supplementary Figure treatment in 697 NT cells compared to very modest levels
S6A). MSH6 shRNA1 cells had only a modest decrease in in the MSH6 shRNA1-2 cells (Figure 4A), suggesting that
growth with no clear S phase arrest (6-TG, P=0.011, and the functional MMR system in the NT cells was attempt-
6-MP, P=0.001, for percent of cells in S phase compared to ing to repair the DNA, leading to nicks. Additionally, the
NT at 120 h using Tukey’s multiple comparison test), even levels of phosphorylated and total p53 were higher in NT
at higher concentrations of 6-TG (Figure 3A and Online cells at 72 and 96 h compared to MSH6 shRNA1-2 cells
Supplementary Figure S6A and B). MSH6 shRNA2 cells had (Figure 4A) and the shRNA2 cells had higher levels than
a more moderate accumulation of cells in S phase and the shRNA1 cells. Similar results were found in UOCB1
drop of cells in G1 (Figure 3B and Online Supplementary cells (Figure 4B).
Figure S6A), which is consistent with the modest levels of We next examined the impact of MSH6 knockdown on
knockdown and apoptosis. Similar trends were observed mutation rate by performing two assays that measure
with UOCB1 cells (6-TG, P=0.31; and 6-MP, P=0.34 at 120 genomic instability and mutation burden. Microsatellite
h) (Figure 3B). This more moderate effect observed with instability (MSI) is a marker for genomic instability and
the UOCB1 cells is consistent with the degree of impact has been observed in cases where expression of MLH1 or
MSH6 knockdown had on chemoresistance compared to MSH2 is lost.25,33 We investigated MSI on 2 patient sample
the 697 cells. Neither NT nor MSH6 shRNA1 Reh cells pairs that we previously found to have deletions of MSH6
showed alterations in cell cycle upon exposure (Online at relapse, as well as on 697 NT and MSH6 shRNA1 cells
Supplementary Figure S6B). treated with 6-TG for 120 h. No MSI was observed in the
To gain a more complete understanding of the mecha- patient samples comparing diagnosis to relapse or in the
nism leading to apoptosis following TGN incorporation, 697 cells comparing either untreated to 6-TG treated or
we analyzed downstream pathways in 697 NT and MSH6 NT to MSH6 shRNA1 cells (Figure 5A). These data are
shRNA1 cells after treatment with 6-TG. Based on the consistent with previous literature that found alterations
observed S phase arrest and previous research demonstrat- in MSH6 expression alone do not lead to high MSI.34 To
ing activation of the ataxia telangiectasia and Rad3-related investigate the effect of MSH6 disruption on the rate of
(ATR)-Chk1 pathway downstream of MMR,30,31 we first spontaneous mutations in PIG-A, which is required for
assessed the level of activation of Chk1 by probing for expression of GPI, we used a flow cytometry-based assay
phosphorylation of serine 317 (pChk1). 697 NT cells had that measures surface expression of several GPI-depen-
a low level of pChk1 at 48 h with a significant increase dent markers (CD48, CD52, and CD59).22,35 Although
through 96 h of exposure. 697 MSH6 shRNA1 cells had there was a trend to suggest that 697 MSH6 shRNA1 cells
minimal to low levels of pChk1 at 72 h with minimal had a slightly higher mutation rate, statistical significance
increase over time (Figure 4A). The 697 cells expressing was not achieved (Figure 5B). Furthermore, treatment of
shRNA2 had a similar pattern but slightly lower levels of the clones from each cell line with 6-TG did not lead to an
pChk1 compared to NT cells, which is consistent with the increased mutation rate (Figure 5B).
cell cycle data. We next assessed the level of phosphory- To investigate the clinical relevance of reduced MSH6
lated H2AX (γH2AX), a marker of DNA damage that is expression and drug resistance, we utilized an in vivo
phosphorylated downstream of the ATR/ATM pathways mouse model. We injected mice with either UOCB1 NT
following drug treatment,32 as well as levels of the apopto- or UOCB1 MSH6 shRNA1 cell lines (knockdown con-

A B

Figure 4. Thiopurine treatment leads to activation of cell cycle regulator Chk1 and DNA repair that ultimately resulted in DNA damage and cell death. Western
blot analysis of whole cell lysates from 697 (A) and UOCB1 (B) non-targeting (NT), MSH6 shRNA1, and shRNA2 cells after treatment with 6-thioguanine (0.1 μg/mL,
and 0.025 μg/mL, respectively). (C) Untreated cells; numbers are hours after treatment. Blots were probed for Chk1 activation, γH2AX for DNA damage, and apop-
tosis marker p53. Total Chk1, actin, and total H2AX were used as loading controls. Images are representative of 3 individual experiments.

haematologica | 2018; 103(5) 835


N.A. Evensen et al.

firmed day of injection; Figure 6C) and, following confir- specific mutations that confer drug resistance. Some of the
mation of leukemic burden on day 6, the mice were treat- most common relapse specific mutations found thus far
ed with PBS (control) or purixan (an oral suspension form occur in NT5C2 and PRPS1 and lead to the outgrowth of
of 6-MP). Following the 10-day course of purixan treat- thiopurine resistant clones.2,4 Our data presented here
ment, the leukemic burden was significantly diminished demonstrate that reduction of MSH6 in ALL also leads to
in the mice harboring NT cells compared to that observed decreased sensitivity to purine analogs due to a failure to
in the NT PBS treated mice (P=0.0001), suggesting that initiate the apoptotic cascade directly in response to
these cells were unable to survive and expand under the nucleotide mismatches. Even with only 50-60% reduced
selective pressure of the purixan (Figure 6A and B). In con- expression, which potentially mimics levels in patients
trast, the MSH6 shRNA1 mice treated with purixan were with heterozygous loss, we demonstrate a significant
not significantly different from the NT PBS group decrease in sensitivity to thiopurines. Our data are consis-
(P=0.828). Although purixan also had a statistically signif- tent with the recent work of Diouf et al. who showed that
icant impact on MSH6 shRNA1 cells compared to MSH6 lower levels of MSH2 in cell lines were associated with
shRNA1 PBS control (P=0.0005), these cells were able to resistance to 6-TG and 6-MP. They found 11% of ALL
continue proliferating under the selective pressure, unlike samples showed decreased protein levels of MSH2
the NT cells (Figure 6A and B). Finally, a comparison through copy number loss of genes controlling MSH2
between PBS MSH6 shRNA1 and PBS control NT cells at degradation.17 Thus defects in MMR, including heterozy-
day 17 showed that MSH6 depleted cells also had a gous deletion of MSH6, can be added to the list of genetic
growth advantage in vivo (P<0.0001). alterations that result in the development of resistance to
purine analogs, the foundation of maintenance therapy.
The variety of mutations that lead to selective outgrowth
Discussion of such clones in a substantial number of patients under-
scores the selective pressure of thiopurines on tumor cells.
In recent years, there has been an abundance of evi- The outgrowth of MSH6 deleted/mutated clones not
dence demonstrating the outgrowth of clones at relapse in found at diagnosis has been observed at relapse in malig-
ALL that are associated with unique or enriched relapse nant gliomas following treatment with temozolo-

A B

Figure 5. Knockdown of MSH6 did not lead to a mutator phenotype or increased mutation rate. (A) Microsatellite instability (MSI) was measured in diagnosis/relapse
pairs that had relapse specific, heterozygous MSH6 deletions and in 697 non-targeting (NT) and MSH6 shRNA1 cells left untreated or treated with 0.05 μg/mL of
6-thioguanine (6-TG) for 5 days. (B) Mutation rate in the PIC-A gene was measured in 697 NT and MSH6 shRNA1 clones that were expanded for 2-3 weeks with or
without 6-TG. The cells were analyzed for loss of GPI-dependent cell surface markers, including FLAER, CD48, CD52, and CD59 using flow cytometry. (Left) Individual
mutation rates/cell divisions for each clone; the line represents mean+Standard Deviation. (Right) Mutation rates/cell divisions for three clones with and without 6-
TG treatment.

836 haematologica | 2018; 103(5)


MSH6 haploinsufficiency contributes to resistance

mide,28,36,37 which produces DNA O6-methyguanine, a leads to apoptosis.20,42,43 This pathway is not fully activated
lesion structurally similar to 6-TG.38 Our data demonstrat- in cells with reduced MSH6 because the mismatch goes
ing decreased sensitivity of MSH6 knockdown cells to undetected, allowing these cells to tolerate excess TGN
temozolomide support the hypothesis that MMR defi- mismatches and, ultimately, to continue to survive and
cient clones gain an advantage under this selective pres- proliferate while under treatment. Our data provide evi-
sure leading to resistant recurrences.27,39 The difference in dence that, upon recognition of mismatches, NT cells
TMZ sensitivity between the two UOCB1 shRNA knock- slow their progression through S phase by activating
down cell lines could be due to the interplay between Chk1 as they begin to repair their DNA. Due to the mis-
MSH6 and SETD2 protein levels since UOCB1 cells have match being on the daughter strand, the excision/repair
a copy number loss of SETD2 (NA Evensen et al., 2018, process is unsuccessful, and over time nicks build up in the
unpublished data) and there is a greater reduction of MSH6 DNA, demonstrated by increased levels of γH2AX.
with shRNA1 compared to shRNA2. SETD2, the gene that Eventually, the damage becomes overwhelming and cells
codes for the methyltransferase responsible for the initiate apoptosis, as shown by increased p53. MSH6
trimethylation of H3K36 that serves as the docking site for shRNA1 cells exhibited minimal to no change in cell cycle,
MSH6,40 is among the epigenetic regulators commonly activation of Chk1, or increased γH2AX and p53. The
found mutated in relapse patients.41 Ongoing studies in our moderate changes observed with the MSH6 shRNA2 cells
lab are focused on identifying the relationship of epigenet- highlight the idea that even a more modest reduction in
ic readers, writers, and erasers, such as SETD2, MSH6, MSH6 expression could lead to subtle changes that have a
and WHSC1 in chemoresistance. significant impact on chemoresistance. The MMR defi-
Mechanistically, our in vitro and in vivo data support the cient Reh cells also had no alteration in their cell cycle,
hypothesis that the delayed cytotoxic response to thiop- suggesting that recognition of mismatches by MutSa is
urines is due to the MMR system recognizing a mismatch not sufficient for full activation of this cascade, but rather
and initiating futile, damaging DNA repair that ultimately damage induced by the repair, which is orchestrated by

B C

Figure 6. Knockdown of MSH6 leads to decreased sensitivity to purixan in vivo. (A) Bioluminescence imaging (BLI) of mice injected with UOCB1 non-targeting (NT)
or MSH6 shRNA1 cells. Six days after injection mice were imaged and then randomized to treatment. Treatment was started on day 7 and images were taken again
on days 13 and 17. C: PBS control treatment; T: purixan treatment. (B) Quantification of total flux was determined by analyzing the BLI images using Living Image
software. (C) Western blot to confirm knockdown of MSH6 in cells used to inject mice. Actin was used as loading control.

haematologica | 2018; 103(5) 837


N.A. Evensen et al.

MutLa, is what initiates apoptosis. Our data demonstrat- gosity.50 Thus, our work supports a model whereby hap-
ing the involvement of the ATR-Chk1-H2AX signaling loinsufficiency of MSH6 results in TGN tolerance and
cascade is supported by the work of Eich et al. which resistance directly rather than by generation of secondary
demonstrated activation of this pathway upon treatment mutations. However, it does not rule out the possibility
with temozolomide.32 Interestingly, understanding how that haploinsufficiency, along with other defects in the
MMR deficient cells respond to thiopurines in terms of MMR pathway, may result in a mutator phenotype.
TGN incorporation could prove essential given the emerg- Overall, it has become increasingly evident that the
ing idea of measuring these parameters in patients on genetic and epigenetic landscape of cancer cells is vital to
maintenance therapy.44 the overall effectiveness of treatment. These studies illus-
The data presented here do not support the hypothesis trate yet another example of a mutation/deletion found at
of increased mutation burden, genomic instability, or MSI relapse that directly influences the response to a therapeu-
when MSH6 is reduced. Our inability to demonstrate tic agent that is currently heavily relied on. Continuous
MSI-high, which is considered a standard method for clin- efforts to elucidate the potential functions and mecha-
ical testing of MMR deficiencies in tumors,45 in MSH6 nisms of genes found mutated at relapse will help lead us
depleted cell lines and clinical samples is consistent with to novel treatment strategies.
the lack of MSI in glioma samples with MSH6 deletions or
mutations.46,47 Haploinsufficiency of MSH6 or compensa- Funding
tion by MSH2/MSH3 may account for this observation.25,48 This work was supported by the Leukemia and Lymphoma
In addition, ALL clonal evolution from diagnosis to relapse Society SCOR grant: 7010-14 (WLC, JY, DT, SPH), the US
is not associated with increased mutation burden support- National Institutes of Health (NIH) funded grant RO1
ing our mutation rate analysis, although Ma et al. reported CA140729 (WLC), and the Perlmutter Cancer Center Support
a subset of hypermutated relapse cases.49 Of these, one Grant: P30 DA016087.
had a bialleic mutation of PMS2, another had multiple
damaging MSH6 mutations as well as an MLH1 splice site Acknowledgments
mutation, while the others harbored no MMR muta- We gratefully acknowledge the Children’s Oncology Group
tions.49 Furthermore, one case demonstrated that a het- (COG) Specimen Bank for samples. Support for flow cytometry
erozygous deletion of MSH6 at diagnosis was not suffi- was provided by NYU School of Medicine’s Cytometry and Cell
cient to cause a hypermutator phenotype, but the acquisi- Sorting Laboratory, which is supported in part by grant
tion of a second hit in the WT allele at relapse was.49 P30CA016087 from the NIH/NCI, and the CHOP Flow
Likewise, the majority of hypermutated gliomas at relapse Cytometry Core. We acknowledge the VA-Mertid award
show defects in multiple MMR genes or loss of heterozy- 1I01BX-000670, which helped support this work.

References reveals therapeutic strategies. Blood. 17. Diouf B, Cheng Q, Krynetskaia NF, et al.
2011;118(19):5218-5226. Somatic deletions of genes regulating
9. Li GM. Mechanisms and functions of MSH2 protein stability cause DNA mis-
1. Pui CH, Evans WE. Treatment of acute DNA mismatch repair. Cell Res. 2008;18 match repair deficiency and drug resistance
lymphoblastic leukemia. N Engl J Med. (1):85-98. in human leukemia cells. Nat Med.
2006;354(2):166-178. 10. Edelbrock MA, Kaliyaperumal S, Williams 2011;17(10):1298-1303.
2. Meyer JA, Wang J, Hogan LE, et al. Relapse- KJ. Structural, molecular and cellular func- 18. Swann PF, Waters TR, Moulton DC, et al.
specific mutations in NT5C2 in childhood tions of MSH2 and MSH6 during DNA Role of postreplicative DNA mismatch
acute lymphoblastic leukemia. Nat Genet. mismatch repair, damage signaling and repair in the cytotoxic action of thiogua-
2013;45(3):290-294. other noncanonical activities. Mutat Res. nine. Science. 1996;273(5278):1109-1111.
3. Jones CL, Bhatla T, Blum R, et al. Loss of 2013;743-744:53-66. 19. Waters TR, Swann PF. Cytotoxic mecha-
TBL1XR1 disrupts glucocorticoid receptor 11. Dunlop MG, Farrington SM, Carothers AD, nism of 6-thioguanine: hMutSalpha, the
recruitment to chromatin and results in glu- et al. Cancer risk associated with germline human mismatch binding heterodimer,
cocorticoid resistance in a B-lymphoblastic DNA mismatch repair gene mutations. binds to DNA containing S6-methylth-
leukemia model. J Biol Chem. 2014; Hum Mol Genet. 1997;6(1):105-110. ioguanine. Biochemistry. 1997;36(9):2501-
289(30):20502-20515. 12. Tiwari AK, Roy HK, Lynch HT. Lynch syn- 2506.
4. Li B, Li H, Bai Y, et al. Negative feedback- drome in the 21st century: clinical perspec- 20. Karran P, Attard N. Thiopurines in current
defective PRPS1 mutants drive thiopurine tives. QJM. 2016;109(3):151-158. medical practice: molecular mechanisms
resistance in relapsed childhood ALL. Nat 13. Goecke T, Schulmann K, Engel C, et al. and contributions to therapy-related cancer.
Med. 2015;21(6):563-571. Genotype-phenotype comparison of Nat Rev Cancer. 2008;8(1):24-36.
5. Mullighan CG, Zhang J, Kasper LH, et al. German MLH1 and MSH2 mutation carri- 21. Morrison DJ, Hogan LE, Condos G, et al.
CREBBP mutations in relapsed acute lym- ers clinically affected with Lynch syn- Endogenous knockdown of survivin
phoblastic leukaemia. Nature. 2011; drome: a report by the German HNPCC improves chemotherapeutic response in
471(7337):235-239. Consortium. J Clin Oncol. 2006; ALL models. Leukemia. 2012;26(2):271-
6. Nielsen SN, Grell K, Nersting J, et al. DNA- 24(26):4285-4292. 279.
thioguanine nucleotide concentration and 14. Ripperger T, Schlegelberger B. Acute lym- 22. Araten DJ, Golde DW, Zhang RH, et al. A
relapse-free survival during maintenance phoblastic leukemia and lymphoma in the quantitative measurement of the human
therapy of childhood acute lymphoblastic context of constitutional mismatch repair somatic mutation rate. Cancer Res.
leukaemia (NOPHO ALL2008): a prospec- deficiency syndrome. Eur J Med Genet. 2005;65(18):8111-8117.
tive substudy of a phase 3 trial. Lancet 2016;59(3):133-142. 23. Araten DJ, Sanders KJ, Anscher D,
Oncol. 2017;18(4):515-524. 15. Ripperger T, Beger C, Rahner N, et al. Zamechek L, Hunger SP, Ibrahim S.
7. Yang JJ, Bhojwani D, Yang W, et al. Constitutional mismatch repair deficiency Leukemic blasts with the paroxysmal noc-
Genome-wide copy number profiling and childhood leukemia/lymphoma-- turnal hemoglobinuria phenotype in chil-
reveals molecular evolution from diagnosis report on a novel biallelic MSH6 mutation. dren with acute lymphoblastic leukemia.
to relapse in childhood acute lymphoblastic Haematologica. 2010;95(5):841-844. Am J Pathol. 2012;181(5):1862-1869.
leukemia. Blood. 2008;112(10):4178-4183. 16. Fink D, Aebi S, Howell SB. The role of 24. Jacobsen JH, Schmiegelow K, Nersting J.
8. Hogan LE, Meyer JA, Yang J, et al. DNA mismatch repair in drug resistance. Liquid chromatography-tandem mass spec-
Integrated genomic analysis of relapsed Clin Cancer Res. 1998;4(1):1-6. trometry quantification of 6-thioguanine in
childhood acute lymphoblastic leukemia

838 haematologica | 2018; 103(5)


MSH6 haploinsufficiency contributes to resistance

DNA using endogenous guanine as internal 33. Verma L, Kane MF, Brassett C, et al. diatric acute lymphoblastic leukaemia. Nat
standard. J Chromatogr B Analyt Technol Mononucleotide microsatellite instability Commun. 2014;5:3469.
Biomed Life Sci. 2012;881-882:115-118. and germline MSH6 mutation analysis in 42. Yan T, Berry SE, Desai AB, Kinsella TJ.
25. Gu L, Cline-Brown B, Zhang F, Qiu L, Li early onset colorectal cancer. J Med Genet. DNA mismatch repair (MMR) mediates 6-
GM. Mismatch repair deficiency in hema- 1999;36(9):678-682. thioguanine genotoxicity by introducing
tological malignancies with microsatellite 34. Bodo S, Colas C, Buhard O, et al. Diagnosis single-strand breaks to signal a G2-M arrest
instability. Oncogene. 2002;21(37):5758- of Constitutional Mismatch Repair- in MMR-proficient RKO cells. Clin Cancer
5764. Deficiency Syndrome Based on Res. 2003;9(6):2327-2334.
26. Marra G, Iaccarino I, Lettieri T, Roscilli G, Microsatellite Instability and Lymphocyte 43. Tidd DM, Paterson AR. Distinction
Delmastro P, Jiricny J. Mismatch repair defi- Tolerance to Methylating Agents. between inhibition of purine nucleotide
ciency associated with overexpression of Gastroenterology. 2015;149(4):1017-1029. synthesis and the delayed cytotoxic reac-
the MSH3 gene. Proc Natl Acad Sci USA. e1013. tion of 6-mercaptopurine. Cancer Res.
1998;95(15):8568-8573. 35. Araten DJ, Krejci O, Ditata K, et al. The rate 1974;34(4):733-737.
27. McFaline-Figueroa JL, Braun CJ, Stanciu M, of spontaneous mutations in human 44. Nielsen SN, Grell K, Nersting J, Frandsen
et al. Minor Changes in Expression of the myeloid cells. Mutat Res. 2013;749(1-2):49- TL, Hjalgrim LL, Schmiegelow K. Measures
Mismatch Repair Protein MSH2 Exert a 57. of 6-mercaptopurine and methotrexate
Major Impact on Glioblastoma Response to 36. Cahill DP, Codd PJ, Batchelor TT, Curry maintenance therapy intensity in child-
Temozolomide. Cancer Res. 2015; WT, Louis DN. MSH6 inactivation and hood acute lymphoblastic leukemia.
75(15):3127-3138. emergent temozolomide resistance in Cancer Chemother Pharmacol. 2016;
28. Xie C, Sheng H, Zhang N, Li S, Wei X, human glioblastomas. Clin Neurosurg. 78(5):983-994.
Zheng X. Association of MSH6 mutation 2008;55:165-171. 45. Buza N, Ziai J, Hui P. Mismatch repair defi-
with glioma susceptibility, drug resistance 37. Cahill DP, Levine KK, Betensky RA, et al. ciency testing in clinical practice. Expert
and progression. Mol Clin Oncol. 2016; Loss of the mismatch repair protein MSH6 Rev Mol Diagn. 2016;16(5):591-604.
5(2):236-240. in human glioblastomas is associated with 46. Yip S, Miao J, Cahill DP, et al. MSH6 muta-
29. Matheson EC, Hall AG. Assessment of mis- tumor progression during temozolomide tions arise in glioblastomas during temo-
match repair function in leukaemic cell treatment. Clin Cancer Res. 2007; zolomide therapy and mediate temozolo-
lines and blasts from children with acute 13(7):2038-2045. mide resistance. Clin Cancer Res.
lymphoblastic leukaemia. Carcinogenesis. 38. Zhang J, Stevens MF, Laughton CA, 2009;15(14):4622-4629.
2003;24(1):31-38. Madhusudan S, Bradshaw TD. Acquired 47. Maxwell JA, Johnson SP, McLendon RE, et
30. Yoshioka K, Yoshioka Y, Hsieh P. ATR resistance to temozolomide in glioma cell al. Mismatch repair deficiency does not
kinase activation mediated by MutSalpha lines: molecular mechanisms and potential mediate clinical resistance to temozolo-
and MutLalpha in response to cytotoxic translational applications. Oncology. 2010; mide in malignant glioma. Clin Cancer Res.
O6-methylguanine adducts. Mol Cell. 78(2):103-114. 2008;14(15):4859-4868.
2006;22(4):501-510. 39. Hunter C, Smith R, Cahill DP, et al. A 48. Umar A, Risinger JI, Glaab WE, Tindall KR,
31. Yan T, Desai AB, Jacobberger JW, hypermutation phenotype and somatic Barrett JC, Kunkel TA. Functional overlap
Sramkoski RM, Loh T, Kinsella TJ. CHK1 MSH6 mutations in recurrent human in mismatch repair by human MSH3 and
and CHK2 are differentially involved in malignant gliomas after alkylator MSH6. Genetics. 1998;148(4):1637-1646.
mismatch repair-mediated 6-thioguanine- chemotherapy. Cancer Res. 2006; 49. Ma X, Edmonson M, Yergeau D, et al. Rise
induced cell cycle checkpoint responses. 66(8):3987-3991. and fall of subclones from diagnosis to
Mol Cancer Ther. 2004;3(9):1147-1157. 40. Li F, Mao G, Tong D, et al. The histone relapse in pediatric B-acute lymphoblastic
32. Eich M, Roos WP, Nikolova T, Kaina B. mark H3K36me3 regulates human DNA leukaemia. Nat Commun. 2015;6:6604.
Contribution of ATM and ATR to the mismatch repair through its interaction 50. van Thuijl HF, Mazor T, Johnson BE, et al.
resistance of glioblastoma and malignant with MutSalpha. Cell. 2013;153(3):590-600. Evolution of DNA repair defects during
melanoma cells to the methylating anti- 41. Mar BG, Bullinger LB, McLean KM, et al. malignant progression of low-grade
cancer drug temozolomide. Mol Cancer Mutations in epigenetic regulators includ- gliomas after temozolomide treatment.
Ther. 2013;12(11):2529-2540. ing SETD2 are gained during relapse in pae- Acta Neuropathol. 2015;129(4):597-607.

haematologica | 2018; 103(5) 839


ARTICLE Hodgkin Lymphoma

A phase II study of the oral JAK1/JAK2


Ferrata Storti
Foundation inhibitor ruxolitinib in advanced
relapsed/refractory Hodgkin lymphoma
Eric Van Den Neste,1 Marc André,2 Thomas Gastinne,3 Aspasia Stamatoullas,4
Corinne Haioun,5 Amine Belhabri,6 Oumedaly Reman, 7Olivier Casasnovas,8
Hervé Ghesquieres,9 Gregor Verhoef,10 Marie-José Claessen,11 Hélène A.
Poirel,12 Marie-Christine Copin,13 Romain Dubois,13 Peter Vandenberghe,14
Ioanna-Andrea Stoian,15 Anne S. Cottereau,16 Sarah Bailly,1 Laurent Knoops17
and Franck Morschhauser18
Haematologica 2018
Department of Hematology, Cliniques Universitaires Saint-Luc, UCL Brussels, Belgium;
Volume 103(5):840-848
1

2
Hematology Department, CHU UCL Namur, Yvoir, Belgium; 3Hematology, CHU Nantes,
France; 4Clinical Hematology, Centre Henri Becquerel, Rouen, France; 5Lymphoid
Malignancies Unit, AP-HP, Groupe Hospitalier Mondor, Créteil, France; 6Onco-hematology,
Centre Leon Berard, University Claude Bernard Lyon 1, France; 7Hematology, Centre
Hospitalier Universitaire, Caen, France; 8Hematology Department, Hopital Le Bocage, CHU
Dijon, France; 9Hospices Civils de Lyon, Université Claude Bernard, Centre Hospitalier Lyon-
Sud, Pierre Bénite, France; 10Department of Hematology, University Hospitals Leuven,
Belgium; 11Erasmus MC, Rotterdam, the Netherlands; 12Center for Human Genetics,
Cliniques universitaires Saint-Luc, Université Catholique de Louvain, Brussels, Belgium;
13
CHRU de Lille, France; 14Center for Human Genetics, Katholieke Universiteit - Leuven,
Belgium; 15Nuclear Medicine, Cliniques Universitaires Saint-Luc, UCL Brussels, Belgium;
16
Nuclear Medicine, Hôpital Tenon, Paris, France; 17Cliniques Universitaires Saint-Luc and
de Duve Institute, Université Catholique de Louvain, Brussels, Belgium and 18CHU Lille,
Hematology Department, and Université de Lille, GRITA, France
Preliminary results were presented at the 58th
Annual Meeting of the American Society of
Hematology, held on December 3 – 6, 2016,
in San Diego, USA.

ABSTRACT

J
AK2 constitutive activation/overexpression is common in classical
Correspondence: Hodgkin lymphoma, and several cytokines stimulate Hodgkin lym-
franck.morschhauser@chru-lille.fr
phoma cells by recognizing JAK1-/JAK2-bound receptors. JAK block-
ade may thus be therapeutically beneficial in Hodgkin lymphoma. In this
phase II study we assessed the safety and efficacy of ruxolitinib, an oral
JAK1/2 inhibitor, in patients with relapsed/refractory Hodgkin lym-
Received: September 12, 2017.
phoma. The primary objective was overall response rate according to the
Accepted: January 10, 2018.
International Harmonization Project 2007 criteria. Thirty-three patients
Pre-published: January 19, 2018. with advanced disease (median number of prior lines of treatment: 5;
refractory: 82%) were included; nine (27.3%) received at least six cycles
of ruxolitinib and six (18.2%) received more than six cycles. The overall
doi:10.3324/haematol.2017.180554
response rate after six cycles was 9.4% (3/32 patients). All three respon-
Check the online version for the most updated ders had partial responses; another 11 patients had transient stable dis-
information on this article, online supplements, ease. Best overall response rate was 18.8% (6/32 patients). Rapid allevia-
and information on authorship & disclosures: tion of B-symptoms was common. The median duration of response was
www.haematologica.org/content/103/5/840
7.7 months, median progression-free survival 3.5 months (95% CI: 1.9-
4.6), and the median overall survival 27.1 months (95% CI: 14.4-27.1).
©2018 Ferrata Storti Foundation Forty adverse events were reported in 14/33 patients (42.4%). One event
Material published in Haematologica is covered by copyright. led to treatment discontinuation, while 87.5% of patients recovered
All rights are reserved to the Ferrata Storti Foundation. Use of without sequelae. Twenty-five adverse events were grade 3 or higher.
published material is allowed under the following terms and
conditions:
These events were mostly anemia (n=11), all considered related to ruxoli-
https://creativecommons.org/licenses/by-nc/4.0/legalcode. tinib. Other main causes of grade 3 or higher adverse events included
Copies of published material are allowed for personal or inter- lymphopenia and infections. Of note, no cases of grade 4 neutropenia or
nal use. Sharing published material for non-commercial pur-
poses is subject to the following conditions: thrombocytopenia were observed. Ruxolitinib shows signs of activity,
https://creativecommons.org/licenses/by-nc/4.0/legalcode, albeit short-lived, beyond a simple anti-inflammatory effect. Its limited
sect. 3. Reproducing and sharing published material for com-
mercial purposes is not allowed without permission in writing
toxicity suggests that it has the potential to be combined with other ther-
from the publisher. apeutic modalities. ClinicalTrials.gov: NCT01877005

840 haematologica | 2018; 103(5)


Ruxolitinib in advanced relapsed/refractory HL

Introduction Methods
Hodgkin lymphoma (HL) is regarded as a curable malig- Patients’ eligibility
nancy in most cases, yet treatment failure still occurs in Patients aged 18 years or older with a diagnosis of R/R HL for
about 10% of patients with early-stage disease.1 In whom no treatment with proven efficacy was available were eli-
advanced-stage disease, up to 10% of cases do not reach gible to enter the trial after having receiving at least one prior
complete remission and are thus considered to have pri- therapy provided that they had measurable nodal disease at
mary refractory HL,2 while 20-30% of primary responders baseline (≥1 cm in the longest transverse diameter, clearly meas-
eventually relapse following first-line treatment.3 urable in at least two perpendicular dimensions) on computed
For most patients with relapsed or refractory HL (R/R tomography or magnetic resonance imaging, as well as an
HL), the standard of care consists of high-dose salvage Eastern Cooperative Oncology Group performance score of ≤3.
chemotherapy followed by autologous stem-cell transplan- Additional inclusion criteria were an absolute neutrophil count
tation (SCT). For patients who develop R/R HL within 1 ≥1.0 x 109/L, platelet count ≥75 x 109/L, serum creatinine ≤1.5 x
year of autologous SCT, the prognosis proves extremely upper limit of normal, serum bilirubin ≤1.5 x upper limit of nor-
poor, since they have a median survival of 1.2 years.4 For mal, and ALT and AST levels ≤2.5 or ≤5.0 x upper limit of nor-
patients in whom all classical approaches have failed, new mal in the event the transaminase increase was due to HL-relat-
strategies, including checkpoint inhibitors targeting PD-1 ed liver disease. Pregnant or lactating patients were not allowed
and antibody-drug conjugates targeting CD30, have to enter the trial, and men and women of childbearing potential
become part of the therapeutic armamentarium against had to agree to employ an adequate contraceptive method dur-
R/R HL.5-8 However, patients with multiple relapses or ing the study treatment. Patients were permitted to have
those who develop refractory disease remain in medical received an undefined number of prior lines of therapy, and a
need, especially those in whom treatment with brentux- previous allogeneic SCT was likewise allowed provided that
imab-vedotin (BV) and PD-1 blockers fails. patients had not received any immunosuppressive therapy with-
Classical HL is characterized by the presence of in the 90 days prior to starting the screening procedures. Patients
Hodgkin and Reed-Sternberg (HRS) cells and their vari- were required to have a life expectancy of ≥3 months.
ants.9 HRS cells were demonstrated to shape their envi-
ronment by secreting immunosuppressive cytokines and Study design and treatment
chemokines.10 With this in mind, the Janus kinase (JAK) – This multicenter, open-label, phase II study (HIJAK,
signal transducer and activator of transcription (STAT) NCT01877005) was conducted at ten LYSA centers in France and
pathway appears to be a relevant cytokine-induced signal Belgium, with patients recruited from July 2013 through
transduction pathway that has been shown to transfer sig- December 2014. Its primary efficacy endpoint was overall
nals directly from cell surface cytokine receptors to the cell response rate (ORR), defined as the proportion of patients with a
nucleus. Given that enhanced JAK-mediated signaling has complete response or partial response at 6 months of treatment by
been demonstrated in a significant number of HL investigator assessment based on the revised 2007 International
patients,11 this signaling pathway has become a focus for Harmonization Project response criteria for malignant lym-
developing novel therapeutic agents for the disease. Van phoma.20 Secondary objectives included relief of B symptoms, best
Roosbroeck et al. reported the translocation of JAK2 in ORR (occurring at any time during study), duration of response,
several cases of HL,12 and JAK inhibition was shown to progression-free survival, overall survival, as well as the incidence
decrease the proliferation of cell lines. Although such and severity of adverse events.
translocations are relatively rare, 9p24.1 genomic amplifi- The study was carried out in line with the ethical principles of
cation including the JAK2 locus appears common in HL, the Helsinki Declaration and in compliance with the International
along with increased protein expression and activity, Conference on Harmonization Guideline for Good Clinical
resulting in the constitutive activation of STAT6, an essen- Practice. The protocol was approved by the institutional review
tial messenger of tumor cell growth.13-15 In corollary, JAK board of each study site and written informed consent was
1/2 inhibition may be suitable to target the constitutive obtained from all patients.
activation caused by either JAK2 translocation or JAK2 The starting dose of ruxolitinib was 20 mg given twice daily dur-
amplification and to modify the reactive microenviron- ing six 28-day cycles for the induction period if the platelet count
ment which contributes to HL growth via aberrant was >200 x 109/L. The ruxolitinib dose was decreased to 15 mg
cytokine production.16 twice daily in patients with platelet counts between 75 x 109/L and
Ruxolitinib is the first potent, selective, and oral inhibitor 200 x 109/L. Patients who achieved at least stable disease at the end
of JAK1/2 being developed for clinical use.17 Its major of cycle 6 and who had, in the investigator's opinion, a clinical ben-
effects include inhibition of proliferation, induction of efit were eligible to continue ruxolitinib (15 mg or 20 mg), which
apoptosis, and reduction in cytokine plasma levels, all was defined as “maintenance” therapy. Treatment could be contin-
mediated by the drug's ability to inhibit JAK-induced phos- ued for up to 2 years or until progressive disease, intolerability, or
phorylation of STAT.18 Used in the treatment of myelofi- as long as the investigator thought that there was clinical benefit.
brosis, ruxolitinib had durable efficacy in reducing Administration of the study drug could be stopped for any grade
splenomegaly and alleviating constitutional symptoms, the ≥3 non-hematologic toxicity, with the exception of deep venous
patients gained weight and their general physical condition thrombosis and alopecia. Following event resolution to grade ≤1,
improved.19 The dose-limiting toxicity was thrombocy- ruxolitinib could be resumed, with a 5 mg dose reduction and a
topenia, which was fairly well managed by dose reduc- maximum delay of 4 weeks. Mandatory dose decreases or inter-
tions or brief interruptions of treatment. In the present ruptions for hematologic toxicity as well as the rules for perma-
phase II study, we sought to investigate the safety and effi- nent discontinuation are detailed in the Online Supplementary
cacy of ruxolitinib in patients with R/R HL. Exploratory Appendix. Growth factors were allowed as per American Society of
biomarker analyses pertaining to plasma cytokine profiles Clinical Oncology guidelines and infectious prophylaxis as per the
and aberrations of JAK2 were also carried out. guidelines of Heine et al.21

haematologica | 2018; 103(5) 841


E. Van Den Neste et al.

Study assessments safety analysis comprised all patients who received at least one
Baseline assessments comprised documentation of disease- dose of the study drug. All statistical analyses were performed
related symptoms, physical examination, laboratory tests, and using SAS software, version 9.2. P-values <0.05 were consid-
imaging studies of the neck, chest, abdomen, and pelvis, using ered statistically significant. All available data were included in
computed tomography or magnetic resonance imaging. Biopsy data listings and tabulations, with no imputations of values for
prior to inclusion was recommended, but not mandatory. Tumors missing data. An interim analysis was neither planned nor per-
were measured at baseline, at the end of every two cycles of rux- formed.
olitinib, and following the six-cycle induction, as well as during
maintenance therapy. Given the exploratory nature of the study,
there was no centralized review of computed tomography Results
response. However, positron emission tomographic images of the
responders were all centrally reviewed by a nuclear physician Patients’ disposition and characteristics
(ASC) to confirm partial or complete metabolic response based on The patients’ characteristics are listed in Table 1. From
the Deauville five-point scale. The evaluable study population for July 2013 to December 2014, a total of 33 patients with
efficacy was restricted to patients who had received at least 28 R/R HL were recruited. Their median age was 37 years
days of the study drug. (range, 19-80). Most of the patients had advanced HL
Safety was monitored for up to 1 month after treatment. (stage III/IV) and had been heavily pretreated, with a
Adverse events were summarized by means of the Medical median number of five prior regimens including autolo-
Dictionary for Regulatory Activities, and graded using the gous SCT (54%), allogeneic SCT(15%), and BV (82%).
National Cancer Institute’s Common Terminology Criteria for Of the 33 patients recruited, 27 (82%) had refractory HL
Adverse Events (NCI-CTCAE), version 3.0. Laboratory abnormal- and 22 had biopsy-confirmed relapse of HL. Among the
ities were assessed according to NCI-CTCAE version 4.0. Only six patients displaying a response, a biopsy was per-
grade 3 or 4 toxicities and grade 2 infections were to be reported. formed in five of them at relapse [8 days, 12 days, 6
All patients were included in the toxicity analysis. weeks (n=2) and 14 months prior to inclusion in the
study].
Exploratory biomarker analysis
Blood samples (5 mL) were taken at baseline prior to drug
administration and on day 1 of cycle 2 for the measurement of Table 1. Patient’s demographics and characteristics.
27 cytokines related to the immune system using bead-based Patients’ demographics and characteristics All patients (n=33)
immunoassays. JAK2 gains, amplifications, and gene rearrange-
Gender, n (%)
ments were also investigated using fluorescent in situ hybridiza-
Male 21 (63.6%)
tion with two tri-color sets of probes associating JAK2/9p24 Female 12 (36.4%)
break-apart probes with a control centromeric probe
Age in years, median (range) 37.0 (19.0-80.0)
(CEP9/9q21): the already prepared probes from Empire
genomics on the one hand, and the association of the JAK2 B/A ECOG score
probe from Kreatech with the CEP9 probe from Vysis on the 0 11 (33.3%)
other hand. The CD274/PDL1 and PDCD1LG2/PDL2 loci at 1 15 (45.5%)
9p24 were studied with home-made prepared bacterial artificial 2 5 (15.2%)
chromosome probes purchased from the Chori BACPAC
3 2 (6.1%)
Resources Center (Oakland, CA, USA). Extraction, labeling and Ann Arbor stage
hybridization were performed on paraffin-embedded tissue, as I 1 (3.0%)
previously reported.22 II 7 (21.2%)
III 3 (9.1%)
Statistical methods IV 22 (66.7%)
The sample size for this phase II study was calculated using B symptoms
an exact single-stage phase II design.23 A two-stage design with Yes 16 (48.5%)
interim analysis for activity or toxicity was not planned given No 17 (51.5%)
the very advanced stage of the patients, the relative paucity of Extranodal involvement
alternative options, and the potential toxicity of ruxolitinib that Bone 13 (39.4%)
was expected to be in the low range, based on myelofibrosis Liver 6 (18.2%)
data. The treatment was considered ineffective if the ORR was Lung 12 (36.4%)
≤15%, and effective if the ORR was ≥35%. Under the assump- Soft tissues 4 (12.1%)
tion of an alpha first-order risk error set at 5% and beta at 20% Time since initial diagnosis in months, median (range) 55.4 (8.7 – 216.1)
with a one-sided test, it was deemed necessary to include a Prior therapies
total of 28 evaluable patients with a cut-off number of eight. If Prior lines, median (range) 5 (1 – 16)
at least eight patients had a response, the hypothesis of an ORR Chemotherapy 33 (100%)
≤15% was rejected with both a target error rate and an actual Radiotherapy 18 (54.5%)
error rate of 0.05. If seven or fewer patients had a response, the Brentuximab vedotin 27 (82%)
hypothesis of an ORR ≥35% was rejected with a target error Autologous SCT 18 (54.5%)
rate of 0.2 and an actual error rate of 0.187. The ORR estimate Allogeneic SCT 5 (15.2%)
and its 90% confidence intervals (CI) were calculated for all Interval since last treatment in months, median (range) 6 (1.1 – 75.0)
patients who completed at least one cycle of the study drug. Disease status at inclusion
The Kaplan-Meier method was employed to estimate the Relapse 6
median value and its 95% CI for time to response, duration of Refractory 27 (81.8%)
response, progression-free survival and overall survival. The SCT: stem cell transplantation; ECOG: Eastern Cooperative Oncology Group.

842 haematologica | 2018; 103(5)


Ruxolitinib in advanced relapsed/refractory HL

Patients’ exposure to the study drug response after six cycles of treatment, eventually entered
The median number of ruxolitinib cycles administered complete remission during the follow-up, beyond the six
was four (range, 1 to 12) (Table 2). Nine patients received cycles. Achievement of complete metabolic response was
all six of the planned cycles of ruxolitinib and six of these confirmed by central review. At the time of writing, two
patients continued on maintenance therapy with the JAK patients (UPN 611001 and 881001) are still taking ruxoli-
inhibitor. The remainder discontinued ruxolitinib therapy, tinib. Figure 4 illustrates changes in target tumor measure-
most because of progressive disease and in one case due to ments in individual patients. The best reduction, if any, at
adverse events. any time throughout treatment is shown.
In addition, during the 6-month induction, transient sta-
Responses and outcomes ble disease was recorded in 11 patients, albeit of limited
The patients’ disposition through the study is illustrated duration. Overall, the disease control rate (including stable
in Figure 1. Among the 33 HL patients included in the trial, disease with complete and partial responses) was 53.1%
one patient did not complete the first cycle of treatment (17/32 patients) (95% CI: 34.7-70.9%) with a median
because of progressive disease and was not, therefore, duration of 1.9 months.
included in the efficacy analysis. At the end of the ruxoli- The alleviating effect on systemic symptoms, such as
tinib induction period (6 months) three of 32 patients had pruritus, fever, and sweating, was noteworthy, starting
a response, for an ORR of 9.4% (90% CI: 2.6-22.5%); the within the first month of drug administration and com-
response in all three was partial. At some point during monly lasting. The impact was most remarkable on the
induction six of the 32 patients had a response, which
was, in all six cases a partial response, for a best ORR of
18.8% (95% CI: 7.2-36.4%). A detailed analysis of the
responders’ characteristics is provided in Table 3. Figures 2
and 3 illustrate metabolic evolution in two patients.
Interestingly, UPN 611001, who had achieved a partial

Table 2. Treatment exposure and modifications.


Treatment exposure and modifications All patients (n=33)
Cycles given, number, median (range) 4 (1-22)
Received full induction (6 cycles), % 9 (27.3%)
Received maintenance (> 6 cycles), % 6 (18.2%)
Percentage of planned dosea
<75% 3 (9.1%)
75%-90% 4 (12.1%)
90%-110% 25 (75.8%)
110%-125% 1 (3.0%)
Dose modification
Yes 20 (60.6%)
Type of modificationb
Dose reduction 2 (10.0%)
Dose interruption 18 (90.0%)
Dose increase 3 (15.0%)
Number of days of interruption if any, 4 (1 – 28)
median (range)
a
Defined as follows: (total number of tablets taken/total expected number of tablets)

Figure 1. Patients’ disposition. *N months on maintenance therapy: 4, 6, 6, 21,


*100, taking into account protocol-defined dose reduction. bThe total sum of the per-

16, 22; PD: progressive disease.


centages for the type and the reasons of modification may be greater than 100.0% as
a patient may have had several types of modification and reasons for treatment mod-
ification.

Table 3. Characteristics of responders (best response achieved during the 6-month ruxolitinib induction).
UPN Prior treatment Extranodal involvement Response (Cheson 2007)20
N Type
611001 9 ABVD, BEACOPP, MINE, IGEV, GVD, CAELYX, GVD, RT, BV Liver PR*,1
211004 8 ABVD, RT, IVA, transplantation, MINE, GVD, BV, ASHAP Breast PR
601001 5 BEACOPP, DHAP, IGEV, transplantation-RT, BV Liver, bone, lung PR
601004 5 ABVD, DHAP, RT, RT, BV None PR
641001 1 ABVD2 None PR
881001 5 ABVD, transplantation, MINE, BV, GEMOX Lung PR1
*Patient eventually achieved a complete response during maintenance therapy. 1Patients still under treatment with ruxolitinib at the time of writing; 2Patient with morbid obesity
not eligible for standard approaches with chemo/immunotherapy. UPN:unique patient number; RT: radiotherapy; BV: brentuximab vedotin; PR: partial response.

haematologica | 2018; 103(5) 843


E. Van Den Neste et al.

control of pruritus, which affected 35.5% of patients prior Safety


to initiating therapy but only 6.6% after the first cycle of All patients enrolled in the study received at least one
ruxolitinib. Sweating, which was present in 32.2% of the dose of study medication and were, therefore, included in
patients at inclusion, was reduced, with 20% still having the safety analysis. Overall, 40 adverse events were
this symptom after one cycle of treatment. Fever was observed in 14/33 patients (42.4%). In six patients, the
abolished in 3/4 patients with this symptom at inclusion. adverse events were related to the ruxolitinib therapy. In
The median follow-up was 17.5 months. As illustrated eight of them, the events were of grade ≥3 (Table 4A).
in Figure 5, the median progression-free survival was 3.5 Among the 40 adverse events recorded, 30 (75%) occurred
months (95% CI: 1.9-4.6). The median duration of during induction and 18 (45%) were related to ruxolitinib.
response was 7.7 months (95% CI: 1.8-NA) for the six No drug-related deaths were recorded. One adverse event
patients who eventually a achieved response (data not resulted in permanent drug discontinuation, while 87.5%
shown). Overall, 30 patients had progressive disease, with of the adverse events resolved without sequelae. The
97% at the initial site and/or 60% at new sites. Following characteristics and grade of the adverse events, listed by
ruxolitinib discontinuation, 25 (83.3%) patients were system organ class and preferred terms, are displayed in
given further treatments, consisting of chemotherapy in Table 4B. Twenty-five (62.5%) were of grade ≥3. These
19, and immunotherapy in nine, the latter comprising rit- were mostly anemia (n=11), all considered related to rux-
uximab in four, BV in three, and nivolumab in two. olitinib. Other main causes of grade ≥3 adverse events
Transplantation was eventually carried out in five included lymphopenia (n=4), infections (n=3) and miscel-
patients, and was allogeneic in four cases and autologous laneous causes. Of note, no cases of grade 4 neutropenia
in the remaining one. Among the 25 patients prescribed or thrombocytopenia were observed.
further therapy, the observed complete and partial Eight serious adverse events were reported in four
response rates were 10% and 15%, respectively. Overall, patients, during induction (n=5), maintenance (n=1) or
12 patients died on account of lymphoma progression after the end of treatment (n=2). These serious adverse
(83.3%), toxicity of other treatments (8.3%), or other rea- events consisted of infection in three patients (device-
sons (8.3%). The median overall survival was 27.1 months related sepsis, gastroenteritis, and lung infection). The
(95% CI: 14.4-27.1). other serious adverse events were anemia, diarrhea, sub-
dural hematoma, bone pain, and pulmonary embolism.
Two serious adverse events (anemia, lung infection) were
deemed drug-related and six were considered grade ≥3:
infection (n=3), anemia, subdural hematoma, and pul-
A monary embolism. Of the eight serious adverse events, six
resolved without sequelae, while the device-related sepsis
and pulmonary embolism, observed in the same patient,
persisted until the patient died due to progressive disease
and were thus not considered as the cause of death.
Among the 33 patients, one second primary malignancy
was observed (adenocarcinoma of the colon in an 80-year
old male patient).

Biomarker analysis
Using bead-based immunoassays, plasma levels of 27
cytokines related to the immune system were measured at

Figure 2. Response after ruxolitinib. Illustrative patient (UPN 601004). (A) Figure 3. Response after ruxolitinib. Illustrative patient (UPN 601001).
Positron emission tomography (PET)-computer tomography (CT) frontal view. (B) Comparison of frontal positron emission tomography (PET)-scan prior to inclu-
PET-CT sagittal view. Partial response with allievation of B symptoms and blood sion and after 2 months of ruxolitinib. There was a rapid improvement of con-
inflammation was achieved 2 months after starting ruxolitinib. At month 6, the stitutional symptoms after a few days on ruxolitinib. PET after 2 months showed
patient had slowly progressive disease but refused to stop ruxolitinib. CRP: C- metabolic partial response with a total volume reduction of tumor lung lesions
reactive protein. of 64%.

844 haematologica | 2018; 103(5)


Ruxolitinib in advanced relapsed/refractory HL

baseline and after the first cycle of treatment. At baseline, achieved a partial response as determined by computed
there was no difference in cytokine levels between tomography criteria and also a positron emission tomog-
responders and non-responders. In responders, the only raphy-determined response lasting 4 months. It is note-
cytokine that decreased significantly was CX-CL10 worthy that the PDL1 and PDL2 loci (which are in the
(P=0.01). In patients presenting with pruritus (n=11), the vicinity of the JAK2 locus at 9p24), analyzed by fluores-
levels of platelet-derived growth factor-BB (PDGF-BB) cent in situ hybridization with bacterial artificial chromo-
(Online Supplementary Appendix), interleukin (IL)-5, IL-10, some probes, showed the same pattern of gains as for the
IL-12, IL-13, IL-17, eotaxin, fibroblast growth factor basic JAK2 locus.
(FGF basic), macrophage inflammatory protein 1b
(MIP1b), regulated on activation, normal T-cell expressed
and secreted (RANTES), and vascular endothelial growth Discussion
factor (VEGF) were significantly increased. In the latter
patients, ruxolitinib treatment significantly decreased the JAK/STAT activation, driven by an aberrant network of
levels of PDGF-BB, IL-10, IL-12, IL-13, IL-17, FGF basic cytokines and chemokines in the HL microenvironment,
and VEGF. Among the patients who could be analyzed for is critical for the proliferation and survival of neoplastic
JAK2 amplification in HRS cells (n=12), polysomy (sug- HRS cells.24,25 The JAK/STAT pathway also plays a role in
gesting hyperdiploidy) was detected in all of them, and immune evasion by HL cells via the secretion of
specific JAK2 amplification in only one. This latter patient chemokines leading to Th2 homing or via the regulation

Figure 4. Waterfall plot demonstrating percent


change from baseline in target tumor dimensions
(best response, n=27). Of note, among the 32
patients evaluable for disease response, five had no
end-of-treatment SPD measurements by computer
tomography (CT) scan as planned by protocol
because there were obvious signs of disease progres-
sion. *Persisting positive positron emission tomogra-
phy scan, considered as partial response.

Figure 5. Kaplan-Meier estimate of progression-free


survival in 32 evaluable patients with Hodgkin lym-
phoma receiving ruxolitinib.

haematologica | 2018; 103(5) 845


E. Van Den Neste et al.

of PD-L1/L2 expression, which confers an immune privi- Table 4. Treatment-emergent adverse events.
lege to HRS cells. Chromosome 9p24.1/PD-L1/PD-L2 A. Patients with an adverse event.
alterations increase the abundance of the PD-1 ligands, Treatment-emergent adverse events1 All patients (n=33)
PD-L1 and PD-L2, and their further induction through
JAK/STAT signaling.26-28 This complex crosstalk between Patients with > 1 AE 14 (42.4%)
malignant HRS cells and the reactive microenvironment N. of AE/patient, median (range) 2 (1-11)
N. of patients with AE > grade 3 8 (24.2%)
could be targeted to overcome chemoresistance. Based on
this rationale, we explored JAK 1/2 inhibition in a phase Patients with AE related to ruxolitinib 6 (18.2%)
II study of fixed dose ruxolitinib in patients with Patients with AE leading to drug discontinuation 1 (3%)
advanced HL patients before the onset of the era of PD-1 Patients with AE leading to death 0 (0%)
blockers. With an ORR of 9.4% at the end of the 6-month AE: adverse event. 1Total number of AE, 40.
induction period, this study did not reach its primary effi-
cacy goal. Nevertheless, when including transient B. Characteristics of the adverse event (N = 40) by system organ class
responses seen before the 6-month evaluation, the ORR and preferred terms
was 18.8% in some heavily pretreated patients, most of
whom were refractory and had failed treatment with BV.
Adverse event Any grade Grade 2 Grades > 3
These responses were sometimes durable (median=7.7 Any adverse event 40 15 25
months). Some other patients had disease control, but Infections and infestations 13 10 31
with uncertain clinical benefit. A notable finding to be Anemia 11 0 11
highlighted was the relief of B symptoms and pruritus,
which was quick and long-lasting, resulting in a number Lymphopenia 4 0 4
of patients being reluctant to discontinue the compound, Thrombocytopenia 2 0 2
despite progressive disease. The latter effect should not Weight decrease 1 0 1
be interpreted as a proven surrogate of anti-lymphoma Respiratory and thoracic disorders 3 2 1
activity.
Diarrhea 1 1 0
These results tend to lend some support to the concept
of JAK1/2 inhibition as a potential therapeutic means in Infuenza-like illness 1 1 0
HL. There are presently only scarce data available on the Subdural hematoma 1 0 1
use of ruxolitinib in HL. In a preliminary report of an Bone pain 1 1 0
ongoing study, Kim et al. described rapid achievement of Epilepsy 2 0 2
disease control (1 complete response, 5 partial responses, 1
Implantable device infection, gastro-enteritis, lung infection.
1 stable disease) in 13 patients with advanced HL treated
with ruxolitinib at a dose of 20 mg bid.29 Younes et al.
reported changes in tumor measurements in HL patients
treated in a phase I study with SB1518, an inhibitor of contains the JAK2 gene, are more frequent in advanced
JAK2 and FLT-3.30 In vitro, AZD1480, an inhibitor of JAK1 disease.28 Surprisingly, in our patients, a low incidence of
and JAK2, could regulate proliferation in HL cell lines.27 JAK2 amplification was seen, suggesting a low proportion
The multikinase inhibitor lestaurtinib also inhibited of patients harboring the target of ruxolitinib, although
growth and increased apoptosis of HL cell lines and HL this inference should be considered with caution since
cells from lymph nodes.31 Finally, a clinical grade JAK2 not all patients could be analyzed.
inhibitor, fedratinib, inhibited the proliferation of classi- With respect to safety, ruxolitinib was by and large
cal HL cell lines in a JAK2 copy number-dependent man- well-tolerated, with no drug-related mortality reported.
ner implying decreased phosphorylation of STAT and The most prominent toxicities included drug-related ane-
expression of downstream targets including PD-L1 show- mia and manageable infectious events with no specific
ing immunomodulation by JAK inhibitors.32 pattern. The relative lack of hematologic toxicity suggests
If JAK2 is actually an appropriate target, questions arise that it could be feasible to combine ruxolitinib treatment
as to why the study outcome was not more convincing. with genotoxic compounds. For patients who discontin-
Could the drug's limited activity be attributed to insuffi- ued ruxolitinib therapy, a switch to chemotherapy and/or
cient dosage? Given that we observed unambiguous immunotherapy was feasible, suggesting that the com-
cytokine profile changes and frequent improvements in B pound does not jeopardize further treatment.
symptoms, it would seem that the dosage of 20 mg twice The question now remains as to how this compound
daily, a dosage at which target inhibition occurs in can best be utilized in the future. The exploratory nature
myelofibrosis,33 was appropriate. Another factor possibly of our study did not allow identification of the best can-
influencing the outcome was our patients’ disease stage, didates on the basis of clinical stage or biomarkers. The
represented by a high percentage of refractoriness. At this cytokine profile showed some changes in patients with
late stage, the genetic changes would be so complex that pruritus, but these changes were not correlated with clin-
selective inhibition of JAK is insufficient in cells depend- ical response. Although JAK2 status was explored in a
ent on other signaling pathways to promote their sur- minority of patients, the only patient with JAK2 amplifi-
vival, thus further curbing the study's potential. It is cation achieved a response. It will be important to focus
known that genomic aberrations, such as chromosome on biomarker results in ongoing studies of JAK inhibition
breakpoints, are more numerous in later clinical stages of in HL. Given ruxolitinib’s limited benefits as monothera-
HL.34 Mechanisms of resistance to JAK/STAT inhibition py, use in combination with other drugs may possibly
have been reported such as a feedback loop of paradoxi- enhance its therapeutic potential. Ruxolitinib, which has
cally activated extracellular signal-regulated kinases 1 and no overlapping toxicity with chemotherapy, has been
2 (ERK1/2).27 Aberrations of the 9p24.1 amplicon, which combined with hypomethylating agents, lenalidomide,

846 haematologica | 2018; 103(5)


Ruxolitinib in advanced relapsed/refractory HL

and even intensive chemotherapy.35-38 In vitro data have phase II study of ruxolitinib in R/R HL patients. The
shown that ruxolitinib could restore the sensitivity of cis- study failed to fulfill the efficacy criteria for further devel-
platin-resistant cell lines with higher Jak2 expression.39 opment of the drug as monotherapy. Nonetheless, in
Interestingly, the combination of BV with ruxolitinib patients with very advanced disease ruxolitinib showed
resulted in additive and synergistic killing in a xenograft hints of activity that surpassed solely an anti-inflammato-
mouse model of HL through a mechanism involving ry effect. This may suggest that further improvements
mitochondrial control of apoptosis.40 Another means to will come from a more complete inhibition of signaling
boost ruxolitinib’s potential would be to combine it with pathways involved in HRS cell survival or from combina-
agents blocking other signaling pathways. Interestingly, tion with chemotherapy, such as BV.
the combination of ruxolitinib with a Bcl2/Bcl-xL
inhibitor displayed dramatic synergy in an adult T-cell Acknowledgments
leukemia cell line via a mechanism implying BAX activa- The authors would like to thank the patients, their families and
tion.41 Finally, the effect of combining chemical JAK their caregivers who made this study possible. We also thank all
blockade and an anti-PD1/L1 strategy should be analyzed the study investigators and study staff at each of the clinical sites.
in HL, keeping in mind, however, that a potential antag- For the LYSARC, we acknowledge the project manager and all
onism may be encountered due to these two drugs acting members of the data monitoring committee. We express our
on the same target, given that PD1-L1 expression is thanks to Loïc Chartier and Sami Boussetta, biostatisticians at
dependent on JAK2 activity. the LYSARC, who contributed to the statistical design and analy-
In conclusion, based on a strong biological rationale for sis of the study. Editorial assistance was provided by Cremer
clinical evaluation of JAK2 blockade in HL, we initiated a Consulting.

References Blood. 2009;114(14):2945-2951. ing PRDM16 in myeloid and lymphoid


12. Van Roosbroeck K, Cox L, Tousseyn T, et al. malignancies. PLoS One. 2011;6(10):e26311.
JAK2 rearrangements, including the novel 23. A'Hern RP. Sample size tables for exact sin-
1. Meyer RM, Hoppe RT. Point/counterpoint: SEC31A-JAK2 fusion, are recurrent in classi- gle-stage phase II designs. Stat Med.
early-stage Hodgkin lymphoma and the role cal Hodgkin lymphoma. Blood. 2011;117 2001;20(6):859-866.
of radiation therapy. Hematology Am Soc (15):4056-4064. 24. Aldinucci D, Pinto A, Gloghini A, Carbone
Hematol Educ Program. 2012;2012:313-321. 13. Meier C, Hoeller S, Bourgau C, et al. A. Chemokine receptors as therapeutic tools
2. Horning SJ. Primary refractory Hodgkin's Recurrent numerical aberrations of JAK2 in Hodgkin lymphoma: CCR4 and beyond.
disease. Ann Oncol. 1998; 9 (Suppl 5): S97- and deregulation of the JAK2-STAT cascade Blood. 2010;115(3):746-747; author reply
101. in lymphomas. Mod Pathol. 2009;22(3):476- 748.
3. Kuruvilla J, Keating A, Crump M. How I 487. 25. Carbone A, Gloghini A, Castagna L, Santoro
treat relapsed and refractory Hodgkin lym- 14. Hartmann S, Martin-Subero JI, Gesk S, et al. A, Carlo-Stella C. Primary refractory and
phoma. Blood. 2011;117(16):4208-4217. Detection of genomic imbalances in early-relapsed Hodgkin's lymphoma: strate-
4. Arai S, Fanale M, DeVos S, et al. Defining a microdissected Hodgkin and Reed- gies for therapeutic targeting based on the
Hodgkin lymphoma population for novel Sternberg cells of classical Hodgkin's lym- tumour microenvironment. J Pathol.
therapeutics after relapse from autologous phoma by array-based comparative genom- 2015;237(1):4-13.
hematopoietic cell transplant. Leuk ic hybridization. Haematologica. 2008;93(9): 26. Green MR, Monti S, Rodig SJ, et al.
Lymphoma. 2013;54(11):2531-2533. 1318-1326. Integrative analysis reveals selective 9p24.1
5. Ansell SM. Hodgkin lymphoma: MOPP 15. Joos S, Granzow M, Holtgreve-Grez H, et al. amplification, increased PD-1 ligand expres-
chemotherapy to PD-1 blockade and Hodgkin's lymphoma cell lines are charac- sion, and further induction via JAK2 in
beyond. Am J Hematol. 2016;91(1):109-112. terized by frequent aberrations on chromo- nodular sclerosing Hodgkin lymphoma and
6. Armand P, Shipp MA, Ribrag V, et al. somes 2p and 9p including REL and JAK2. primary mediastinal large B-cell lymphoma.
Programmed death-1 blockade with pem- Int J Cancer. 2003;103(4):489-495. Blood. 2010;116(17):3268-3277.
brolizumab in patients with classical 16. Aldinucci D, Celegato M, Casagrande N. 27. Derenzini E, Lemoine M, Buglio D, et al.
hodgkin lymphoma after brentuximab Microenvironmental interactions in classical The JAK inhibitor AZD1480 regulates prolif-
vedotin failure. J Clin Oncol. 2016 Jun 27. Hodgkin lymphoma and their role in pro- eration and immunity in Hodgkin lym-
[Epub ahead of print]. moting tumor growth, immune escape and phoma. Blood Cancer J. 2011;1(12):e46.
7. Younes A, Bartlett NL, Leonard JP, et al. drug resistance. Cancer Lett. 2016;380(1): 28. Roemer MG, Advani RH, Ligon AH, et al.
Brentuximab vedotin (SGN-35) for relapsed 243-252. PD-L1 and PD-L2 genetic alterations define
CD30-positive lymphomas. N Engl J Med. 17. Assi R, Verstovsek S, Daver N. 'JAK-ing' up classical Hodgkin lymphoma and predict
2010;363(19):1812-1821. the treatment of primary myelofibrosis: outcome. J Clin Oncol. 2016;34(23):2690-
8. Moskowitz C. Novel agents and strategies building better combination strategies. Curr 2697.
in transplant-eligible patients with relapsed Opin Hematol. 2017;24(2):115-124. 29. Kim SJ, Kang HJ, Dong-Yeop S, et al. The
and refractory Hodgkin lymphoma. 18. Vannucchi AM, Harrison CN. Emerging efficacy of JAK2 inhibitor in heavily pre-
Hematology Am Soc Hematol Educ treatments for classical myeloproliferative treated classical Hodgkin lymphoma: a
Program. 2016;2016(1):331-338. neoplasms. Blood. 2017;129(6):693-703. prospective pilot study of ruxolitinib in
9. Slovak ML, Bedell V, Hsu YH, et al. 19. Massaro F, Molica M, Breccia M. How rux- relapsed or refractory classical Hodgkin
Molecular karyotypes of Hodgkin and Reed- olitinib modified the outcome in myelofi- lymphoma and primary mediastinal large
Sternberg cells at disease onset reveal dis- brosis: focus on overall survival, allele bur- B-cell lymphoma. Blood. 2016;128(22):
tinct copy number alterations in chemosen- den reduction and fibrosis changes. Expert 1820.
sitive versus refractory Hodgkin lymphoma. Rev Hematol. 2017;10(2):155-159. 30. Younes A, Romaguera J, Fanale M, et al.
Clin Cancer Res. 2011;17(10):3443-3454. 20. Cheson BD, Pfistner B, Juweid ME, et al. Phase I study of a novel oral Janus kinase 2
10. Aldinucci D, Gloghini A, Pinto A, De Filippi Revised response criteria for malignant lym- inhibitor, SB1518, in patients with relapsed
R, Carbone A. The classical Hodgkin's lym- phoma. J Clin Oncol. 2007;25(5):579-586. lymphoma: evidence of clinical and biologic
phoma microenvironment and its role in 21. Heine A, Brossart P, Wolf D. Ruxolitinib is a activity in multiple lymphoma subtypes. J
promoting tumour growth and immune potent immunosuppressive compound: is it Clin Oncol. 2012;30(33):4161-4167.
escape. J Pathol. 2010;221(3):248-263. time for anti-infective prophylaxis? Blood. 31. Diaz T, Navarro A, Ferrer G, et al.
11. Navarro A, Diaz T, Martinez A, et al. 2013;122(23):3843-3844. Lestaurtinib inhibition of the Jak/STAT sig-
Regulation of JAK2 by miR-135a: prognostic 22. Duhoux FP, Ameye G, Lambot V, et al. naling pathway in Hodgkin lymphoma
impact in classic Hodgkin lymphoma. Refinement of 1p36 alterations not involv- inhibits proliferation and induces apoptosis.

haematologica | 2018; 103(5) 847


E. Van Den Neste et al.

PLoS One. 2011;6(4):e18856. Clinical use of ruxolitinib in an academic F694L mutation in B-precursor acute lym-
32. Hao Y, Chapuy B, Monti S, Sun HH, Rodig medical center in unselected patients with phoblastic leukemia. Pediatr Blood Cancer.
SJ, Shipp MA. Selective JAK2 inhibition myeloproliferative neoplasms not on clini- 2017;64(5).
specifically decreases Hodgkin lymphoma cal study. Leuk Lymphoma. 2017;58(4): 39. Hu Y, Hong Y, Xu Y, Liu P, Guo DH, Chen Y.
and mediastinal large B-cell lymphoma 866-871. Inhibition of the JAK/STAT pathway with
growth in vitro and in vivo. Clin Cancer Res. 36. Daver N, Cortes J, Newberry K, et al. ruxolitinib overcomes cisplatin resistance in
2014;20(10):2674-2683. Ruxolitinib in combination with lenalido- non-small-cell lung cancer NSCLC.
33. Cervantes F, Vannucchi AM, Kiladjian JJ, et mide as therapy for patients with myelofi- Apoptosis. 2014;19(11):1627-1636.
al. Three-year efficacy, safety, and survival brosis. Haematologica. 2015;100(8):1058- 40. Ju W, Zhang M, Wilson KM, et al.
findings from COMFORT-II, a phase 3 1063. Augmented efficacy of brentuximab vedotin
study comparing ruxolitinib with best avail- 37. Devillier R, Raffoux E, Rey J, et al. combined with ruxolitinib and/or
able therapy for myelofibrosis. Blood. Combination therapy with ruxolitinib plus Navitoclax in a murine model of human
2013;122(25):4047-4053. intensive treatment strategy is feasible in Hodgkin's lymphoma. Proc Natl Acad Sci
34. Falzetti D, Crescenzi B, Matteuci C, et al. patients with blast-phase myeloproliferative USA. 2016;113(6):1624-1629.
Genomic instability and recurrent break- neoplasms. Br J Haematol. 2016;172(4):628- 41. Zhang M, Mathews Griner LA, Ju W, et al.
points are main cytogenetic findings in 630. Selective targeting of JAK/STAT signaling is
Hodgkin's disease. Haematologica. 1999;84 38. Mayfield JR, Czuchlewski DR, Gale JM, et potentiated by Bcl-xL blockade in IL-2-
(4):298-305. al. Integration of ruxolitinib into dose-inten- dependent adult T-cell leukemia. Proc Natl
35. Naqvi K, Daver N, Pemmaraju N, et al. sified therapy targeted against a novel JAK2 Acad Sci USA. 2015;112(40):12480-12485.

848 haematologica | 2018; 103(5)


Non-Hodgkin Lymphoma ARTICLE

A B-cell receptor-related gene signature


predicts survival in mantle cell lymphoma: Ferrata Storti
Foundation
results from the Fondazione Italiana Linfomi
MCL-0208 trial
Riccardo Bomben,1* Simone Ferrero,2,3* Tiziana D'Agaro,1 Michele Dal Bo,1
Alessandro Re,4 Andrea Evangelista,5 Angelo Michele Carella,6 Alberto Zamò,7
Umberto Vitolo,8 Paola Omedè,3 Chiara Rusconi,9 Luca Arcaini,10
Luigi Rigacci,11 Stefano Luminari,12,13 Andrea Piccin,14 Delong Liu,15
Adrian Wiestner,15 Gianluca Gaidano,16 Sergio Cortelazzo,17 Marco Ladetto2,18
and Valter Gattei1**
Haematologica 2018
Volume 103(5):849-856
1
Clinical and Experimental Onco-Hematology Unit, Centro di Riferimento Oncologico,
IRCCS, Aviano (PN), Italy; 2Department of Molecular Biotechnologies and Health
Sciences, Hematology Division 1, University of Torino, Italy; 3Hematology Division 1, AOU
“Città della Salute e della Scienza di Torino” University-Hospital, Italy; 4Hematology, AO
“Spedali Civili di Brescia”, Italy; 5Unit of Cancer Epidemiology, AOU “Città della Salute e
della Scienza di Torino” University-Hospital, Italy; 6Hematology Division 1, IRCCS AOU
San Martino IST, Genova, Italy; 7Department of Diagnostics and Public Health, University
of Verona, Italy; 8Hematology Division 1, AOU “Città della Salute e della Scienza di
Torino” University-Hospital, Italy; 9Hematology Division, “Niguarda Ca’ Granda” Hospital,
Milano, Italy; 10Hematology Division, Department of Molecular Medicine, IRCCS
Fondazione San Matteo, Pavia, Italy; 11Hematology Division, AOU “Careggi”, University of
Firenze, Italy; 12Hematology, Azienda Sanitaria Locale IRCCS, Reggio Emilia, Italy;
13
Department of Diagnostic, Clinical and Public Health Medicine, University of Modena
and Reggio Emilia, Reggio Emilia, Italy; 14Department of Hematology and BMT Unit,
Bolzano/Bozen Regional Hospital, Italy; 15Hematology Branch, National Heart, Lung, and
Blood Institute, NIH, Bethesda, MD, USA; 16Division of Haematology, Department of
Translational Medicine –University of Eastern Piedmont, Novara, Italy; 17Hematology,
Medical Oncology and Hematology Division, “Istituto Clinico Humanitas Gavazzeni”,
Bergamo, Italy and 18SC Ematologia Azienda Ospedaliera Nazionale SS. Antonio e Biagio
e Cesare Arrigo, Alessandria, Italy
*RB and SF contributed equally to this work as first authors: **ML and VG contributed equally to
this work as senior authors

Correspondence:
ABSTRACT rbomben@cro.it

M
antle cell lymphoma patients have variable clinical courses,
ranging from indolent cases that do not require immediate treat-
ment to aggressive, rapidly progressing diseases. Thus, diagnos- Received: November 10, 2017.
tic tools capable of stratifying patients according to their risk of relapse Accepted: February 14, 2018.
and death are needed. This study included 83 samples from the
Fondazione Italiana Linfomi MCL-0208 clinical trial. Through gene Pre-published: February 22, 2018.
expression profiling and quantitative real-time PCR we analyzed 46
peripheral blood and 43 formalin-fixed paraffin-embedded lymph node doi:10.3324/haematol.2017.184325
samples. A prediction model to classify patients was developed. By ana-
lyzing the transcriptome of 27 peripheral blood samples, two subgroups Check the online version for the most updated
characterized by a differential expression of genes from the B-cell recep- information on this article, online supplements,
tor pathway (B-cell receptorlow and B-cell receptorhigh) were identified. and information on authorship & disclosures:
The prediction model based on the quantitative real-time PCR values of www.haematologica.org/content/103/5/849
six representative genes (AKT3, BCL2, BTK, CD79B, PIK3CD, and SYK),
was used to classify the 83 cases (43 B-cell receptorlow and 40 B-cell
receptorhigh). The B-cell receptorhigh signature associated with shorter pro- ©2018 Ferrata Storti Foundation
gression-free survival (P=0.0074), selected the mantle cell lymphoma Material published in Haematologica is covered by copyright.
subgroup with the shortest progression-free survival and overall survival All rights are reserved to the Ferrata Storti Foundation. Use of
(P=0.0014 and P=0.029, respectively) in combination with high (>30%) published material is allowed under the following terms and
Ki-67 staining, and was an independent predictor of short progression- conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode.
free survival along with the Mantle Cell Lymphoma International Copies of published material are allowed for personal or inter-
Prognostic Index-combined score. Moreover, the clinical impact of the 6- nal use. Sharing published material for non-commercial pur-
gene signature related to the B-cell receptor pathway identified a mantle poses is subject to the following conditions:
cell lymphoma subset with shorter progression-free survival intervals https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com-
also in an external independent mantle cell lymphoma cohort homoge- mercial purposes is not allowed without permission in writing
nously treated with different schedules. In conclusion, this 6-gene signa- from the publisher.
ture associates with a poor clinical response in the context of the MCL-
0208 clinical trial. (clinicaltrials.gov identifier: 02354313).

Haematologica | 2018; 103(5) 849


R. Bomben et al.

Introduction as the strongest predictor of OS in a large MCL series.20 In


this context, a cohort of 20 proliferation-associated genes
Mantle cell lymphoma (MCL) is a distinctive B-cell constructed on the basis of gene expression analysis was
malignancy accounting for 5-10% of all lymphomas,1-3 demonstrated to be superior to other molecular markers.20
whose molecular hallmark and initiating oncogenic event, Since approaches based on microarray technology have
the t(11;14)(q13;q32) translocation, leads to constitutive not yet been incorporated into routine clinical practice, a
overexpression of the proto-oncogene cyclin D1 PCR-based surrogate method investigating expression of
(CCND1).2,4 five genes has been proposed and applied to paraffin-
Once considered as uniformly characterized by a poor embedded tissues.18
prognosis, MCL has been demonstrated to have unexpect- Recent evidence suggests that the B-cell receptor (BCR)
edly variable clinical courses, ranging from indolent cases pathway may contribute to the pathogenesis of several
that do not require immediate treatment to aggressive, histological types of B-cell non-Hodgkin lymphomas,
rapidly progressing disease.2,5-10 Even among patients including MCL.27-30 The importance of BCR signaling path-
requiring treatment, prognosis is highly heterogeneous, way in B-cell malignancy pathogenesis has driven interest
with patients experiencing prolonged remissions and oth- in the use of small-molecule inhibitors of BCR-associated
ers rapidly relapsing even after cytarabine-containing kinases, potentially preventing the activation of one or
induction regimens followed by autologous transplanta- more of the distal BCR signaling pathway proteins.28,31
tion. Thus, diagnostic tools capable of stratifying MCL In the present study, we developed a survival predictive
patients in different risk classes are warranted in order to model for younger patients with advanced MCL treated in
direct treatment strategies.11 For this reason, many the context of the Fondazione Italiana Linfomi (FIL) MCL-
attempts have been made to identify clinical, histological, 0208 Phase III randomized clinical trial. This model is
and molecular markers that can stratify patients according based upon the quantitative evaluation of six genes, most-
to their risk of relapse and death.12-25 ly from the BCR pathway, selected from a gene expression
In addition to the clinical MCL prognostic score (MCL- profile (GEP) of peripheral blood (PB) MCL cells and was
International Prognostic Index, MIPI)12,14 capable of strati- applied to formalin-fixed paraffin-embedded (FFPE) tissue
fying patients into risk groups with different overall sur- specimens. Notably, the model predicts poor response in
vival (OS),14 the Ki-67 proliferation index has been pro- the context of the FIL-MCL-0208 trial.
posed as one of the most powerful and independent pre-
dictors of survival in MCL even in the context of prospec-
tive trials and modern therapies,5,13,26 and for these reasons Methods
has been integrated into the so-called MIPI-combined
(MIPI-c) score.13,26 Moreover, effective prognostic discrimi- Primary MCL cases
nation is achieved by post-treatment response monitoring The study included 83 out of 300 samples of adult patients
by positron emission tomography (PET)-scan and minimal under 66 years of age with advanced stage MCL, enrolled in the
residual disease (MRD). Furthermore, a seminal study FIL-MCL-0208 prospective, multicenter, Phase III randomized
identified a specific signature associated with proliferation clinical trial (clinicaltrials.gov identifier: 02354313),32 divided as fol-
lows: i) a panel of 27 PB samples utilized for GEP upon positive
sorting of the clonal CD5+/CD19+ MCL cells; ii) an additional
panel of 19 PB samples utilized for quantitative real-time PCR
(qRT-PCR) of the identified gene signature in the purified MCL
Table 1. Characteristics of 83 mantle cell lymphoma cases entering
the study. cell component; iii) a panel of 43 lymph node (LN) samples uti-
lized for qRT-PCR of the identified gene signature (in this LN
Samples
panel 6 samples had a matched PB sample). The clinical and histo-
Number of cases 83 pathological details of the 83 MCL cases used in this study are
Median age, years (range) 56 (28-65) reported in Table 1. No significant differences were found
between the 83 cases entering the study versus the 217 remaining
Ratio male/female (%) 57/26 (68)
cases enrolled in the clinical trial in terms of median age, MIPI
Abnormal LDH (%) 45/29 (39) score, Ki-67 index and PFS intervals (Online Supplementary Table S1
Median WBC (x109/L) 13.7 and Online Supplementary Figure S1). No differences in clinical and
Typical morphology 74 biological parameters were observed between PB and LN MCL
Blastoid morphology 5 samples (data not shown). All patients were treated according to the
FIL-MCL-0208 clinical trial, as reported in Online Supplementary
Median Proliferation Index (Ki-67 staining), % 20.0 (0-99)
Figure S2.
MIPI-c class Mantle cell lymphoma diagnosis was prospectively confirmed
Low 38 (46%) by centralized histological review according to the World Health
Low/intermediate 23 (28%) Organization (WHO) 2008 criteria.3,33 All patients provided
High/intermediate 11 (13%) informed consent in accordance with Institutional Review Board
requirements (0016331-BZ 09/02/2010) and the Declaration of
High 6 (7%)
Helsinki, and protocol consent included use of MRD sample left-
na 5 (6%) overs for the study.
Median survival, months (range) 34.7 (1.4-73.4) All the procedures employed for RNA extraction, GEP and
Median progression-free survival, months (range) 31.3 (1.4-73.4) downstream analyses, qRT-PCR, analyses and qRT-PCR valida-
tions were carried out according to standard protocols, as report-
LDH: lactate dehydrogenase; WBC: white blood count; MIPI-c: MCL:-International
Prognostic Index-combined; na: not available.
ed previously.34-37 (See the Online Supplementary Appendix for
details). Microarray data are available in Gene Expression

850 haematologica | 2018; 103(5)


B-cell receptor signature in mantle cell lymphoma

Omnibus (http://www.ncbi.nlm.nih.gov/geo/) under accession num- 2017.32 Investigators are still blinded to the investigation arm as
ber GSE89447. Cases used for these procedures are reported in the primary study end point has still not been met.
Online Supplementary Table S2.
Results
Validation procedures
The 6-gene signature was tested in the MCL cohort described GEP identifies MCL patients with distinct expression of
by Saba et al.,30 enrolled in another clinical trial (clinicaltrials.gov genes belonging to the BCR pathway
identifier: 00114738), by using the sum of the array gene expression Global GEP was performed in purified MCL cells from
values, as reported.30 Gene signatures related to MCL outcome 27 PB samples. An unsupervised analysis performed by
were retrieved from previous papers,30,38 and imported in the principal component analysis (PCA) divided the cohort
GeneSpring GX and tested in the present cohort with GEP data into two groups of 14 cases and 13 cases, respectively
available. (Figure 1A). Consistently, a hierarchical clustering, which
was run with all the GEP features, split MCL cases into
Statistical analysis two major groups perfectly resembling the PCA groups
Overall survival was computed from trial registration to death (Figure 1B).
as a result of any cause, censored at the latest follow up in patients Supervised analysis according to the PCA classification
who were still alive. Progression-free survival (PFS) was computed defined a gene expression signature composed of 922
from trial registration to progression or death as a result of any probes, 713 up-regulated and 209 down-regulated in
cause, censored at the latest tumor assessment if no progression group-2 versus group-1 samples (Figure 1C and Online
was observed. Clinical correlations, performed with the MedCalc Supplementary Table S3).
v.9.5 software, were made using Kaplan-Meier plots and log-rank Pathway analysis revealed that “Antigen processing and
test. The Cox proportional model was chosen for multivariable presentation” and “B-cell receptor signaling pathway”
analysis. Clinical outcome results were up-dated as of January were among the top ranked pathways enriched in the

A B

Figure 1. Gene Expression Profile (GEP) analysis of 27 mantle cell lym-


phoma samples. (A) Principal Component Analysis (PCA) scores represent-
ed in a 3D scatter plot. One point per array/sample is shown. Black line indi-
cates separation between PCA classes. (B) Hierarchical clustering of 14
group-1 cases and 13 group-2 cases, using 50,739 probes. (C) Hierarchical
clustering of 14 group1 cases and 13 group2 cases, using the 922 differ-
entially expressed probes. Color codes for gene expression values refer to
mean centered log-ratio values.

haematologica | 2018; 103(5) 851


R. Bomben et al.

group-2 category (Online Supplementary Table S4). Similar A 6-gene signature identifies BCRlow and BCRhigh MCL
results were obtained by GSEA which highlighted a con- samples
stitutive overexpression of genes related to the BCR sig- Having identified two different groups of MCL patients
naling pathways in the context of group-2 patients (Figure at diagnosis with a different expression of genes related
2A and Online Supplementary Table S5). Therefore, here- to the BCR pathway, we overlapped the genes included in
after the two PCA groups were identified as BCRlow the gene sets related to the BCR pathway (115 probes)
(group-1) and BCRhigh (group-2). and the differentially expressed genes (922 probes) to cre-

A B

D E

Figure 2. 6-gene signature and Decision Tree (DT) prediction model. (A) Gene Expression Profile data of BCRlow and BCRhigh MCL samples were tested using Gene set
enrichment analysis (GSEA). Reported are the significant gene sets differentially expressed and related to the B-Cell Receptor (BCR) pathway. (B) Venn diagram
derived by merging the differentially expressed probes and the genes belonging to the BCR related gene sets. In bold genes selected as the 6-gene signature. (C)
Hierarchical clustering of 14 BCRlow cases and 13 BCRhigh cases, using the six gene values. Color codes for gene expression values refer to mean centered log-ratio
values. (D) Hierarchical clustering of 8 BCRlow cases and 9 BCRhigh cases belonging to the training set of DT prediction model, using the six gene qRT-PCR values. (E)
Hierarchical clustering of 6 BCRlow cases and 4 BCRhigh cases belonging to the validation set of DT prediction model, using the six gene qRT-PCR values. Bar under the
heat-map refers to prediction generated by the DT prediction model. Color codes for gene expression values refer to mean centered log-ratio values.

852 haematologica | 2018; 103(5)


B-cell receptor signature in mantle cell lymphoma

ate a reduced signature (Figure 2B). In this way, 18 probes (median PFS: 21.6 months vs. not reached; P=0.0375)
corresponding to 15 genes, all over-expressed in BCRhigh (Figure 3).
cases were identified (Figure 2B). Among these genes, a
subgroup of six genes (AKT3, BCL2, BTK, CD79B, Application of the 6-gene signature to LN samples
PIK3CD, and SYK) was selected for further validations from MCL patients
due to their direct involvement in the BCR pathway To evaluate the capability of the 6-gene signature to
and/or the existence of drugs targeting the related pro- identify different subgroups also in the context of MCL
teins. A hierarchical cluster using only these six genes was LN cases, we tested our qRT-PCR approach in a series of
able to discriminate patients belonging to the BCRlow or 43 LN samples preserved as FFPE LN specimens. Thirty-
BCRhigh groups (Figure 2C). five (81%) out of 43 samples were amplifiable for all six
genes, and using a DT model based on qRT-PCR values
Development of a qRT-PCR-based predictor for BCRlow from FFPE, 23 cases were classified as BCRlow and 20 clas-
and BCRhigh in MCL samples sified as BCRhigh (Online Supplementary Table S2). Notably,
By analyzing the expression levels of the selected six for 6 out of 43 LN samples, a PB matched sample was
genes in the same 27 MCL PB samples by qRT-PCR available, and by comparing qRT-PCR results performed
approach, a strict correlation with GEP data was found on PB samples and LN FFPE samples from these cases, a
(Online Supplementary Figure S3B). Moreover, the 27 MCL good concordance was overall observed, although FFPE
cases were randomly divided into a training set (17 cases; samples generally amplified at higher Ct values (Online
8 BCRlow and 9 BCRhigh samples) and a validation set (10 Supplementary Figure S5). Of note, 5 out of 6 these MCL
cases; 6 BCRlow and 4 BCRhigh samples) to develop and test cases were consistently classified. The misclassified case
a decision tree (DT) model based on qRT-PCR data capa-
ble of categorizing patients into one of the two categories.
The DT model based on qRT-PCR data correctly classified
16 of 17 cases belonging to the training set and 10 of 10
cases of the validation cohort, and allowed the classifica-
tion of 19 additional PB samples screened with qRT-PCR
(9 BCRlow and 10 BCRhigh (Figure 2D and E and Online
Supplementary Table S2).

Association between BCR categories and biological


and clinical parameters
Collectively, the 6-gene signature was re-evaluated by
setting up a validated qRT-PCR approach (see Online
Supplementary Appendix and Online Supplementary Table S6)
in PB samples from 46 MCL cases, 23 were identified as
BCRlow and 23 as BCRhigh. By correlating the BCR groups
with the available biological parameters, no association
was found between the 6-gene signature and IGHV gene
status (P=0.93) (Online Supplementary Table S2 and Online
Supplementary Figure S4A), Ki-67 expression, white blood
cells, hemoglobin, lymphocytes, platelets, and neutrophil
count (data not shown). The only significant difference was
between the BCR classification and lactate dehydrogenase
Figure 3. BCRhigh mantle cell lymphoma (MCL) group is associated with a worse
(LDH) levels; BCRhigh cases showed a higher level of LDH
clinical outcome. Kaplan-Meier curves obtained by comparing progression-free
with respect to BCRlow MCL (416.6±191.6 vs. 292.2±127.4;
P=0.023) (Online Supplementary Figure S4B). survival intervals of 23 BCRlow MCL cases with 23 BCRhigh MCL cases. The number
Clinically, MCL patients classified as BCRhigh experi- of patients in each group is reported under relative categories; P refers to log-
rank test.
enced shorter PFS with respect to BCRlow MCL cases

Table 2. Cox regression analysis on mantle cell lymphoma cases.


Univariable Multivariable
Variable HR (95%CI) P HR (95%CI) P
BCR signature
BCR high 2.81 (1.28-6.19) 0.01 3.48 (1.47-8.25) 0.005
MIPI-c
Low/intermediate 1.5 (0.6-3.73) 0.384
High/intermediate 1.41 (0.45-4.43) 0.557
High 3.46 (1.1-10.91) 0.034 4.17 (1.3-13.34) 0.016
Multivariable Cox regression analysis of progression-free survival was performed by including the 6-gene BCR categorization and the mantle cell lymphoma International
Prognostic Index-combined (MIPI-c) score as defined by Hoster et al.13 HR: Hazard Ratio; CI: Confidence Interval; BCR: B-cell receptor.

haematologica | 2018; 103(5) 853


R. Bomben et al.

was considered as BCRlow according to GEP data. Also in cases classified as BCRlow had similar longer PFS intervals
the context of LN samples, no correlation was found irrespective of the high or low Ki-67 score (median PFS:
between the different biological parameters and BCR 20.5 months vs. not reached for all the other combinations;
groups (data not shown). P=0.0014) (Figure 4B). Consistently, multivariable analysis
By merging the MCL cases analyzed either in PB or in carried out by including the BCR signature and the MIPI-c
LN, a total of 83 cases were collected, 43 BCRlow and 40 categories selected the BCRhigh and the high risk MIPI-c cat-
BCRhigh. BCRhigh patients had a shorter PFS with respect to egory as independent predictors of PFS (Table 2). Regarding
BCRlow patients (median PFS: 42.1 months vs. not reached; OS, while the BCR readout failed to identify groups with
P=0.0074) (Figure 4A). Since Ki-67 is a well-known prog- different OS intervals, possibly due to the low rate of
nosticator in MCL,26 we combined the BCR groups with events and short follow up (Figure 4C), the combination of
the prognostic groups defined by Ki-67 score. Cases with high Ki-67 score and a BCRhigh 6-gene signature was able
high Ki-67 (≥30% of Ki-67 expressing cells) and classified again to select the MCL subgroup with the shortest OS
in the BCRhigh group experienced the shortest PFS, while (46.7 vs. not reached; P=0.029) (Figure 4D).

A B

Figure 4. BCRhigh mantle cell


lymphoma (MCL) group is
associated with a worse clini-
cal outcome (overall series).
C (A) Kaplan-Meier curves
obtained by comparing pro-
D
gression-free survival (PFS)
intervals of 43 BCRlow MCL
cases with 40 BCRhigh MCL
cases. (B) Kaplan-Meier
curves obtained by compar-
ing PFS intervals of 19 BCRlow
and low Ki-67 MCL cases,
with 20 BCRlow and high Ki-67
MCL cases, with 21 BCRhigh
and Ki-67 low MCL cases,
with 10 BCRhigh and Ki-67 high
MCL cases. (C) Kaplan-Meier
curves obtained by compar-
ing overall survival (OS) inter-
vals of 43 BCRlow MCL cases
with 40 BCRhigh MCL cases.
(D) Kaplan-Meier curves
obtained by comparing OS
intervals of 19 BCRlow and low
Ki-67 MCL cases, with 20
BCRlow and high Ki-67 MCL
cases, with 21 BCRhigh and Ki-
67 low MCL cases, with 10
BCRhigh and Ki-67 high MCL
cases. The number of
patients in each group is
reported under relative cate-
gories; P refers to log-rank
test.

854 haematologica | 2018; 103(5)


B-cell receptor signature in mantle cell lymphoma

Validations of BCR signature expression of the selected six genes.28 This DT model was
To verify whether the BCR signature maintained its applied in an independent cohort of PB samples and then
prognostic impact in an independent set of patients, we to a further series of FFPE LN samples, thus demonstrating
used the gene expression data of MCL LN biopsies report- that two MCL subsets with different expression levels of
ed by Saba et al.30 Also in this different setting, a high BCR-related genes could also be recognized in the LN
expression of the 6-gene signature, as in the context of compartment, mirroring PB. Taken together, by combin-
BCRhigh cases, identified an MCL patient subset with infe- ing data from the PB and LN compartments, MCL cases
rior PFS (P=0.049) (Online Supplementary Figure S6). classified as BCRhigh showed higher LDH levels and shorter
In another set of analyses, by taking advantage of our 27 PFS with respect to BCRlow patients, suggesting that activa-
MCL cases with GEP data available, we correlated our tion of BCR signaling drives tumor proliferation and deter-
BCR signature with other MCL signatures with proven mines clinical outcome of MCL patients, which is in keep-
clinical impact.30,38 As reported in Online Supplementary ing with recent findings.30
Figure S7A, the BCR signature reported in Saba et al.30 By combining the predictive capacity of the 6-gene BCR
divided MCL cases in two groups that corresponded signature with the Ki-67 index, we identified a particularly
exactly to our BCR definition (Online Supplementary Figure unfavorable category (BCRhigh and high Ki-67) with a sub-
S7B).30 Similarly, the 17 genes of the proliferation signa- stantially shorter PFS and OS than the other groups.
ture reported by Scott et al.38 split our MCL cases in 3 dif- Consistently, the BCRhigh signature turned out to be an
ferent groups resembling the 3 different groups originally independent prognosticator along with the high-risk
defined (Online Supplementary Figure S8A). In this context, MIPI-c category for short PFS by multivariate analysis.
the shortest PFS and OS intervals were observed in the There is no indication that the validity of the model may
third group characterized by a higher expression of genes be affected by the different recruitment site (PB vs. LN), or
related to proliferation and a BCRhigh phenotype in keeping by different sample storage (frozen vs. FFPE) because the
with our findings (Online Supplementary Figure S8A-C). main clinical parameters were equally distributed
between the different series (PB/frozen vs. LN/FFPE) (R
Bomben et al., 2018, unpublished observation). In this regard,
Discussion an important feature of this model/assay is its applicabili-
ty to both PB and LN FFPE samples, having, therefore, the
In this study, we demonstrated that a BCR-derived sig- chance to combine results of qRT-PCR with Ki-67 staining
nature based on the differential expression of six genes in all the cases.
correlated with shorter PFS intervals in the context of a Our data underscore the increasing importance of BCR-
Phase III prospective clinical trial (FIL-MCL-0208) for related genes in the pathogenesis and development of
younger MCL patients receiving R-CHOP induction, fol- MCL, further underlined by the clinical significance of
lowed by high-dose cytarabine and autologous stem cell drugs specifically targeting genes belonging to this path-
transplantation (clinicaltrials.gov identifier: 023541313).32 way. In particular, therapeutic targeting of BTK41 can be
Notably, the BCR-related 6-gene signature reported here rationally exploited in lymphoid malignancies that have
was able to identify an MCL subset with shorter PFS inter- been proved to be dependent on an antigen-dependent
vals also in the context of an external independent MCL BCR-mediated active signaling. However, despite the rel-
cohort homogenously treated with different schedules.30 atively high response rate to single agent ibrutinib in
On the other hand, when the signature described by Saba relapsed/refractory MCL, it remained unclear as to why
et al.30 and Scott et al.38 was applied to our MCL cases, the some patients showed clear responses, while others
patient subsets with the worse prognosis turned out to be received little therapeutic benefit.31,42 The BCR-related sig-
particularly enriched in BCRhigh cases, even though these nature described here may provide insights into molecular
signatures did not include any gene from our signature. factors that explain the divergent responses of MCL
Therefore, although composed of genes located upstream patients to ibrutinib, although other causes of primary
of the BCR machinery, our signature was able to identify resistance might be related to gene mutations in the other
cases with an active BCR pathway as defined by other sig- pathways, e.g. NF-κB pathway and epigenetic modifiers,
natures.30 In this regard, however, experiments with pri- as recently reported.43,44
mary MCL cases and/or MCL cell lines combining BCR In conclusion, in the present study we developed a sur-
stimulation with the use of specific BCR inhibitors should vival model for patients with MCL composed of six genes
be performed to investigate the contribution of the 6-gene (AKT3, BTK, CD79B, PIK3CD, SYK, BCL2) whose expres-
signatures described here to the actual activation of the sion can easily be investigated by qRT-PCR and also in
BCR pathway. FFPE specimens. The signature was associated with a poor
Again in agreement with this line of reasoning, BCRhigh clinical response in the context of a high-dose chemo-
samples presented a significant upregulation of PAX5 (see immunotherapy regimen, and might, therefore, be con-
GEP data in Online Supplementary Table S3), a gene whose sidered for validation and application in future clinical tri-
product is known to prevent plasma cell differentiation als.
thus preserving the capacity to respond to antigen-
induced activation and proliferation.39 Taken together Acknowledgments
these data corroborate recent findings of ongoing active The authors would like to thank Progetto Giovani Ricercatori
BCR signaling in MCL cell in vivo,29,30 and further underline GR-2011-02347441, GR-2009-1475467, and GR-2011-
the role of antigen stimulation in the ontogeny of MCL, as 02351370, Ministero della Salute, Rome, Italy; Progetto Ricerca
suggested by the skewed IGVH gene repertoire found in Finalizzata RF-2009-1469205, and RF-2010-2307262,
MCL cells.40 Ministero della Salute, Rome, Italy; Associazione Italiana contro
In order to discriminate between BCRlow and BCRhigh le Leucemie, linfomi e mielomi (AIL), Venezia Section,
MCL samples, we developed a DT model based on the Pramaggiore Group, Italy; Associazione Italiana Ricerca Cancro

haematologica | 2018; 103(5) 855


R. Bomben et al.

(AIRC), Investigator Grant IG-2015 (17622); “5x1000 Ricerca Locale, Università degli Studi di Torino, Italy;
Intramural Program”, Centro di Riferimento Oncologico, Aviano, Fondazione Neoplasie Del Sangue (Fo.Ne.Sa), Torino, Italy;
Italy; Provincia Autonoma di Bolzano/Bozen, Italy; A.O. S. CRT 2015.1044, Fondazione CRT, Torino, Italy. We are grate-
Maurizio, Bolzano/Bozen, Italy; Progetto di Rilevante Interesse ful to all the Clinical Investigators, to the Pathologists, to Luigia
Nazionale (PRIN2009) 7.07.02.60 AE01, Ministero Italiano Monitillo, Daniela Barbero, Marina Ruggeri, Paola Ghione, and
dell'Università e della Ricerca (MIUR), Roma, Italy; Fondi di Gian Maria Zaccaria.

17. Katzenberger T, Petzoldt C, Holler S, et al. and canonical NF-kappaB activation in man-
References The Ki67 proliferation index is a quantita- tle cell lymphoma. Blood. 2016; 128(1):82-
tive indicator of clinical risk in mantle cell 92.
1. Dreyling M, Ferrero S, Hermine O. How to lymphoma. Blood. 2006;107(8):3407. 31. Wang ML, Rule S, Martin P, et al. Targeting
manage mantle cell lymphoma. Leukemia. 18. Hartmann E, Fernandez V, Moreno V, et al. BTK with ibrutinib in relapsed or refractory
2014;28(11):2117-2130. Five-gene model to predict survival in man- mantle-cell lymphoma. N Engl J Med.
2. Cheah CY, Seymour JF, Wang ML. Mantle tle-cell lymphoma using frozen or formalin- 2013;369(6):507-516.
Cell Lymphoma. J Clin Oncol. 2016; fixed, paraffin-embedded tissue. J Clin 32. Cortellazzo S, Martelli M, Ladetto M,
34(11):1256-1269. Oncol. 2008;26(30):4966-4972. Ferrero S, Ciccone G, Evangelista A, et al.
3. Campo E, Swerdlow SH, Harris NL, Pileri S, 19. Ek S, Bjorck E, Porwit-MacDonald A, High dose sequential chemotherapy with
Stein H, Jaffe ES. The 2008 WHO classifica- Nordenskjold M, Borrebaeck CA. Increased rituximab and ASCT as first line therapy in
tion of lymphoid neoplasms and beyond: expression of Ki-67 in mantle cell lym- adult MCL patients: clinical and molecular
evolving concepts and practical applications. phoma is associated with de-regulation of response of the MCL-0208 trial, a FIL study.
Blood. 2011;117(19):5019-5032. several cell cycle regulatory components, as Haematologica. 2015;100(s1):3.
4. Jares P, Colomer D, Campo E. Molecular identified by global gene expression analy- 33. Swerdlow SH, Campo E, Pileri SA, et al. The
pathogenesis of mantle cell lymphoma. J sis. Haematologica. 2004;89(6):686-695. 2016 revision of the World Health
Clin Invest. 2012;122(10):3416-3423. 20. Rosenwald A, Wright G, Wiestner A, et al. Organization (WHO) classification of lym-
5. Dreyling M, Ferrero S, Vogt N, Klapper W. The proliferation gene expression signature phoid neoplasms. Blood. 2016;
New paradigms in mantle cell lymphoma: is is a quantitative integrator of oncogenic 127(20):2375-2390.
it time to risk-stratify treatment based on events that predicts survival in mantle cell 34. Bomben R, Gobessi S, Dal BM, et al. The
the proliferative signature? Clin Cancer Res. lymphoma. Cancer Cell. 2003;3(2):185-197. miR-17-92 family regulates the response to
2014;20(20):5194-5206. 21. Mozos A, Royo C, Hartmann E, et al. Toll-like receptor 9 triggering of CLL cells
6. Ghielmini M, Zucca E. How I treat mantle SOX11 expression is highly specific for with unmutated IGHV genes. Leukemia.
cell lymphoma. Blood. 2009;114(8):1469- mantle cell lymphoma and identifies the 2012;26(7):1584-1593.
1476. cyclin D1-negative subtype. Haematologica. 35. Dal Bo M, D'Agaro T, Gobessi S, et al. The
7. Herrmann A, Hoster E, Zwingers T, et al. 2009;94(11):1555-1562. SIRT1/TP53 axis is activated upon B-cell
Improvement of overall survival in advanced 22. Navarro A, Clot G, Royo C, et al. Molecular receptor triggering via miR-132 up-regula-
stage mantle cell lymphoma. J Clin Oncol. subsets of mantle cell lymphoma defined by tion in chronic lymphocytic leukemia cells.
2009;27(4):511-518. the IGHV mutational status and SOX11 Oncotarget. 2015;6(22):19102-19117.
8. Zucca E, Roggero E, Pinotti G, et al. Patterns expression have distinct biologic and clinical 36. Draghici S, Khatri P, Martins RP, Ostermeier
of survival in mantle cell lymphoma. Ann features. Cancer Res. 2012;72(20):5307- GC, Krawetz SA. Global functional profiling
Oncol. 1995;6(3):257-262. 5316. of gene expression. Genomics.
9. Barista I, Romaguera JE, Cabanillas F. 23. Majlis A, Pugh WC, Rodriguez MA, 2003;81(2):98-104.
Mantle-cell lymphoma. Lancet Oncol. Benedict WF, Cabanillas F. Mantle cell lym- 37. Subramanian A, Tamayo P, Mootha VK, et
2001;2(3):141-148. phoma: correlation of clinical outcome and al. Gene set enrichment analysis: a knowl-
10. Martin P, Chadburn A, Christos P, et al. biologic features with three histologic vari- edge-based approach for interpreting
Outcome of deferred initial therapy in man- ants. J Clin Oncol. 1997;15(4):1664-1671. genome-wide expression profiles. Proc Natl
tle-cell lymphoma. J Clin Oncol. 2009; 24. Wiestner A, Tehrani M, Chiorazzi M, et al. Acad Sci USA. 2005;102(43):15545-15550.
27(8):1209-1213. Point mutations and genomic deletions in 38. Scott DW, Abrisqueta P, Wright GW et al.
11. Dreyling M, Ferrero S. The role of targeted CCND1 create stable truncated cyclin D1 New Molecular Assay for the Proliferation
treatment in mantle cell lymphoma: is trans- mRNAs that are associated with increased Signature in Mantle Cell Lymphoma
plant dead or alive? Haematologica. proliferation rate and shorter survival. Applicable to Formalin-Fixed Paraffin-
2016;101(2):104-114. Blood. 2007;109(11):4599-4606. Embedded Biopsies. J Clin Oncol. 2017;
12. Geisler CH, Kolstad A, Laurell A, et al. The 25. Jares P, Colomer D, Campo E. Genetic and 35(15):1668-1677.
Mantle Cell Lymphoma International molecular pathogenesis of mantle cell lym- 39. Nera KP, Kohonen P, Narvi E, et al. Loss of
Prognostic Index (MIPI) is superior to the phoma: perspectives for new targeted thera- Pax5 promotes plasma cell differentiation.
International Prognostic Index (IPI) in pre- peutics. Nat Rev Cancer. 2007;7(10):750- Immunity. 2006;24(3):283-293.
dicting survival following intensive first-line 762. 40. Hadzidimitriou A, Agathangelidis A,
immunochemotherapy and autologous 26. Determann O, Hoster E, Ott G, et al. Ki-67 Darzentas N, et al. Is there a role for antigen
stem cell transplantation (ASCT). Blood. predicts outcome in advanced-stage mantle selection in mantle cell lymphoma?
2010;115(8):1530-1533. cell lymphoma patients treated with anti- Immunogenetic support from a series of 807
13. Hoster E, Rosenwald A, Berger F, et al. CD20 immunochemotherapy: results from cases. Blood. 2011;118(11):3088-3095.
Prognostic Value of Ki-67 Index, Cytology, randomized trials of the European MCL 41. Rickert RC. New insights into pre-BCR and
and Growth Pattern in Mantle-Cell Network and the German Low Grade BCR signalling with relevance to B cell
Lymphoma: Results From Randomized Lymphoma Study Group. Blood. 2008; malignancies. Nat Rev Immunol. 2013;
Trials of the European Mantle Cell 111(4):2385-2387. 13(8):578-591.
Lymphoma Network. J Clin Oncol. 2016; 27. Perez-Galan P, Dreyling M, Wiestner A. 42. Wang ML, Blum KA, Martin P, et al. Long-
34(12):1386-1396. Mantle cell lymphoma: biology, pathogene- term follow-up of MCL patients treated
14. Hoster E, Dreyling M, Klapper W, et al. A sis, and the molecular basis of treatment in with single-agent ibrutinib: updated safety
new prognostic index (MIPI) for patients the genomic era. Blood. 2011;117(1):26-38. and efficacy results. Blood. 2015; 126(6):739-
with advanced-stage mantle cell lymphoma. 28. Young RM, Staudt LM. Targeting pathologi- 745.
Blood. 2008;111(2):558-565. cal B cell receptor signalling in lymphoid 43. Rahal R, Frick M, Romero R, et al.
15. Bea S, Valdes-Mas R, Navarro A, et al. malignancies. Nat Rev Drug Discov. 2013; Pharmacological and genomic profiling iden-
Landscape of somatic mutations and clonal 12(3):229-243. tifies NF-kappaB-targeted treatment strate-
evolution in mantle cell lymphoma. Proc 29. Akhter A, Street L, Ghosh S, et al. gies for mantle cell lymphoma. Nat Med.
Natl Acad Sci USA. 2013;110(45):18250- Concomitant high expression of Toll-like 2014;20(1):87-92.
18255. receptor (TLR) and B-cell receptor (BCR) sig- 44. Lenz G, Balasubramanian S, Goldberg J,
16. Zhang J, Jima D, Moffitt AB, et al. The nalling molecules has clinical implications in Rizo A, Schaffer M, Phelps C, et al.
genomic landscape of mantle cell lymphoma mantle cell lymphoma. Hematol Oncol. Sequence variants in patients with primary
is related to the epigenetically determined 2015;35(1):79-86. and acquired resistance to ibrutinib in the
chromatin state of normal B cells. Blood. 30. Saba NS, Liu D, Herman SE, et al. phase 3 MCL3001 (RAY) trial.
2014;123(19):2988-2996. Pathogenic role of B-cell receptor signaling Haematologica. 2016;101(s1):155.

856 haematologica | 2018; 103(5)


Non-Hodgkin Lymphoma ARTICLE

Incidence and risk factors for relapses in


HIV-associated non-Hodgkin lymphoma as Ferrata Storti
Foundation
observed in the German HIV-related lymphoma
cohort study
Philipp Schommers,1,2,* Daniel Gillor,1,* Marcus Hentrich,3 Christoph Wyen,1,4
Timo Wolf,5 Mark Oette,6 Alexander Zoufaly,7 Jan-Christian Wasmuth,8
Johannes R. Bogner,9 Markus Müller,10 Stefan Esser,11 Alisa Schleicher,12
Björn Jensen,13 Albrecht Stoehr,14 Georg Behrens,15,16 Alexander Schultze,17
Haematologica 2018
Jan Siehl,18 Jan Thoden,19 Ninon Taylor20 and Christian Hoffmann12,21
Volume 103(5):857-864

1
Department I of Internal Medicine, University Hospital Cologne, Germany; 2German
Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Germany; 3Department
of Medicine III, Red Cross Hospital Munich, Germany; 4Praxis am Ebertplatz, Cologne,
Germany; 5Department of Medicine II, University of Frankfurt, Germany; 6Department of
General Medicine, Gastroenterology and Infectious Diseases, Augustinerinnen Hospital,
Cologne, Germany; 7Department of Medicine IV, Kaiser Franz Josef Hospital, Vienna,
Austria; 8Department of Internal Medicine I, University of Bonn, Germany; 9Department
of Medicine IV, University of Munich, Munich, Germany; 10Department of Infectious
Diseases, Vivantes Auguste-Viktoria- Hospital, Berlin, Germany; 11Department of
Dermatology, University Hospital Essen, Germany; 12University of Schleswig Holstein,
Campus Kiel, Kiel, Germany; 13Department of Gastroenterology, Hepatology and
Infectious Diseases, Düsseldorf University Hospital, Germany; 14Ifi-Institute for
Interdisciplinary Medicine, Hamburg, Germany; 15Department of Clinical Immunology
and Rheumatology, Hannover Medical School, Germany; 16German Center for Infection
Research (DZIF), Hannover, Germany; 17Department of Emergency Medicine, University
Medical Center Hamburg-Eppendorf, Hamburg, Germany; 18Ärzteforum Seestraße,
Berlin, Germany; 19Medical Group Practice for Internal Medicine and Rheumatology,
Freiburg, Germany; 20Department of Internal Medicine III with Hematology, Medical
Oncology, Hemostaseology, Infectious Diseases, Rheumatology, Oncologic Center,
Laboratory of Immunological and Molecular Cancer Research, Paracelsus Medical
University Salzburg, Austria and 21IPM Study Center, Hamburg, Germany
*PS and DG contributed equally to this work.
Correspondence:
philipp.schommers@uk-koeln.de
ABSTRACT

O
utcome of HIV-infected patients with AIDS-related lymphomas
Received: September 17, 2017.
has improved during recent years. However, data on incidence,
risk factors, and outcome of relapses in AIDS-related lym- Accepted: February 2, 2018.
phomas after achieving complete remission are still limited. This Pre-published: February 8, 2018.
prospective observational multicenter study includes HIV-infected
patients with biopsy- or cytology-proven malignant lymphomas since
2005. Data on HIV infection and lymphoma characteristics, treatment doi:10.3324/haematol.2017.180893
and outcome were recorded. For this analysis, AIDS-related lymphomas
patients in complete remission were analyzed in terms of their relapse- Check the online version for the most updated
information on this article, online supplements,
free survival and potential risk factors for relapses. In total, 254 of 399 and information on authorship & disclosures:
(63.7%) patients with AIDS-related lymphomas reached a complete www.haematologica.org/content/103/5/857
remission with their first-line chemotherapy. After a median follow up
of 4.6 years, 5-year overall survival of the 254 patients was 87.8%
(Standard Error 3.1%). Twenty-nine patients relapsed (11.4%). Several ©2018 Ferrata Storti Foundation
factors were independently associated with a higher relapse rate, includ- Material published in Haematologica is covered by copyright.
ing an unclassifiable histology, a stage III or IV according to the Ann All rights are reserved to the Ferrata Storti Foundation. Use of
Arbor Staging System, no concomitant combined antiretroviral therapy published material is allowed under the following terms and
conditions:
during chemotherapy and R-CHOP-based compared to more intensive https://creativecommons.org/licenses/by-nc/4.0/legalcode.
chemotherapy regimens in Burkitt lymphomas. In conclusion, complete Copies of published material are allowed for personal or inter-
remission and relapse rates observed in our study are similar to those nal use. Sharing published material for non-commercial pur-
reported in HIV-negative non-Hodgkin lymphomas. These data provide poses is subject to the following conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
further evidence for the use of concomitant combined antiretroviral ther- sect. 3. Reproducing and sharing published material for com-
apy during chemotherapy and a benefit from more intensive chemother- mercial purposes is not allowed without permission in writing
apy regimens in Burkitt lymphomas. Modifications to the chemotherapy from the publisher.
regimen appear to have only a limited impact on relapse rate.

Haematologica | 2018; 103(5) 857


P. Schommers et al.

Introduction apy were classified as “cycle reduction” and the treatment dura-
tion was calculated according to the number of cycles given. If the
Over the last two decades, the incidence of AIDS-relat- dose of any chemotherapy drug was reduced by 20% or more,
ed lymphomas (ARL) has markedly declined due to the treatment intensity was considered to be reduced.
introduction of combination antiretroviral therapy As positron emission tomography (PET) scans were not routine-
(cART). However, ARL remain a major cause of morbidity ly performed, the 1999 standardized response criteria for non-
and mortality, and represent the highest proportion of all Hodgkin’s lymphomas13 were used rather than the 2007 criteria.14
AIDS-related deaths.1 Patients with ARL are usually treat- CR was defined as the disappearance of all disease manifestations
ed with the same chemotherapy protocols established in for at least three months. This definition also includes uncertain
the HIV-negative setting,2 and the rates of complete remis- complete remission (CRu) that implied a residual mass of 1.5 cm
sion (CR) achieved are comparable to those reported in or smaller that remained unchanged over at least three months.
their HIV-negative counterparts.3,4 However, available data
on the incidence and potential risk factors of recurrent dis- Statistical analysis
ease in ARL are scarce, and treatment of disease relapse Statistical analyses were performed using IBM SPSS Statistics
remains challenging.4,5 In recent studies on HIV-negative software (IBM, Armonk, NY, USA), v.24.0. Univariate statistics
patients with diffuse large B-cell lymphoma (DLBCL), R- were performed using Pearson’s χ², Fisher’s exact one-way
CHOP-based regimens (rituximab, cyclophosphamide, Analysis of Variance (ANOVA) with Bonferroni-corrected post-
adriamycin, vincristine, and prednisone) resulted in CR hoc test, or Kruskal-Wallis test depending on data. For the multi-
rates of around 65-80%. Differences in response rates variate Cox regression analysis, continuous clinically meaningful
largely depend on the pre-treatment International breakpoints that showed P-values below 0.1 in the univariate
Prognostic Index (IPI) for aggressive lymphomas.6,7 In analysis were considered. Kaplan-Meier curves were used to illus-
patients who had achieved a CR, relapse rates ranged trate the relapse-free survival (RFS) and overall survival.
from 6-10%.6,8 The second most common ARL are Burkitt Differences between subgroups were assessed with the log-rank
or Burkitt-like lymphomas (BL).9,10 In the HIV-negative set- test. RFS was defined as the period between first diagnosis and
ting, CR and overall survival (OS) rates of around 80-90% any lymphoma relapse according to the STEEP criteria.15 OS was
were reported by different groups.10-12 In a large prospec- defined as the period between first diagnosis and death from any
tive trial on short-intensive chemotherapy combined with cause. All-cause deaths as well as “lost to follow up” were cen-
rituximab for patients with BL, the relapse rate was 12%.10 sored. All P-values were two-sided. P<0.05 was considered statis-
Although this approach also proved feasible in HIV-relat- tically significant.
ed BL,9 it remains unclear whether relapse rates reported
in HIV-negative DLBCL and BL are different to those in
ARL. Thus, we investigated the risk factors and incidence Results
of relapse in a large cohort of ARL patients who had
achieved a CR after first-line treatment. Patients' characteristics and outcome
Numbers and characteristics of patients included in the
present analysis are depicted in Figure 1. In total, 254 of
Methods 399 (63.7%) patients with high-grade NHL of B-cell origin
(classified as ARL) reached a CR with their first-line
Study design chemotherapy. Of those, 127 had DLBCL, 91 BL, 29 PBL,
The German HIV Lymphoma Cohort is an ongoing, prospective and 7 ARL, not further classified. ARL was CD20-negative
observational multicenter study including all adult HIV infected in 24 of 254 cases (9.5%), among them 22 PBL and 2
patients who are diagnosed with biopsy- or cytology-proven DLBCL cases. Among 22 PBL cases with information on
malignant lymphoma in 33 participating centers since January Epstein-Barr-Virus (EBV) status, EBV was present in 15
2005. Data on HIV-infection and lymphoma characteristics, treat- (68%). Overall, 86.2% of patients with CD20+ lym-
ment and outcome are recorded. From the time of lymphoma phomas received rituximab. Notably, patients diagnosed
diagnosis, patients are followed every six months. Ethics approval before 2010 were less frequently treated with rituximab
was obtained from the ethics committees of the University of than those diagnosed from 2010 onwards (79% vs. 96%,
Cologne (IRC Cologne: 05-174), Germany, and written informed P<0.001). Further, 73% of patients with CD4 cell counts
consent was given by each participating patient. less than 50/µl received rituximab compared to 88% with
The present analysis includes only patients with aggressive B- CD4 counts 50/mL or over (P=0.096). Patients' character-
cell lymphoma in first CR. Lymphomas were grouped in DLBCL, istics with respect to treatment outcomes are listed in
BL, plasmablastic lymphoma (PBL) and ARL, not further classifi- Table 1. OS of patients who achieved CR with first-line
able, the latter group representing aggressive B-cell non-Hodgkin therapy was significantly better than that of patients in
lymphomas (B-NHL) that could not be classified into any subtype. other response groups (Figure 2). After a median follow up
To study the impact of chemotherapy dose intensity on the risk of of 4.6 years, 5-year OS of the entire group of all 262
relapse in patients treated with either R-CHOP-based regimens or patients in first CR was 87.1% [standard error (SE) 2.3%]
the short intensive GMALL protocol,10 we performed an analysis (Figure 3A) with differences between lymphoma sub-
of dose reductions and delays in chemotherapy cycles. types: 87.8% (SE 3.1%) in DLBCL, 87.6% (SE 3.7%) in BL,
(Information about the GMALL and R-CHOP protocol can be 79.6% (SE 11.3%) in PBL, and 83.3% (SE 15.2%) in ARL,
found in the Online Supplementary Tables S1 and S2, respectively). not further classified (P=0.994) (Figure 3B).
A full-intensity treatment was considered to consist of six cycles
of chemotherapy according to the R-CHOP or GMALL protocol Incidence of recurrent disease in ARL
administered at 3-week intervals without dose reductions and After a median follow up of 4.6 years, a relapse of the
within a period of 120 days (5x21 days for 6 cycles plus a maxi- ARL had occurred in 29 of 254 patients (11.4%). Relapses
mum of 3 days delay per cycle). Less than 6 cycles of chemother- were observed in 14 patients with DLBCL (11.0%), 9 with

858 haematologica | 2018; 103(5)


Relapses in AIDS-related lymphomas

BL (9.9 %), 3 with PBL (11.5%) and 3 with ARL, not fur- (P=0.884) (Figure 3F), although the number of patients
ther classified (42.9%), after a median follow up of 5.0, with subtypes other than BL was very small in this analy-
4.6, 3.5 and 6.0 years, respectively. Isolated central nerv- sis. Of note, patients with BL who received the GMALL
ous system (CNS) relapses were observed in 3 of 29 protocol had a significantly better 5yRFS than those
patients (DLBCL: n=2; BL: n=1). RFS depicted by Kaplan receiving R-CHOP-based protocols [94.2% (SE 2.8%) vs.
Meier curves is shown in Figure 3C and D. Five-year RFS 65.5% (SE 12.6%); P=0.001].
(5yRFS) was 88.4% (SE 2.9%) in DLBCL, 88.9% (SE 3.5%)
in BL, and 88.6% (SE 6.2%) in PBL. By contrast, 5yRFS Risk factors for recurrent disease in ARL
was lower in patients with ARL, not further classified Univariate analysis identified several factors associated
[57.1% (SE 18.7%); P=0.057) (Figure 3D). with a lower risk for ARL relapse such as a low IPI, stage
Among patients who achieved CR with first-line R- I or II according to the Ann Arbor Staging System, cART
CHOP-based protocols, 5yRFS was 87.8% (SE 3.1%) and given during chemotherapy, CD4 T-cell counts
84.4% (SE 8.3%) in DLBCL and PBL, respectively, as com- >200x109/L, pathology other than ARL, not further classi-
pared to 65.5% (SE 12.6) in BL and 40.0% in ARL, not fur- fied, and chemotherapy according to the GMALL-proto-
ther classified (SE 21.9%; P=0.005) (Figure 3E). No signifi- col (Table 2). These factors were analyzed in a multivari-
cant differences in 5yRFS between ARL subtypes were ate Cox proportional hazards model, with backward step-
observed in patients treated with the GMALL protocol wise elimination based on a Wald statistic with P≤0.1.

Figure 1. Flow chart of patients included in the present analysis. NHL: non-
Hodgkin lymphoma; T-NHL: T-cell non-Hodgkin lymphoma; CR: complete remis-
sion; BL: Burkitt lymphoma; DLBCL: diffuse large B-cell lymphoma; PBL: plas-
mablastic lymphoma; ARL: AIDS-related lymphoma.

haematologica | 2018; 103(5) 859


P. Schommers et al.

After two elimination steps, histology [BL: Hazard ratio Dose intensity of chemotherapy
(HR) 2.60 95% Confidence Interval (95%CI): 0.92 – 7.4, Since chemotherapy regimen (R-CHOP or GMALL)
PBL: HR 1.28 95% CI 0.36-4.57, ARL, not further classi- seems to be critical for RFS, we investigated how many
fied: HR 5.08 95% CI 1.13 – 22.90, indicator = DLBCL), patients of all aggressive B-NHL had any kind of reduc-
stage III or IV according to the Ann Arbor Staging System tion (either in the number of chemotherapy cycles or in
(HR 4.85 95%CI 1.44 – 16.34), no concomitant cART (HR the treatment intensity) or a delay during their treatment.
4.28 95%CI 1.19 – 15.39) and use of R-CHOP (HR: 7.59 Results of dose intensity analysis are shown in Online
95%CI 1.87 – 30.81) remained in the model (Table 2). A Supplementary Table S3. Overall, 32.7% of the patients
higher IPI was no longer predictive anymore in the multi- had a treatment delay, 13.8% had dose reductions, and
variate model. 16.5% had reduced numbers of chemotherapy cycles

Table 1. Patients’ characteristics based on their treatment outcome.


CR after CR after Progressive Partial On Treatment No Total P
first-line further lines disease remission chemotherapy related chemotherapy (N=387)
chemotherapy chemotherapy (n=45) (n=22) (n=15) deaths given
(n=254) (n=21) (n=23) (n=7)
Median age (years) 44 48 45 44 50 48 46 45 0.104b
Male 230 (91%) 20 (95%) 44 (98%) 21 (96%) 15 (100%) 21 (91%) 6 (86%) 357 (92%) 0.514a
Median viral load 19031 26790 40208 7159 11387 2390 105000 18557 0.784b
(copies/mL)
HIV-RNA below limit of detection 74 (30%) 5 (24%) 11 (25%) 8 ((36%) 4 (29%) 8 (35%) 2 (29%) 112 (30%) 0.903a
Median CD4+ 248 190 111 186 153 157 58 212 0.006b
T cells (x109/L)
CD20+ lymphoma 213 (90%) 20 (95%) 34 (81%) 18 (90%) 10 (83%) 13 (68%) 4 (67%) 312 (87%) 0.044a
BM involvement 47 (20%) 7 (33%) 15 (39%) 6 (29%) 3 (21%) 6 (21%) 1(17%) 85 (23%) 0.190a
CNS involvement 17 (8%) 2 (10%) 8 (21%) 2 (11%) 1 (9%) 5 (33%) 0 (0%) 36 (11%) 0.023
IPI score
Low 100 (42%) 5 (24%) 8 (18%) 2 (11%) 5 (39%) 2 (9%) 0 (0%) 122 (36%)
Intermediate 104 (44%) 11 (52%) 20 (46%) 13 (68%) 6 (42%) 11 (50%) 3 (43%) 168 (46%)
High 34 (14%) 5 (24%) 16 (36%) 4 (21%) 2 (15%) 9 (41%) 4 (57%) 74 (20%) <0.001a
Lymphoma subtype
DLBCL 127 (50%) 7 (33%) 24 (53%) 13 (59%) 7 (47%) 7 (30%) 4 (57%) 189 (49%)
BL 91 (36%) 11 (52%) 12 (27%) 5 (23%) 4 (27%) 9 (39%) 1 (14%) 133 (34%)
PBL 29 (11%) 3 (14%) 7 (16%) 3 (14%) 3 (20%) 7 (30%) 2 (29%) 54 (14%)
ARL, not further classifiable 7 (3%) 0 (0%) 2 (4%) 1 (5%) 1 (7%) 0 (0%) 0 (0%) 11 (3%) 0.420a
Median follow up (years) 4.64 5.4 0.5 0.7 0.1 0.2 0 2.4 <0.001b
BL: Burkitt lymphoma; DLBCL: diffuse large B-cell lymphoma; PBL: plasmablastic lymphoma; IPI: International Prognostic Index; BM: bone marrow; CNS: central nervous sys-
tem. aTwo-sided Pearson’s χ2. bKruskal-Wallis test.

Figure 2. Kaplan-Meier estimates for overall survival (OS) of the differ-


ent observed treatment outcomes of aggressive non-Hodgkin lym-
phomas. CR: complete remission; Prog. Disease: progressive disease;
Part. Remission: partial remission. Dotted line indicates 3 months.

860 haematologica | 2018; 103(5)


Relapses in AIDS-related lymphomas

given. Only 37.0% of patients received their full planned received rituximab. There was no difference in the relapse
course of therapy. rate between patients with or without administration of
Patients who experienced treatment delays and/or dose rituximab (P=0.75), and our results remained consistent
reductions or received a reduced number of chemotherapy when patients without rituximab were excluded (data not
cycles were significantly older (42 vs. 45 years; P=0.042) shown).
and had received the GMALL-protocol significantly more
often than patients who completed the full planned course Factors influencing the RFS
of therapy (P=<0.001) (Online Supplementary Table S3). To investigate the influence of different factors, includ-
Overall, 86.2% of patients with CD20+ lymphomas ing treatment reduction or delay on 5yRFS, we investigat-

Table 2. Risk factors for 5-year relapse-free survival (including all aggressive non-Hodgkin lymphoma in first complete remission; n=254).
Aggressive
NHL (N=254)
5-year relapse-free P P
survival % (univariate) (multivariate)
Sex Male (n=230) 87
Female (n=24) 96 0.229
Age >60Y (n=24) 77
<60Y (n=228) 89 0.178
CNS involvement Yes (n=17) 82
No (n=204) 90 0.305
BM involvement Yes (n=47) 81
No (n=193) 90 0.101
Bulky Disease Yes (n=44) 86
No (n=137) 88 0.637
CD4+ T cells <50x109/l Yes (n=37) 86
No (n=202) 88 0.573
Prior AIDS-defining illness Yes (n=56) 87
No (n=193) 88 0797
IPI score Low (n=100) 95 Indicator
Intermediate (n=104) 84 0.760
High (n=34) 82 0.039 0.853
Ann Arbor stage I/II (n=93) 95
III/IV (n=156) 83 0.005 0.011
Extranodal involvement Yes (n=71) 87
No (n=181) 88 0.936
ECOG score 0-1 (n=159) 89
2-5 (n=78) 86 0.635
Elevated LDH Yes (n=146) 86
No (n=96) 92 0.143
Antiretroviral Treatment Viral load b.d. (n=74) 88
Naive (n=134) 88
Therapy failure (n=39) 87 0.986
cART during Chemotherapy Yes (n=234) 89
No (n=9) 67 0.033 0.026
CD20 positive lymphoma Positive (n=213) 88
Negative (n=24) 87 0.888
Lymphoma subtype DLBCL (n=127) 88 Indicator
BL (n=91) 89 0.072
PBL (n=29) 89 0.703
ARL, not further classifiable (n=7) 57 0.064 0.034
Chemotherapy CHOP (n=163) 84
GMALL (n=87) 95 0.013 0.005
Univariate statistics: Log rank test. Multivariate statistics: Cox regression. Viral load b.d.: Viral load below limit of detection; cART: combination antiretroviral therapy; BL: Burkitt
lymphoma; DLBCL: diffuse large B-cell lymphoma; PBL: plasmablastic lymphoma; IPI: International Prognostic Index; BM: bone marrow; CNS: central nervous system; ECOG:
Eastern Cooperative Oncology Group scale.

haematologica | 2018; 103(5) 861


P. Schommers et al.

ed two selected groups: patients with DLBCL receiving R- Discussion


CHOP-based regimens and patients with BL receiving the
GMALL-regimen (see also Figure 1). In patients with In this large prospective cohort study of 254 ARL
DLBCL who received a R-CHOP-based treatment, a high patients who had achieved a CR with first-line
IPI, an elevated LDH, stage III or IV according to the Ann chemotherapy (64% of all cases), the total relapse rate was
Arbor Staging System, and bone marrow involvement at 11% after a median follow up of 4.6 years. Patients with
first diagnosis were associated with a significantly DLBCL who were mainly treated with R-CHOP-based
increased risk of relapse (Online Supplementary Table S4). regimens had a relapse rate of 11%. These rates are in line
However, none of these parameters turned out to be an with the 6-10% relapse rates reported in HIV-negative
independent risk factor in the Cox proportional hazards DLBCL in first CR.6,8 Notably, the 10% relapse rate of
model. Of note, chemotherapy dose reductions, treatment patients with BL who were mainly treated with the
delays or a reduced number of R-CHOP-cycles given did GMALL protocol compares favorably with the 12%
not adversely affect 5yRFS (Online Supplementary Table S4). relapse rate reported in the HIV-negative setting.10
By univariate analysis, patients with BL who underwent Outcome of patients with ARL in which histology was
GMALL-chemotherapy had a significantly increased risk not further classifiable was poor with a 5yRFS of only
of relapse if the following factors were present: diagnosis 57%. There was no difference in type and intensity of
of another AIDS-defining disease prior to BL diagnosis, chemotherapy to that used in DLBCL (data not shown),
CNS-involvement, failure of cART defined as measurable therefore these cases may represent a subgroup of highly
viral loads despite concomitant cART, concomitant cART aggressive lymphomas that may benefit from intensive
during chemotherapy, and reduced numbers of chemotherapy regimens.
chemotherapy cycles administered (Online Supplementary Even though the overall survival of patients with PBL
Table S4). However, none of these factors remained signif- was shown to be significantly worse than that in DLBCL
icant in the Cox proportional hazards model. and BL,16,17 there was no difference in relapse rates

A B

C D

E F

Figure 3. Kaplan-Meier estimates for aggressive non-Hodgkin lymphoma (NHL) that achieved complete remission (CR) after first-line chemotherapy. (A) Overall
survival of all AIDS-related lymphomas (ARL) and of (B) different subtypes (Log rank test: P=0.982). (C) Relapse-free survival of all ARL and of (D) different subtypes
(P=0.064). (E) Relapse-free survival of different subtypes treated with R-CHOP-based regimens (rituximab, cyclophosphamide, adriamycin, vincristine, and pred-
nisone) and (F) GMALL-based chemotherapeutic regimens (P=0.006 and P=0.79, respectively). DLBCL: diffuse-large B-cell lymphoma; BL: Burkitt-lymphoma; PBL:
plasmablastic lymphoma. Dotted line indicates 3 months.

862 haematologica | 2018; 103(5)


Relapses in AIDS-related lymphomas

between patients who have achieved a CR (12%) to those treated with the planned number of intensive chemother-
observed in DLBCL and BL. The inferior OS of the small apy cycles.3,8 By contrast, reduced relative dose intensity
patient group of PBL may at least in part be explained by did not negatively impact 5yRFS in patients treated with
3 late deaths 4-7 years after first diagnosis that were unre- R-CHOP-based regimens for DLBCL. This finding does
lated to lymphoma: one case of sepsis due to pneumonia not correspond to data reported in HIV-negative DLBCL
and 2 cases of secondary malignancies (lung cancer and and warrants further investigation.22,23
oral cavity cancer). It is important to note that this analysis focuses on
AIDS-related lymphoma patients with an intermediate patients in first CR, and that factors that predict RFS were
and high IPI had higher relapse rates and a lower 5yRFS not necessarily associated with initial treatment response.
than those with a low IPI in univariate analysis. The lack Our study has several limitations. First, given the uncon-
of significance in the multivariate analysis was somewhat trolled design selection biases cannot be ruled out. Second,
surprising as previous studies have demonstrated strong the analysis of risk factors associated with outcome is
prognostic relevance of the IPI in ARL.17,18 Whether more exploratory in nature. Given the relatively low num-
patients with HIV-related DLBCL and intermediate or ber of patients in some of the selected subgroups, the sta-
high IPI may benefit from more intensive treatments such tistical power of the analysis is limited and does not allow
as the CHOEP regimen, as has been shown in the HIV- any firm conclusions to be drawn. Notably, data on poten-
negative setting, remains to be seen.19,20 tial risk factors for lymphoma relapse such as adherence to
Previous studies have shown that concomitant cART ART or cumulative viremia between CR and relapse are
was associated with improved CR rates and a trend not available.24 Nevertheless, if a CR has been reached, the
toward improved OS.21 Our results also support a concur- relapse rate was low regardless of whether the CR was
rent use of cART as it was associated with better 5yRFS. achieved with or without dose reduction and whether rit-
Several cART regimens with a good safety and tolerability uximab was used or not. Fourth, CRs were not generally
profile and low interaction potential are now available, confirmed by negative positron emission tomography
strongly arguing for a simultaneous cART during ARL (PET) scans as recommended by current guidelines for
chemotherapy.4 HIV-negative lymphomas.14,25 However, the role of PET-
The use of R-CHOP-based regimens showed signifi- scanning in HIV-lymphoma remains controversial as the
cantly less treatment delays and reductions, as compared rate of false positive results appears to be higher than in
to the GMALL protocol. However, the majority of the HIV-negative setting.26,27 Finally, the number of
patients with BL (84%) received chemotherapy according patients with ARL, not further classified, may have been
to the GMALL-protocol which resulted in significantly lowered by reference pathology services which, in turn,
lower relapse rates compared to R-CHOP-based regimens may have slightly altered our findings.
(Figure 3E and F). Notably, treatment delays and a reduced In conclusion, both CR rates and relapse rates observed
chemotherapy intensity appeared to have no impact on in the German HIV-related Lymphoma Cohort Study are
the relapse-rate in GMALL-treated BL, while, at least in similar to those reported in HIV-negative NHL. These data
the univariate analysis, a reduced number of chemothera- add to the growing body of evidence showing that treat-
py cycles was associated with lower 5yRFS. Thus, our ment outcomes compare favorably with those in patients
data indicate that HIV-infected patients with BL should be with NHL and no HIV infection.

6. Pfreundschuh M, Kuhnt E, Trumper L, et al. multicenter trial. Blood. 2014;124(26):3870-


References CHOP-like chemotherapy with or without 3879.
rituximab in young patients with good- 11. Dunleavy K, Pittaluga S, Shovlin M, et al.
1. Lewden C, May T, Rosenthal E, et al. prognosis diffuse large-B-cell lymphoma: 6- Low-intensity therapy in adults with
Changes in causes of death among adults year results of an open-label randomised Burkitt's lymphoma. N Engl J Med. 2013;
infected by HIV between 2000 and 2005: study of the MabThera International Trial 369(20):1915-1925.
The "Mortalite 2000 and 2005" surveys (MInT) Group. Lancet Oncol. 2011; 12. Ribrag V, Koscielny S, Bosq J, et al.
(ANRS EN19 and Mortavic). J Acquir 12(11):1013-1022. Rituximab and dose-dense chemotherapy
Immune Defic Syndr. 2008;48(5):590-598. 7. Feugier P, Van Hoof A, Sebban C, et al. for adults with Burkitt's lymphoma: a ran-
2. Barta SK, Dunleavy K, Mounier N. Diffuse Long-term results of the R-CHOP study in domised, controlled, open-label, phase 3
large B-cell lymphoma. Hentrich M, Barta the treatment of elderly patients with dif- trial. Lancet. 2016;387(10036):2402-2411.
SK (eds.). HIV-associated Hematological fuse large B-cell lymphoma: a study by the 13. Cheson BD, Horning SJ, Coiffier B, et al.
Malignancies. Springer International Groupe d'Etude des Lymphomes de Report of an international workshop to
Publishing; 2016. l'Adulte. J Clin Oncol. 2005;23(18):4117- standardize response criteria for non-
3. Dunleavy K, Wilson WH. How I treat HIV- 4126. Hodgkin's lymphomas. NCI Sponsored
associated lymphoma. Blood. 2012; 8. Rovira J, Valera A, Colomo L, et al. International Working Group. J Clin Oncol.
119(14):3245-3255. Prognosis of patients with diffuse large B 1999;17(4):1244.
4. Brunnberg U, Hentrich M, Hoffmann C, cell lymphoma not reaching complete 14. Cheson BD, Pfistner B, Juweid ME, et al.
Wolf T, Hubel K. HIV-Associated response or relapsing after frontline Revised response criteria for malignant
Malignant Lymphoma. Oncol Res Treat. chemotherapy or immunochemotherapy. lymphoma. J Clin Oncol. 2007;25(5):579-
2017;40(3):82-87. Ann Hematol. 2015;94(5):803-812. 586.
5. Re A, Michieli M, Casari S, et al. High-dose 9. Montoto S, Noy A, Ribera JM. Burkitt lym- 15. Hudis CA, Barlow WE, Costantino JP, et al.
therapy and autologous peripheral blood phoma. Hentrich M, Barta SK (eds): HIV- Proposal for standardized definitions for
stem cell transplantation as salvage treat- associated Hematological Malignancies. efficacy end points in adjuvant breast can-
ment for AIDS-related lymphoma: long- Springer International Publishing; 2016. cer trials: the STEEP system. J Clin Oncol.
term results of the Italian Cooperative 10. Hoelzer D, Walewski J, Dohner H, et al. 2007;25(15):2127-2132.
Group on AIDS and Tumors (GICAT) study Improved outcome of adult Burkitt lym- 16. Schommers P, Wyen C, Hentrich M, et al.
with analysis of prognostic factors. Blood. phoma/leukemia with rituximab and Poor outcome of HIV-infected patients
2009;114(7):1306-1313. chemotherapy: report of a large prospective with plasmablastic lymphoma: results from

haematologica | 2018; 103(5) 863


P. Schommers et al.

the German AIDS-related lymphoma with rituximab or high-dose chemotherapy Ann Hematol. 2010;89(9):897-904.
cohort study. Aids. 2013;27(5):842-845. (MegaCHOEP) with rituximab for young, 24. Zoufaly A, Stellbrink HJ, Heiden MA, et al.
17. Schommers P, Hentrich M, Hoffmann C, et high-risk patients with aggressive B-cell Cumulative HIV viremia during highly
al. Survival of AIDS-related diffuse large B- lymphoma: an open-label, randomised, active antiretroviral therapy is a strong pre-
cell lymphoma, Burkitt lymphoma, and phase 3 trial (DSHNHL 2002-1). Lancet dictor of AIDS-related lymphoma. J Infect
plasmablastic lymphoma in the German Oncol. 2012;13(12):1250-1259. Dis. 2009;200(1):79-87.
HIV Lymphoma Cohort. Br J Haematol. 21. Barta SK, Xue X, Wang D, et al. Treatment 25. Younes A, Hilden P, Coiffier B, et al.
2015;168(6):806-810. factors affecting outcomes in HIV-associat- International Working Group consensus
18. Barta SK, Xue X, Wang D, et al. A new ed non-Hodgkin lymphomas: a pooled response evaluation criteria in lymphoma
prognostic score for AIDS-related lym- analysis of 1546 patients. Blood. 2013; (RECIL 2017). Ann Oncol. 2017;28(7):1436-
phomas in the rituximab-era. 122(19):3251-3262. 1447
Haematologica. 2014;99(11):1731-1737. 22. Bosly A, Bron D, Van Hoof A, et al. 26. Mhlanga JC, Durand D, Tsai HL, et al.
19. Hentrich M, Hoffmann C, Mosthaf F, et al. Achievement of optimal average relative Differentiation of HIV-associated lym-
Therapy of HIV-associated lymphoma-rec- dose intensity and correlation with survival phoma from HIV-associated reactive
ommendations of the oncology working in diffuse large B-cell lymphoma patients adenopathy using quantitative FDG PET
group of the German Study Group of treated with CHOP. Ann Hematol. 2008; and symmetry. Eur J Nucl Med Mol
Physicians in Private Practice Treating HIV- 87(4):277-283. Imaging. 2014;41(4):596-604.
Infected Patients (DAGNA), in cooperation 23. Hirakawa T, Yamaguchi H, Yokose N, Gomi 27. Sathekge M. Differentiation of HIV-associ-
with the German AIDS Society (DAIG). S, Inokuchi K, Dan K. Importance of main- ated lymphoma from HIV-reactive
Ann Hematol. 2014;93(6):913-921. taining the relative dose intensity of CHOP- adenopathy using quantitative FDG-PET
20. Schmitz N, Nickelsen M, Ziepert M, et al. like regimens combined with rituximab in and symmetry. Eur J Nucl Med Mol
Conventional chemotherapy (CHOEP-14) patients with diffuse large B-cell lymphoma. Imaging. 2014;41(4):593-595.

864 haematologica | 2018; 103(5)


Chronic Lymphocytic Leukemia ARTICLE

Highly similar genomic landscapes in


monoclonal B-cell lymphocytosis and Ferrata Storti
Foundation
ultra-stable chronic lymphocytic leukemia with
low frequency of driver mutations
Andreas Agathangelidis,1* Viktor Ljungström,2* Lydia Scarfò,1 Claudia Fazi,1
Maria Gounari,1,3 Tatjana Pandzic,2 Lesley-Ann Sutton,2,4
Kostas Stamatopoulos,3 Giovanni Tonon,5 Richard Rosenquist2,4**
and Paolo Ghia1**
Haematologica 2018
1
Strategic Research Program on CLL and B-cell Neoplasia Unit, Division of Experimental Volume 103(5):865-873
Oncology, Università Vita-Salute San Raffaele and IRCCS Istituto Scientifico San
Raffaele, Milan, Italy; 2Department of Immunology, Genetics and Pathology, Science for
Life Laboratory, Uppsala University, Sweden; 3Institute of Applied Biosciences, Center for
Research and Technology Hellas, Thessaloniki, Greece; 4Department of Molecular
Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden and 5Functional
Genomics of Cancer Unit, Division of Experimental Oncology, IRCCS Istituto Scientifico
San Raffaele, Milan, Italy
*
AA and VL contributed equally as first authors. **RR and PG contributed equally as last authors

ABSTRACT

D
espite the recent discovery of recurrent driver mutations in chron-
ic lymphocytic leukemia, the genetic factors involved in disease
onset remain largely unknown. To address this issue, we per-
formed whole-genome sequencing in 11 individuals with monoclonal B-
cell lymphocytosis, both of the low-count and high-count subtypes, and
5 patients with ultra-stable chronic lymphocytic leukemia (>10 years
without progression from initial diagnosis). All three entities were indis-
tinguishable at the genomic level exhibiting low genomic complexity
and similar types of somatic mutations. Exonic mutations were not fre-
quently identified in putative chronic lymphocytic leukemia driver genes Correspondence:
in all settings, including low-count monoclonal B-cell lymphocytosis. To ghia.paolo@hsr.it
corroborate these findings, we also performed deep sequencing in 11
known frequently mutated genes in an extended cohort of 28 monoclon-
al B-cell lymphocytosis/chronic lymphocytic leukemia cases.
Received: August 8, 2017.
Interestingly, shared mutations were detected between clonal B cells and
paired polymorphonuclear cells, strengthening the notion that at least a Accepted: February 7, 2018.
fraction of somatic mutations may occur before disease onset, likely at Pre-published: February 15, 2018.
the hematopoietic stem cell level. Finally, we identified previously unre-
ported non-coding variants targeting pathways relevant to B-cell and
chronic lymphocytic leukemia development, likely associated with the doi:10.3324/haematol.2017.177212
acquisition of the characteristic neoplastic phenotype typical of both
monoclonal B-cell lymphocytosis and chronic lymphocytic leukemia. Check the online version for the most updated
information on this article, online supplements,
and information on authorship & disclosures:
Introduction www.haematologica.org/content/103/5/865

Chronic lymphocytic leukemia (CLL), the most common adult leukemia in the
West, is a clinically heterogeneous disease.1 At one end of the spectrum, CLL
©2018 Ferrata Storti Foundation
patients present with an indolent disease that does not require therapy for Material published in Haematologica is covered by copyright.
decades. At the other end of the spectrum, patients experience a rapidly progres- All rights are reserved to the Ferrata Storti Foundation. Use of
published material is allowed under the following terms and
sive disease, need early treatment, and frequently relapse.2,3 conditions:
High-throughput studies14,15 have established that, though displaying a markedly https://creativecommons.org/licenses/by-nc/4.0/legalcode.
lower mutational burden compared to solid tumors,16 CLL is characterized by a Copies of published material are allowed for personal or inter-
diverse genetic landscape with driver gene mutations in pathways considered cen- nal use. Sharing published material for non-commercial pur-
poses is subject to the following conditions:
tral for disease pathogenesis, e.g. NOTCH and NF-κB signaling.7,9,17 The frequency https://creativecommons.org/licenses/by-nc/4.0/legalcode,
of most driver gene mutations in CLL tends to increase in aggressive/refractory sect. 3. Reproducing and sharing published material for com-
cases supporting their involvement mainly in disease progression.18-20 mercial purposes is not allowed without permission in writing
Chronic lymphocytic leukemia is preceded by a condition termed monoclonal from the publisher.
B-cell lymphocytosis (MBL) that is characterized by the presence of circulating
monoclonal B cells with a CLL phenotype, however, at a lower concentration than

Haematologica | 2018; 103(5) 865


A. Agathangelidis et al.

required for a clinical diagnosis of CLL (≥5x109/L).21-24 DNA Blood Mini kit (Qiagen, Germany) was used for samples
MBL, found in otherwise healthy individuals, is divided with more than 1x106 cells as well as for the buccal samples. DNA
into 2 subtypes based on the number of circulating cells: quantity and quality were assayed using the Qubit dsDNA HS
‘high-count MBL’ (HC-MBL: 0.5-5x109/L) that evolves into Assay Kit (Life Technologies, USA).
CLL requiring therapy at a rate of 1%/year,25 and ‘low-
count MBL’ (LC-MBL: <0.5x109/L) that has not been WGS: library preparation
observed to progress into a clinical disease,26 yet persists The Nextera technology was utilized for the library construc-
over time.26,27 Several typical CLL driver gene mutations tion (Nextera™ DNA Sample Prep Kit, Illumina, USA) as it
have been reported in HC-MBL9,28,29 even years before the requires low input material whilst maintaining library complexity.
transition to CLL,30 and these correlate with adverse dis- Fifty ng of genomic DNA were used for the construction of
ease course.31 Such mutations have been reported in mul- libraries that were sequenced in paired-end mode 2x100bp on a
tipotent hematopoietic progenitor CD34+ cells from HiSeq 2000 (Illumina, USA).
patients with CLL,32 suggesting that such aberrations may A variant allele frequency (VAF) of 10% was used as threshold
also be implicated in CLL onset. for variant calling. More detailed information regarding the bioin-
Here, we aimed to gain insight into the genetic lesions formatics analysis is given in the Online Supplementary Appendix.
that may be involved in the transformation from MBL to
CLL, analyzing LC-MBL cases for the first time. To this Targeted re-sequencing: library preparation
end, we used whole-genome sequencing (WGS) and tar- Probes targeting all coding exons or hotspot regions of 11
geted re-sequencing to profile LC-MBL, HC-MBL and a known or postulated CLL driver genes (ATM, BIRC3, MYD88,
particularly indolent subset of CLL, i.e. patients with ultra- NOTCH1, SF3B1, TP53, EGR2, POT1, NFKBIE, XPO1, FBXW7)
stable disease for more than ten years, thus, clinically anal- (Online Supplementary Table S1) were designed using Agilent’s
ogous to MBL. Moreover, in order to explore the possible SureDesign service (https://earray.chem.agilent.com/suredesign/home.
origin of genetic lesions at the hematopoietic progenitor htm). The target regions were captured using the HaloPlex HS tar-
cell level, we analyzed polymorphonuclear (PMN) cells geting enrichment kit (Agilent Technologies, USA). Paired-end
from the study participants. sequencing (150 bp reads) was performed on the NextSeq instru-
We report that the genomic profiles of ultra-stable CLL ment with the use of the 500/550 High Output Kit (Illumina,
patients are very similar to individuals with LC-MBL and USA).
HC-MBL, characterized by infrequent CLL driver gene
mutations that, however, were not associated with dis- Gene enrichment analysis
ease progression. Furthermore, we observed non-coding The identification of genes/gene pathways (gene enrichment
variants (NCVs) that target key pathways/cellular process- analysis, GEA) enriched within the targets of NCVs and motif-
es relevant to normal and neoplastic B-cell development, breaking events caused by NCVs was performed with Enrichr31
thus, potentially contributing to the leukemic transforma- using the KEGG 2016 gene database.
tion. We also found shared somatic mutations between
MBL/CLL and PMN cells, strengthening the notion that at
least a proportion of somatic mutations may occur before Results
the onset of CLL.
WGS reveals highly similar mutational profiles in MBL
and ultra-stable CLL
Methods Whole-genome sequencing was performed on 6 individ-
uals with LC-MBL, 5 individuals with HC-MBL, and 5
The research protocol was approved by the Institutional Ethics patients with ultra-stable CLL. For each individual/patient,
Committee and all participants gave written informed consent in samples from MBL/CLL cells and PMN cells were evaluat-
accordance with the Declaration of Helsinki. ed against buccal (control) cells resulting in a total of 48
samples sequenced with an average autosomal coverage
Study population of 32X (Online Supplementary Table S2). Basic demographic
The study cohort comprised 9 subjects with LC-MBL, 13 sub- and biological characteristics of the MBL/CLL cases
jects with HC-MBL, and 7 patients with Rai stage 0 CLL, herein included in the WGS analysis are provided in Online
called ‘ultra-stable’ CLL. Detailed information about the study Supplementary Table S3. Overall, 37,033 somatic variants
cohort is provided in the Online Supplementary Appendix. were detected in MBL/CLL samples with an average of
2040 somatic variants in LC-MBL (range: 298-2871), 2558
Cell samples in HC-MBL (range: 1428-3483), and 2400 in CLL (range:
Chronic lymphocytic leukemia cells were stained with anti- 1650-3176), respectively. Notably, 2792 variants were
CD19, anti-CD5 and anti-CD20 antibodies. CD19+CD5+CD20dim identified in the 15 PMN control samples compared with
cells were sorted using a High Speed FACS Sorter MoFLo buccal DNA, with an average of 186 variants/sample (the
(Beckman Coulter) according to previously published methods.26 PMN sample from case CLL_3 was excluded from the
PMN cells were sorted based on physical parameters. Buccal cells analysis due to tumor cell contamination) (Figure 1A).
were collected with the use of appropriate buccal swabs Highly analogous mutation rates were observed in HC-
(Epicentre, Madison, USA). MBL and CLL (0.79 and 0.74 mutations per Mb, respec-
tively), while a slightly lower rate was seen in LC-MBL
DNA extraction (0.63 mutations per Mb); this latter finding was due to
The NucleoSpin® Tissue XS kit (Macherey-Nagel, Germany) sample LC-MBL_1 (excluding this sample, the average
was used for DNA extraction in samples with less than 5x104 cells mutation rate for LC-MBL would have been 0.74 muta-
and the QIAamp DNA Micro kit (Qiagen, Germany) in samples tions per Mb) (Figure 1B). The ratio of single-nucleotide
with cell numbers ranging between 5x104 and 1x106. The QIAamp variants (SNVs)/small indels was again almost identical in

866 haematologica | 2018; 103(5)


WGS in MBL and ultra-stable CLL

the three entities (ranging from 11.4 to 12.6), whereas it calculating the pairwise similarity with the 30 previously
was significantly lower in the PMN samples (3.9 for LC- published signatures, where signature 9 and, to some
MBL and CLL and 4.1 for HC-MBL, respectively) (P<0.005 extent, signature 1 where the main contributors (Figure
for all cases) (Figure 1C). 2C). The same analysis in the PMN samples gave different
The transition to transversion (Ti/Tv) ratio ranged from results, with a strong impact of mutational signatures 3
0.99 to 1.13 in the MBL/CLL samples, while it was slightly and 5 (Figure 2D). Signature 3 had previously been identi-
lower in the PMN samples (0.94) (Figure 2A). No clear dif- fied in solid tumors and is associated with failure of DNA
ferences were observed between MBL/CLL and PMN double-strand break-repair by homologous recombina-
samples when the distribution of mutations among the six tion.16 Signature 5 exhibits transcriptional strand bias for
types was examined (Figure 2B). We then evaluated the C>T and T>C mutations at ApTpN context and displays
sequence context of each mutation by incorporating infor- a correlation between smoking history and mutation con-
mation on the bases immediately upstream and down- tribution.16
stream of the mutated base, hence leading to 96 possible
mutation types in this classification16 (Online MBL and ultra-stable CLL display a paucity of
Supplementary Table S4). Almost all major differences were mutations in putative CLL driver genes
identified between MBL/CLL samples and PMN samples Whole-genome sequencing identified 186 non-synony-
with the former group exhibiting mainly C>T mutations mous exonic variants amongst MBL/CLL samples and 15
at NpCpG trinucleotides (P<0.05 for ultra-stable CLL and amongst PMN samples. The average number was 8.9 for
HC-MBL). LC-MBL (range: 1-16), 14.8 for HC-MBL (range: 9-27),
The MutationalPatterns package33 was used to delineate 11.6 for ultra-stable CLL (range: 7-19), and 0.9 for the
the mutational signatures in our cohort. Mutational pat- PMN samples (range: 0-6), respectively (Figure 3A). In
terns identified in the MBL/CLL samples resembled those MBL/CLL samples, the vast majority of non-synonymous
reported by Puente et al.;9 this finding was corroborated by mutations were missense [LC-MBL: 47 of 53 (88.7%); HC-

B C

Figure 1. Somatic mutational analysis of ultra-stable chronic lymphocytic leukemia (CLL), high-count monoclonal B-cell lymphocytosis (HC-MBL), low-count mon-
oclonal B-cell lymphocytosis (LC-MBL) and control polymorphonuclear (PMN) cell samples. (A) Total number of somatic mutations identified by whole-genome
sequencing (WGS) in CLL cell samples from MBL, CLL and the respective PMN samples. All samples carried similar mutational loads with the exception of a single
LC-MBL sample (LC-MBL_1) that displayed a very low number of mutation events; as can be seen, the corresponding PMN sample had a mutation load similar to
the other PMN samples, where comparison of mutation profiles between the MBL and PMN sample showed few common hits, thus excluding the likelihood of con-
tamination. Concerning PMN control samples, they were also characterized by high homogeneity regarding the mutational load. There was a single sample with a
very high mutational load; detailed comparison against its respective CLL sample showed a high overlap of mutations indicating potential tumor cell contamination,
hence this sample was removed from downstream analysis. (B) Average mutation rates ± Standard Deviation (SD) for LC-MBL, HC-MBL and CLL. Highly analogous
mutation rates were observed in the HC-MBL (0.79 mutations per Mb) and CLL (0.74 mutations per Mb) samples, while LC-MBL samples had a slightly lower ratio
(0.63 mutations per Mb). (C) Average SNV to small indels ratio ± SD for all sample groups. All 3 entities displayed similar ratios in clear contrast to the PMN samples
where the ratio was much lower.

haematologica | 2018; 103(5) 867


A. Agathangelidis et al.

MBL: 61 of 73 (83.7%); ultra-stable CLL: 50 of 59 prediction using Polyphen-2 classified all but the CD79B
(84.7%)], whereas the remainder concerned either non- mutation as probably damaging. No CLL driver gene
sense mutations or frameshift indels (Figure 3B and Online mutations were found in the PMN samples.
Supplementary Table S5). Forty-nine of the 186 mutations To assess whether the non-synonymous mutations
(26.3%) had a VAF more than 50%. Concerning the 7 identified here might be potentially relevant to CLL, we
PMN samples harboring mutations, only a single mutation compared our findings to the variants reported by Puente
(6.7%) had a VAF more than 50% (Figure 3C) (Online et al.9 and the International Cancer Genome Consortium
Supplementary Table S6). The most commonly mutated (ICGC) database.37 Overall, the vast majority of genes car-
gene was IGLL5, in accordance with a recently reported rying mutations in our series were also reported as mutat-
study,6 carrying mutations in 5 different samples (2 LC- ed in either or both datasets: 94% in LC-MBL, 89% in
MBL, 2 HC-MBL and 1 CLL samples), likely introduced by HC-MBL, and 97% in CLL.
the somatic hypermutation (SHM) process. Only 6 of 186 We extended our analysis by performing targeted re-
mutations (1.6%) detected in the MBL/CLL samples con- sequencing of 11 putative CLL driver genes in 8 LC-MBL,
cerned putative CLL driver genes, according to 2 recently 13 HC-MBL and 7 ultra-stable CLL samples as well as 24
reported lists.7,9 In detail, 3 were identified in individuals corresponding PMN samples. All but one LC-MBL case
with HC-MBL: i) a single NOTCH1 p.P2514Rfs*4 deletion (LC-MBL_4) subjected to WGS were included in this
(VAF 20%), a known hotspot mutation in CLL10,28,34-36 in analysis (Online Supplementary Table S7). In total, 5 variants
HC-MBL_4; ii) a single FBXW7 p.W307S mutation (VAF were detected in 3 different HC-MBL samples, including 4
26%) in HC-MBL_2; and iii) a single KIAA0947 p.L2093X missense variants and 1 frameshift deletion. Two variants
(VAF 43%) in HC-MBL_5. Two mutations concerned indi- (targeting the NOTCH1 and FBXW7 genes) had been
viduals with LC-MBL: i) a KLHL6 p.A91D mutation (VAF already identified by WGS, whereas the remaining 3 con-
45%) in LC-MBL_5; and ii) a single CD79A p.E200G cerned the POT1 (n=2) and SF3B1 (n=1) genes. In detail, a
mutation (VAF 53%) in LC-MBL_6. Finally, a CD79B single HC-MBL case (HC-MBL_5) harbored an SF3B1
p.N68S mutation (VAF 41%) was identified in a single mutation (p.K700E; VAF 1.1%), a known hotspot muta-
CLL sample (CLL_5). Although most of these exact muta- tion in CLL,18,29,38,39 and a POT1 mutation (p.M1V; 4.3%),
tions have not previously been reported in CLL, functional while the other POT1 mutation (p.S38R; 6.7%) was found

A B

C D

Figure 2. Detailed analysis of mutation types. (A) Transition to transversion (Ti/Tv) ratios were comparable in all monoclonal B-cell lymphocytosis (MBL)/chronic lym-
phocytic leukemia (CLL) samples and somewhat lower in the polymorphonuclear (PMN) cell samples. (B) Similar distribution of mutations among the 6 mutation
classes for each MBL/CLL entity and PMN samples (average values ± Standard Deviation). Similar profiles were evident for all entities with the G>A mutation pre-
dominating in all cases. (C) Mutational signatures that contribute to the somatic mutations observed in the MBL/CLL samples. (D) Mutational signatures that dom-
inate in the PMN samples.

868 haematologica | 2018; 103(5)


WGS in MBL and ultra-stable CLL

in HC-MBL_2, which also harbored the FBXW7 mutation. a DNase I hypersensitive site, while enrichment analysis
Two somatic non-synonymous variants were identified in of the implicated target genes using Enrichr31 led to the
2 different PMN samples: an ATM variant (p.R337C; VAF identification of genes participating in pathways relevant
20%) detected in an LC-MBL case, frequently reported to CLL pathogenesis, such as the MAPK, WNT and AP-1
and probably with a low functional impact, and a TP53 pathways (P<0.0005) (Online Supplementary Table S11).
mutation (p.G245A, VAF 3%) found in a HC-MBL case, Moreover, we examined the potential relation of the
previously reported in several human cancers and lym- NCVs that affect TF binding to AID activity by checking
phomas40 (Online Supplementary Table S8). if they occurred in the known hotspots (WRCY, RGYW,
WA, TW). According to our findings, 21 of 72 (29.2%)
Non-coding variants in MBL and ultra-stable CLL target NCVs were located at AID hotspots. Gene enrichment
genes in pathways relevant to CLL pathogenesis analysis of the remaining 51 target genes revealed similar
Coding and non-coding regions enriched for mutations pathways as in the original analysis (namely AP-1 and
were detected using Fishhook29 in 10 kilobase windows DNA damage response pathways).
across the genome and with compensation for replication
timing. In line with previous findings,9 the analysis Shared mutations between CLL and polymorphonuclear
revealed highest mutational enrichment in the IG loci and cells indicate that somatic variants can arise before
within sites known to be recurrently affected by off-target CLL onset
somatic hypermutation (e.g. BTG2, BCL6 and TCL1A) Shared mutations between MBL/CLL samples and their
(Online Supplementary Figure S1). respective PMN samples were identified in all samples
Funseq2,41 a bioinformatics tool investigating the link- irrespective of origin. Regarding exonic mutations, the
age between NCVs and target genes using integrated same synonymous GSE1 mutation was found in an HC-
bisulfite sequencing, ChIP-Sequencing, and RNA-sequenc- MBL case and its paired PMN sample with comparable
ing data from the Roadmap Epigenomics Project, was VAF (28% vs. 26%). In addition, a LC-MBL sample and its
used for the examination of the NCVs. This analysis paired PMN sample carried an identical mutation within
revealed a total of 1517 variants in the MBL/CLL samples the ncRNA gene LOC339874, though with different VAF
and 39 in the PMN samples. After stringent filtering, 106 (16% vs. 31%). In the case of non-exonic mutations, 179
NCVs of potential relevance to MBL and CLL were iden- shared NCVs were identified between MBL/CLL and
tified (Online Supplementary Table S9): 29 in LC-MBL (aver- PMN samples (Online Supplementary Table S12); the aver-
age 4.8), 45 in HC-MBL (average 9), and 32 in CLL samples age number per sample was 15.8 for LC-MBL, 8.2 for HC-
(average 6.4), respectively; only 4 NCVs were found in 2 MBL, and 9 for ultra-stable CLL (range: 2-34), respectively.
PMN samples. Since we intentionally selected for NCVs in Most of these mutations were intergenic (128 of 179,
transcription factor (TF) highly occupied regions (see 71.5%) (Figure 4C). Interestingly, 6 NCVs were recurrent-
Online Supplementary Appendix), not unexpectedly most ly found in more than one MBL/CLL-PMN sample pair: 3
variants were located in gene promoter sites (Figure 4A). were intergenic and the other 3 were intronic (Online
Twenty-nine variants (26.4%) concerned 16 cancer-associ- Supplementary Table S13). Finally, we also examined the
ated genes and were evenly distributed amongst the 3 mutational signatures for shared mutations between
entities: 9 in LC-MBL samples, 11 in HC-MBL, and 9 in MBL/CLL and PMN samples but did not observe clear cor-
ultra-stable CLL samples. Three of these cancer-associated relations with any signature (data not shown).
genes were recurrently targeted: 9 variants concerned the In order to exclude the possibility of contamination of
BTG2 gene in 4 samples (2 CLL, 1 HC-MBL and 1 LC- the PMN cell fraction by MBL/CLL DNA, we designed
MBL), 5 variants involved the BCL6 gene in 2 samples (1 allele-specific primers (Online Supplementary Table S14) and
HC-MBL and 1 LC-MBL), and 2 variants targeted the performed PCR amplification of the clonotypic IGH gene
BIRC3 gene in 2 samples (1 CLL and 1 HC-MBL). We also rearrangement in both the MBL/CLL and the respective
identified 6 variants concerning the ST6GAL1 gene in 3 PMN samples in 11 of 16 cases with available material.
CLL samples and the same NKIRAS1-related variant in 2 We identified the clonotypic rearrangement in all 11
CLL samples (Figure 4B). Moreover, pathway analysis MBL/CLL samples but in none of the corresponding PMN
with Enrichr31 showed that 30 of 110 (27.3%) of the vari- samples examined, effectively ruling out the possibility
ants targeted genes were implicated in key CLL pathways that the observed results were due to contamination.
and cellular processes, such as the PI3K-AKT pathway
(TCL1A, CCND1, BCL2, PKN1, DDIT4 and SGK3) Somatic copy-number analysis
(P<0.05), the NF-κB pathway (BIRC3, BCL2 and PLAU) sCNA analysis was performed in 3 samples from each
(P<0.05) and the spliceosome machinery (DDX46 and entity and their respective PMN control samples, as well
HSPA2). In most of these cases (22 of 30, 73.3%), the vari- as in 4 additional PMN samples from 1 HC-MBL and 3
ants were located at promoter sites. Comparison to the LC-MBL cases. In total, 16 sCNAs were identified in the
series by Puente et al.9 identified 4 common gene targets: MBL/CLL samples (average: 1.8, range: 1-6): 7 in LC-MBL,
BTG2, BCL6, BACH2 and TCL1A; none of our samples 4 in HC-MBL, and 5 in the ultra-stable CLL samples, all
carried variants affecting either the 3’ UTR of the but one concerning deletion events (Online Supplementary
NOTCH1 gene or the PAX5 gene enhancer. Table S15). Of the recurrent cytogenetic aberrations
We also analyzed the predicted impact of the NCVs on included in the Döhner hierarchical model,42 del(17p),
TF binding and found that 72 of 110 (65.5%) of the vari- del(11q) and trisomy 12 were not identified in any of the
ants could result in a motif-breaking event (LC-MBL: samples, whereas del(13q) was detected in 7 of 9
n=21, HC-MBL: n=33, ultra-stable CLL: n=18) (Online MBL/CLL cases (2 LC-MBL, 3 HC-MBL and 2 CLL cases).
Supplementary Table S10). We subsequently investigated FISH analysis gave concordant results in 5 of 7 cases
genes and gene pathways that may be affected by such TF where data from both techniques were available; in the
motif breaks. In 55 of 72 (76.4%) cases, variants disrupted remaining 2 cases del(13q) was detected with a single

haematologica | 2018; 103(5) 869


A. Agathangelidis et al.

A B

Figure 3. Exonic mutations in our monoclonal B-


cell lymphocytosis (MBL)/chronic lymphocytic
leukemia (CLL) cohort and polymorphonuclear
(PMN) cell samples. (A) Average numbers of
exonic non-synonymous mutations in MBL/CLL
entities and PMN samples. (B) The vast majority
of non-synonymous mutations were missense in
all 3 MBL/CLL entities. PMN samples failed to
show such predominance. (C) Average numbers
of mutations with VAF≥50% for all 3 MBL/CLL
entities were comparable. A single PMN sample
carried a clonal mutation.

technique each (Online Supplementary Table S16). All other somatic hypermutation.16 The second ranking signature 1
sCNAs represented unique events. In terms of distribution is an age-related signature stemming from spontaneous
across the chromosomes, 7 of the 32 (21.9%) sCNAs were deamination of 5-methylcytosine that has been detected
found in the vicinity of centromeres, whereas 11 of 32 in many cancer types.16 Analogies between MBL and ultra-
(34.4%) were located close to a telomere (distance stable CLL extended also to sCNAs in that all samples,
<10x107 bp). None of the PMN samples demonstrated irrespective of origin, carried very few sCNA. Del(13q)
sCNAs typical of CLL. Only one MBL case showed a predominated in all three entities, as shown in previous
shared del(8)(p11.22) between the LC-MBL sample and its studies.26 Most of the sCNAs were located in close prox-
paired PMN sample. imity to either centromeres or telomeres, in keeping with
previous findings reporting significant over-representa-
tions in these regions due to duplication rates.43 Thus,
Discussion most of the sCNAs identified here may not be directly
related to the MBL/CLL phenotype.
Limited information is available concerning the genomic Interestingly, PMN cells harbored a significantly higher
landscape at the very early or indolent phases of CLL. To load of mutations compared to buccal cells. Mutations
this end, we compared the genomes of ultra-stable CLL detected in the PMN samples were characterized by the
cases, defined as those cases stable for more than ten years dominance of distinct mutational signatures compared to
after diagnosis, to genomes from individuals with: i) LC- the MBL/CLL cohort. However, these samples carried
MBL, a condition that does not progress into a clinically shared somatic mutations with the respective MBL/CLL
relevant leukemia;26 and, ii) HC-MBL, a clinically identifi- cell samples in all analyzed cases. Most shared mutations
able pre-leukemic state.25 concerned intergenic regions, yet we also identified a sin-
Both types of MBL and ultra-stable CLL exhibited the gle shared exonic mutation. This finding supports the
same low level of genomic complexity, similar genome- notion that some mutations present in the CLL clone
wide mutation rates, and average number of exonic muta- could be acquired prior to disease onset, as previously sug-
tions, which were distinct from those of the control sam- gested.32
ples. Reflecting this similarity, analysis relating to pub- Almost all genes that were found mutated in HC-MBL
lished mutational signatures revealed similar patterns in and/or LC-MBL had been previously described as recur-
samples from all 3 entities. In more detail, signature 9 that rently mutated in CLL.9,37 In contrast to our recent WES
predominated in the MBL/CLL cohort has been previous- study on relapsing CLL,8 where the great majority of cases
ly identified in CLL and B-cell lymphomas and is attrib- carried at least one CLL driver mutation, such mutations
uted to polymerase η that is involved in AID-induced were relatively scarce in our cohort. Most importantly, the

870 haematologica | 2018; 103(5)


WGS in MBL and ultra-stable CLL

A B

Figure 4. Analysis of non-coding variants and shared mutations between monoclonal B-cell lymphocytosis (MBL)/chronic lymphocytic leukemia (CLL) and poly-
morphonuclear (PMN) cell samples in the present cohort. (A) Topology of the 106 relevant non-coding variants identified in the present study. The majority con-
cerned gene promoter sites. (B) Recurrent non-coding variants in genes relevant to CLL. The BIRC3, BCL6 and BTG2 genes are known to be associated with various
types of cancer. (C) Topology of shared mutations between MBL/CLL and PMN samples. Intergenic mutations predominated, followed by intronic mutations.

lack of any obvious impact of the identified mutations on ing mutome commonly targets gene pathways and cellu-
disease progression after a prolonged follow up highlights lar processes involved in CLL pathobiology. A sizeable
the fact that the mere presence of a given driver mutation proportion of variants affected the promoter sites of genes
does not axiomatically equate with disease progression, as previously associated with cancer, e.g. BTG2, BCL6,
previously reported.31,44 Additional studies are required in BACH2 and TCL1A. Interestingly, all 4 genes have been
order to clarify this phenomenon. recently reported as non-IG targets of the SHM process in
Chronic lymphocytic leukemia cases have been shown patients with lymphomas,47 implicating this otherwise
to harbor detrimental gene mutations in subclones not vis- normal process in the emergence of CLL-like clones.
ible at diagnosis that are progressively selected, e.g. fol- Additional studies need to be performed to address the
lowing the use of chemotherapy.45,46 Such mutations were relevance of such mutations; however, since AID-related
identified by targeted re-sequencing, yet only in a minor mutations are common in CLL,16 they may indeed be rel-
fraction of the present cohort. In this context, it has been evant to disease pathogenesis. We did not identify any
recently proposed that sequence depths greater than “poor-prognostic” 3’ UTR NOTCH1 mutations;9 howev-
4000X will be essential in order to robustly identify all er, we did discover two NCVs targeting indirectly the
subclonal mutations and predict aggressive cases.12 BIRC3 gene. We also found a number of variants within
Arguably, the absence of such driver mutations when the promoter sites of genes implicated in pathways rele-
applying highly sensitive methods may potentially help to vant to CLL biology, including the PI3K-AKT and NF-κB
identify individuals with very indolent disease for whom pathways as well as the spliceosome machinery.
less frequent, if any follow up will be warranted. Furthermore, we identified TF binding motif breaking
Whole-genome sequencing revealed that the non-cod- events that may arise due to NCVs; most concerned the

haematologica | 2018; 103(5) 871


A. Agathangelidis et al.

MAPK, WNT and AP-1 signaling pathways. In this con- In summary, we report that MBL and ultra-stable CLL
text, preliminary results (data not shown) from our ongoing are virtually indistinguishable at the genomic level. While
high-throughput study on aggressive CLL cases showed a this may be reflective of a passive and slow accumulation
great degree of consistency in the targeting of NCVs: the of mutations, we identified both exonic and NCV-targeted
same “CLL-relevant” gene pathways were again among pathways central for B-cell biology and CLL development,
the most common targets of NCVs, further corroborating likely linked to the acquisition of the MBL/CLL pheno-
our present findings. Having said that, some of these vari- type. Importantly, ultra-stable CLL cases carried few
ants could represent bystander SHM targets of unknown known driver gene mutations, even after ten years of fol-
significance or minor contributors to disease pathogenesis, low up, perhaps reflecting the central role of microenvi-
therefore requiring further studies before definitive con- ronmental signals rather than cell-intrinsic defects in shap-
clusions can be drawn regarding their actual significance. ing clonal behavior. In other words, cell-extrinsic trigger-
It is important to note that a recent study on the epige- ing, specifically mediated through the B-cell receptor,
netic profile of CLL48 reported a novel pathogenic role of might represent the major driving force in the early stages
TF dysregulation in CLL, with increased activity of EGR of CLL, whereas disease progression will require acquisi-
and NFAT as well as loss of EBF and AP-1, causing imbal- tion of genetic driver mutations.
ances in the normal B-cell epigenetic program.
Interestingly, certain members of these networks (e.g. Funding
EBF1, JUN and FOS) were among the most commonly This research project was supported by the Associazione
affected TFs across all sample types tested. Collectively, Italiana per la Ricerca sul Cancro, AIRC (Investigator Grant
our findings support the notion that gene pathways could #15189 to PG and Special Program Molecular Clinical
be indirectly targeted by NCVs with the targets being Oncology – 5 per mille #9965), Milano, Italy, Ricerca
either the genes themselves or other interacting genes, e.g. Finalizzata 2010 (#2318823 to PG); Swedish Cancer Society,
TFs. the Swedish Research Council, Uppsala University, Uppsala
Limitations of the present work involve the relatively University Hospital, Lion’s Cancer Research Foundation, and
small size of the cohort, mainly due to the rarity of sam- Selander’s Foundation, Uppsala; and, H2020 “AEGLE, An
ples meeting the selection criteria. In particular, CLL analytics framework for integrated and personalized healthcare
patients had to have stable disease after a prolonged fol- services in Europe”, by the European Union; “MEDGENET,
low up, whereas all individuals with MBL had to have a Medical Genomics and Epigenomics Network” (No.692298) by
persistent monoclonal B-cell population. Concerning LC- the European Union; “GCH-CLL” funded by the General
MBL, low CLL cell number was an additional challenging Secretariat for Research and Technology (GSRT) of Greece and
factor. Furthermore, although our targeted re-sequencing the Italian Ministry of Health (MoH); and IMI2 “HARMO-
approach covered almost 50% of reported mutations in NY”, funded by the European Union. AA is a fellow of
putative driver genes (as reported by Puente et al.),9 by def- Associazione Italiana per la Ricerca sul Cancro AIRC (Triennial
inition this approach is not exhaustive. fellowship “Guglielmina Lucatello é Gino Mazzega”).

8. Ljungstrom V, Cortese D, Young E, et al. lymphocytic leukemia. N Engl J Med.


References Whole-exome sequencing in relapsing 2011;365(26):2497-2506.
chronic lymphocytic leukemia: clinical 15. Burns A, Alsolami R, Becq J, et al. Whole-
1. Chiorazzi N, Rai KR, Ferrarini M. Chronic impact of recurrent RPS15 mutations. genome sequencing of chronic lymphocyt-
lymphocytic leukemia. N Engl J Med. Blood. 2016;127(8):1007-1016. ic leukaemia reveals distinct differences in
2005;352(8):804-815. 9. Puente XS, Bea S, Valdes-Mas R, et al. Non- the mutational landscape between
2. Parikh SA, Shanafelt TD. Prognostic factors coding recurrent mutations in chronic lym- IgHV(mut) and IgHV(unmut) subgroups.
and risk stratification in chronic lympho- phocytic leukaemia. Nature. 2015; Leukemia. 2018;32(2):332-342.
cytic leukemia. Semin Oncol. 2016; 526(7574):519-524. 16. Alexandrov LB, Nik-Zainal S, Wedge DC,
43(2):233-240. 10. Puente XS, Pinyol M, Quesada V, et al. et al. Signatures of mutational processes in
3. Scarfo L, Ferreri AJ, Ghia P. Chronic lym- Whole-genome sequencing identifies recur- human cancer. Nature. 2013;
phocytic leukaemia. Crit Rev Oncol rent mutations in chronic lymphocytic 500(7463):415-421.
Hematol. 2016;104:169-182. leukaemia. Nature. 2011;475(7354):101- 17. Landau DA, Carter SL, Stojanov P, et al.
4. Fabbri G, Khiabanian H, Holmes AB, et al. 105. Evolution and impact of subclonal muta-
Genetic lesions associated with chronic 11. Quesada V, Conde L, Villamor N, et al. tions in chronic lymphocytic leukemia.
lymphocytic leukemia transformation to Exome sequencing identifies recurrent Cell. 2013;152(4):714-726.
Richter syndrome. J Exp Med. mutations of the splicing factor SF3B1 gene 18. Rossi D, Bruscaggin A, Spina V, et al.
2013;210(11):2273-2288. in chronic lymphocytic leukemia. Nat Mutations of the SF3B1 splicing factor in
5. Fabbri G, Rasi S, Rossi D, et al. Analysis of Genet. 2012;44(1):47-52. chronic lymphocytic leukemia: association
the chronic lymphocytic leukemia coding 12. Rose-Zerilli MJ, Gibson J, Wang J, et al. with progression and fludarabine-refrac-
genome: role of NOTCH1 mutational acti- Longitudinal copy number, whole exome toriness. Blood. 2011;118(26):6904-6908.
vation. J Exp Med. 2011;208(7):1389-1401. and targeted deep sequencing of 'good risk' 19. Rossi D, Fangazio M, Rasi S, et al.
6. Kasar S, Kim J, Improgo R, et al. Whole- IGHV-mutated CLL patients with progres- Disruption of BIRC3 associates with flu-
genome sequencing reveals activation- sive disease. Leukemia. 2016;30(6):1301- darabine chemorefractoriness in TP53
induced cytidine deaminase signatures dur- 1310. wild-type chronic lymphocytic leukemia.
ing indolent chronic lymphocytic 13. Schuh A, Becq J, Humphray S, et al. Blood. 2012;119(12):2854-2862.
leukaemia evolution. Nat Commun. 2016; Monitoring chronic lymphocytic leukemia 20. Schnaiter A, Paschka P, Rossi M, et al.
6(8866). progression by whole genome sequencing NOTCH1, SF3B1, and TP53 mutations in
7. Landau DA, Tausch E, Taylor-Weiner AN, reveals heterogeneous clonal evolution pat- fludarabine-refractory CLL patients treated
et al. Mutations driving CLL and their evo- terns. Blood. 2012;120(20):4191-4196. with alemtuzumab: results from the
lution in progression and relapse. Nature. 14. Wang L, Lawrence MS, Wan Y, et al. SF3B1 CLL2H trial of the GCLLSG. Blood.
2015;526(7574):525-530. and other novel cancer genes in chronic 2013;122(7):1266-1270.

872 haematologica | 2018; 103(5)


WGS in MBL and ultra-stable CLL

21. Landgren O, Albitar M, Ma W, et al. B-cell deaminated cytosines are a source of C --> SF3B1 mutations correlated to cytogenetics
clones as early markers for chronic lym- T transitions in vivo. Proc Natl Acad Sci and mutations in NOTCH1, FBXW7,
phocytic leukemia. N Engl J Med. 2009; USA. 1998;95(7):3578-3582. MYD88, XPO1 and TP53 in 1160 untreated
360(7):659-667. 31. Kuleshov MV, Jones MR, Rouillard AD, et CLL patients. Leukemia. 2014;28(1):108-
22. Marti GE, Rawstron AC, Ghia P, et al. al. Enrichr: a comprehensive gene set 117.
Diagnostic criteria for monoclonal B-cell enrichment analysis web server 2016 40. Wilda M, Bruch J, Harder L, et al.
lymphocytosis. Br J Haematol. 2005; update. Nucleic Acids Res. 2016; Inactivation of the ARF-MDM-2-p53 path-
130(3):325-332. 44(W1):W90-97. way in sporadic Burkitt's lymphoma in
23. Dagklis A, Fazi C, Scarfo L, et al. 32. Damm F, Mylonas E, Cosson A, et al. children. Leukemia. 2004;18(3):584-588.
Monoclonal B lymphocytosis in the general Acquired initiating mutations in early 41. Fu Y, Liu Z, Lou S, et al. FunSeq2: a frame-
population. Leuk Lymphoma. 2009; hematopoietic cells of CLL patients. Cancer work for prioritizing noncoding regulatory
50(3):490-492. Discov. 2014;4(9):1088-1101. variants in cancer. Genome Biol. 2014;
24. Ghia P, Prato G, Scielzo C, et al. 33. Blokzijl FJ, R.; van Boxtel, R.; Cuppen, E. 15(10):480.
Monoclonal CD5+ and CD5- B-lympho- MutationalPatterns: comprehensive 42. Dohner H, Stilgenbauer S, Benner A, et al.
cyte expansions are frequent in the periph- genome-wide analysis of mutational Genomic aberrations and survival in chron-
eral blood of the elderly. Blood. 2004; processes. bioRxiv 071761; doi: https:// ic lymphocytic leukemia. N Engl J Med.
103(6):2337-2342. doi.org/10.1101/071761. 2000;343(26):1910-1916.
25. Rawstron AC, Bennett FL, O'Connor SJ, et 34. Baliakas P, Hadzidimitriou A, Sutton LA, et 43. Nguyen DQ, Webber C, Ponting CP. Bias of
al. Monoclonal B-cell lymphocytosis and al. Recurrent mutations refine prognosis in selection on human copy-number variants.
chronic lymphocytic leukemia. N Engl J chronic lymphocytic leukemia. Leukemia. PLoS Genet. 2006;2(2):e20.
Med. 2008;359(6):575-583. 2015;29(2):329-336. 44. Hurtado AM, Chen-Liang TH,
26. Fazi C, Scarfo L, Pecciarini L, et al. General 35. Sutton LA, Young E, Baliakas P, et al. Przychodzen B, et al. Prognostic signature
population low-count CLL-like MBL per- Different spectra of recurrent gene muta- and clonality pattern of recurrently mutat-
sists over time without clinical progression, tions in subsets of chronic lymphocytic ed genes in inactive chronic lymphocytic
although carrying the same cytogenetic leukemia harboring stereotyped B-cell leukemia. Blood Cancer J. 2015;5:e342.
abnormalities of CLL. Blood. 2011; receptors. Haematologica. 2016;101(8):959- 45. Ojha J, Ayres J, Secreto C, et al. Deep
118(25):6618-6625. 967. sequencing identifies genetic heterogeneity
27. Matos DM, Furtado FM, Falcao RP. 36. Lionetti M, Fabris S, Cutrona G, et al. High- and recurrent convergent evolution in
Monoclonal B-cell lymphocytosis in indi- throughput sequencing for the identifica- chronic lymphocytic leukemia. Blood.
viduals from sporadic (non-familial) chron- tion of NOTCH1 mutations in early stage 2015;125(3):492-498.
ic lymphocytic leukemia families persists chronic lymphocytic leukaemia: biological 46. Rossi D, Khiabanian H, Spina V, et al.
over time, but does not progress to chronic and clinical implications. Br J Haematol. Clinical impact of small TP53 mutated sub-
B-cell lymphoproliferative diseases. Rev 2014;165(5):629-639. clones in chronic lymphocytic leukemia.
Bras Hematol Hemoter. 2015;37(5):292- 37. Zhang J, Baran J, Cros A, et al. International Blood. 2014;123(14):2139-2147.
295. Cancer Genome Consortium Data Portal--a 47. Khodabakhshi AH, Morin RD, Fejes AP, et
28. Rasi S, Monti S, Spina V, et al. Analysis of one-stop shop for cancer genomics data. al. Recurrent targets of aberrant somatic
NOTCH1 mutations in monoclonal B-cell Database (Oxford). 2011;2011:bar026. hypermutation in lymphoma. Oncotarget.
lymphocytosis. Haematologica. 2012; 38. Strefford JC, Sutton LA, Baliakas P, et al. 2012;3(11):1308-1319.
97(1):153-154. Distinct patterns of novel gene mutations 48. Oakes CC, Seifert M, Assenov Y, et al.
29. Imielinski M, Guo G, Meyerson M. in poor-prognostic stereotyped subsets of DNA methylation dynamics during B cell
Insertions and Deletions Target Lineage- chronic lymphocytic leukemia: the case of maturation underlie a continuum of disease
Defining Genes in Human Cancers. Cell. SF3B1 and subset #2. Leukemia. 2013; phenotypes in chronic lymphocytic
2017;168(3):460-472 e414. 27(11):2196-2199. leukemia. Nature Genetics. 2016;48(3):253-
30. Kreutzer DA, Essigmann JM. Oxidized, 39. Jeromin S, Weissmann S, Haferlach C, et al. 264.

haematologica | 2018; 103(5) 873


ARTICLE Chronic Lymphocytic Leukemia

Toxicities and outcomes of 616


Ferrata Storti
Foundation ibrutinib-treated patients in the United States:
a real-world analysis
Anthony R. Mato,1 Chadhi Nabhan,2 Meghan C. Thompson,1 Nicole Lamanna,3
Danielle M. Brander,4 Brian Hill,5 Christina Howlett,6,7 Alan Skarbnik,7 Bruce D.
Cheson,8 Clive Zent,9 Jeffrey Pu,10 Pavel Kiselev,11 Andre Goy,7 David Claxton,10
Krista Isaac,12 Kaitlin H. Kennard,1 Colleen Timlin,1 Daniel Landsburg,1 Allison
Winter,5 Sunita D. Nasta,1 Spencer H. Bachow,3 Stephen J. Schuster,1 Colleen
Dorsey,1 Jakub Svoboda,1 Paul Barr13* and Chaitra S. Ujjani8*
Haematologica 2018
Hematology and Oncology, University of Pennsylvania, Philadelphia, PA; 2Cardinal Health,
Volume 103(5):874-879
1

Dublin, OH 3Hematology/Oncology, Presbyterian/Columbia University Medical Center, New


York, NY; 4Hematology/Oncology, Duke University, Durham, NC; 5Hematology and Medical
Oncology, Cleveland Clinic, OH; 6Pharmacy, Ernest Mario School of Pharmacy, New
Brunswick, NY; 7Hematology/Oncology, John Theurer Cancer Center, Hackensack, NY;
8
Hematology/Oncology, Georgetown University Hospital, Washington DC;
9
Hematology/Oncology, University of Rochester Medical Center, NY; 10Hematology/Oncology,
Penn State Milton S Hershey Medical Center, PA; 11Lymphoma, Celgene Corp, Summit, NY;
12
Hematology/Oncology, Lankenau Hospital, Wynnewood, PA and 13Division of Hematology
and Oncology, University of Rochester, NY, USA
*Indicates shared last authorship

ABSTRACT

C
linical trials that led to ibrutinib’s approval for the treatment of
chronic lymphocytic leukemia showed that its side effects differ
from those of traditional chemotherapy. Reasons for discontinua-
tion in clinical practice have not been adequately studied. We conducted a
retrospective analysis of chronic lymphocytic leukemia patients treated
Correspondence: with ibrutinib either commercially or on clinical trials. We aimed to com-
pare the type and frequency of toxicities reported in either setting, assess
amato@mskcc.org discontinuation rates, and evaluate outcomes. This multicenter, retrospec-
tive analysis included ibrutinib-treated chronic lymphocytic leukemia
patients at nine United States cancer centers or from the Connect®
Received: October 20, 2017. Chronic Lymphocytic Leukemia Registry. We examined demographics,
Accepted: January 26, 2018. dosing, discontinuation rates and reasons, toxicities, and outcomes. The
Pre-published: February 1, 2018. primary endpoint was progression-free survival. Six hundred sixteen ibru-
tinib-treated patients were identified. A total of 546 (88%) patients were
treated with the commercial drug. Clinical trial patients were younger
doi:10.3324/haematol.2017.182907 (mean age 58 versus 61 years, P=0.01) and had a similar time from diagnosis
Check the online version for the most updated to treatment with ibrutinib (mean 85 versus 87 months, P=0.8). With a
information on this article, online supplements, median follow-up of 17 months, an estimated 41% of patients discontin-
and information on authorship & disclosures: ued ibrutinib (median time to ibrutinib discontinuation was 7 months).
www.haematologica.org/content/103/5/874 Notably, ibrutinib toxicity was the most common reason for discontinua-
tion in all settings. The median progression-free survival and overall sur-
©2018 Ferrata Storti Foundation vival for the entire cohort were 35 months and not reached (median fol-
low-up 17 months), respectively. In the largest reported series on ibrutinib-
Material published in Haematologica is covered by copyright.
All rights are reserved to the Ferrata Storti Foundation. Use of treated chronic lymphocytic leukemia patients, we show that 41% of
published material is allowed under the following terms and patients discontinued ibrutinib. Intolerance as opposed to chronic lympho-
conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode.
cytic leukemia progression was the most common reason for discontinua-
Copies of published material are allowed for personal or inter- tion. Outcomes remain excellent and were not affected by line of therapy
nal use. Sharing published material for non-commercial pur- or whether patients were treated on clinical studies or commercially. These
poses is subject to the following conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode, data strongly argue in favor of finding strategies to minimize ibrutinib
sect. 3. Reproducing and sharing published material for com- intolerance so that efficacy can be further maximized. Future clinical trials
mercial purposes is not allowed without permission in writing
from the publisher.
should consider time-limited therapy approaches, particularly in patients
achieving a complete response, in order to minimize ibrutinib exposure.

874 haematologica | 2018; 103(5)


Ibrutinib: treatment toxicities and outcomes

Introduction International Workshop on Chronic Lymphocytic Leukemia


(IWCLL) criteria to define response and progression of disease.18,19
Ibrutinib is an orally bioavailable, irreversible inhibitor Patients were stratified by line of therapy (front-line versus
of Bruton tyrosine kinase (BTK). It is a standard of care in relapsed/refractory), reason for discontinuation (intolerance versus
the treatment of chronic lymphocytic leukemia (CLL) in progressive disease), clinical trial participation versus commercial
the relapsed/refractory1-3 as well as front-line4 settings. use, and depth of response (complete response versus partial
The toxicities of ibrutinib and reasons for its discontin- response and partial response with lymphocytosis). Secondary
uation were initially defined through several landmark endpoints included overall survival and reasons for ibrutinib dis-
studies comparing ibrutinib to chlorambucil (front-line, continuation. Overall survival was defined as the time in months
RESONATE 2),4 ofatumumab (RESONATE)2 and ben- from initiation of ibrutinib to death.
damustine and rituximab +/- ibrutinib versus placebo Reasons for ibrutinib discontinuation were categorized as fol-
(HELIOS).5 Higher proportions of patients discontinuing lows: toxicity, progressive disease, Richter transformation to
therapy (ranging from 25% to 51%) have been found with either diffuse large B-cell lymphoma or Hodgkin lymphoma,
long-term follow-up (median follow-ups ranging between planned cellular therapy (allogeneic hematopoietic stem cell trans-
20 months and approximately 5 years) of these and other plantation or chimeric antigen receptor genetically modified T-cell
ibrutinib clinical trials. Although the percentage of discon- therapy), secondary malignancies, physician’s or patient’s prefer-
tinuations due to progression of disease (21% to 45%) ence, financial concerns, and other/unrelated death. Toxicities
remained similar, there were high proportions of discon- leading to discontinuation were categorized as: hematologic toxi-
tinuations due to adverse events, ranging from 12% to city, infection, atrial fibrillation, congestive heart failure, drug-
32%.6-10 induced pneumonitis, drug-induced colitis, transaminitis, bleed-
Woyach et al. suggested that discontinuation due to CLL ing, arthralgia/myalgia, dermatological, neurotoxicity, other, or
progression (distinguished from disease transformation) unknown.
occurred later in the disease course (cumulative incidence Survival data were compared using the log-rank test.20
of 7.3% at 2 years, 19.1% at 4 years) while other reasons Univariate Cox regression analyses were used to estimate hazard
for discontinuation, such as Richter transformation, ratios.21 All other comparison analyses were descriptive. All tests
peaked early and reached a plateau by 3 years (18.7% at 2 were two-sided at the 5% level. Statistical analyses were per-
years, 23.9% at 3 years).8 In clinical practice, adverse formed using STATA 10.1 (Stata Statistical Software: Release 10.
events were found to be the most common cause of ibru- 2007; StataCorp LP, College Station, TX, USA).
tinib discontinuation among 143 patients, approaching
50%.11 Atrial fibrillation, infectious complications, and
cytopenias were the most commonly described adverse Results
events.11 High percentages of patients discontinuing thera-
py, ranging from 19-41%, were encountered in additional Patients
studies conducted in the USA and outside of the USA; We identified 616 patients who received ibrutinib,
however, these studies were limited by relatively small including 536 relapsed-refractory and 80 previously
sample sizes and short follow-up periods.12-15 untreated patients. Data from the nine contributing aca-
We, therefore, aimed to characterize patterns of care demic centers were collected retrospectively and included
among ibrutinib-treated CLL patients in clinical practice in information for 399 patients treated with ibrutinib; data
the USA focusing on rates and reasons for discontinuation, from the Connect CLL registry were collected prospec-
and how these affect outcomes. To our knowledge, this tively and included information on 217 patients largely
series is the largest report on ibrutinib-treated CLL collected from community sites (80% community). The
patients. patients’ baseline characteristics stratified by line of thera-
py are available in Table 1. A total of 546 (88%) patients
were treated with commercially available drug/off study.
Methods Clinical trial patients were younger (mean age 58 versus 61
years, P=0.01), had a similar time from diagnosis to treat-
We conducted a multicenter, retrospective cohort study of CLL ment with ibrutinib (mean 85 versus 87 months, P=0.8)
patients at nine USA cancer centers and from the Connect® CLL and were more consistently initiated at a dose of 420 mg
Registry (199 USA centers, 80% community sites) who were daily (100% versus 89%).
treated with ibrutinib either as part of a clinical trial or with com-
mercially available drug.16 The institutional review board of each Reasons for discontinuation, toxicities and timing
participating institution approved this study. Investigators at each of events
institution were asked to utilize chart review, institutional elec- At a median follow-up of 17 months (range, 1-60
tronic medical records and clinical/pathological databases to months), 41% of patients discontinued ibrutinib. The
obtain required information for all CLL patients treated with ibru- median time to ibrutinib discontinuation was 7 months
tinib. Data collected included: patients’ demographics, genetic (range, 0.1–41), with a median time of 6 months for
characteristics, number of prior therapies, dosing and dose adjust- patients who discontinued due to intolerance and 10
ments, discontinuation rates and reasons, toxicities and outcomes. months for those who discontinued due to progression of
The period of enrollment was January 2014 to August 2016. disease (Online Supplementary Figure S1). Among patients
The primary study endpoint was progression-free survival, on ibrutinib monotherapy, 41% discontinued therapy
which was defined as time from ibrutinib treatment to progres- while the percentage of discontinuation among patients
sion or death from any cause as per the Kaplan Meier method.17 receiving ibrutinib-based combination therapy was
Patients were otherwise censored, regardless of progression status, 43.9%. Ibrutinib starting dose (420 mg daily versus <420
at the time of last follow-up and at the time of next therapy. When mg daily) did not correlate with the proportion of patients
interpreting medical records, investigators were advised to use the who discontinued ibrutinib due to toxicity (51% versus

haematologica | 2018; 103(5) 875


A.R. Mato et al.

50%) or disease progression (19.6% versus 21.4%). for 5.3% of the discontinuations in the front-line setting
Reasons for ibrutinib discontinuation are listed in Table and 5.0% in the relapsed/refractory setting.
2. Percentages listed indicate the proportion of discontin- Among the patients treated front-line with ibrutinib, the
uations due to each category. Toxicity was the most com- three most common toxicities leading to discontinuation
mon reason for discontinuation in all settings, accounting were arthralgia (41.6%), atrial fibrillation (25%), and rash
for 63.1% of discontinuations in front-line use (n=12/80 (16.7%). In the relapsed/refractory population, the most
front-line patients) and 50.2% of discontinuations in common toxicities leading to discontinuation were atrial
relapsed/refractory use (n=116/536 relapsed/refractory fibrillation (12.3%), infection (10.7%), pneumonitis
patients). Toxicity was the most common reason for dis- (9.9%), bleeding (9%) and diarrhea (6.6%).
continuation in several settings including: commercial use The median time to ibrutinib discontinuation varied by
and clinical trial use (50% of discontinuations in front-line toxicity: bleeding (8 months), diarrhea (7.5 months), atrial
commercial use, 77.7% of discontinuations in front-line fibrillation (7 months), infection (6 months), arthralgia (5
clinical trial use, 52.5% of discontinuations in months), pneumonitis (4.5 months) and rash (3.5 months).
relapsed/refractory commercial use, and 39.7% of discon-
tinuations in relapsed/refractory trial use). Notably, the Outcomes
proportion of discontinuations due to progressive disease At a median follow-up of 17 months, the median pro-
was lower: 15.8% in the front-line setting and 20.9% in gression-free and overall survival for the entire cohort
relapsed/refractory use. Richter transformation to diffuse were 35 months and not reached, respectively (Figure
large B-cell lymphoma or Hodgkin lymphoma accounted 1A,B). Overall survival from the start of ibrutinib therapy,

Table 1. Baseline characteristics.


Baseline characteristics
Ibrutinib in front line Ibrutinib in relapse
Total number 80 536
Median age at diagnosis, years (range) 62 (37-88), n=78 60 (22-95), n=532
Median time from diagnosis to ibrutinib start, months (range) 25.5 (1-232), n=80 78.3 (1-366), n=536
del17p(+) 37%, n=76 26%, n=368
del11q(+) 19%, n=75 35%, n=367
Tp53mut(+) 12%, n=42 13%, n=142
Complex karyotype (+) (>3) 40%, n=60 34%, n=214
Median time diagnosis to ibrutinib, months (range) 26 (1-232) 78 (1-660)
Median ibrutinib starting dose 420 mg 420 mg
Ibrutinib administered as monotherapy 68%, n=80 89%, n=536
Ibrutinib suspension required 30%, n=79 37%, n=310
Ibrutinib dose adjusted 15%, n=79 20%, n=309
Median follow-up 17 months
del17p(+) (deletion 17p mutation positive); del11q(+) (deletion 11q mutation positive); Tp53mut(+) (Tp53 mutation positive); Complex karyotype (+)(>3) (complex karyotype
positive defined as three or more abnormalities).

A B

Figure 1. Outcomes for the entire cohort. Kaplan Meier curves at a median follow-up of 17 months showing (A) progression-free survival (PFS) for the entire cohort
and (B) overall survival (OS) for the entire cohort.

876 haematologica | 2018; 103(5)


Ibrutinib: treatment toxicities and outcomes

stratified by whether the drug was being used in the Investigator-assessed depth of response (complete
front-line versus relapsed/refractory setting, is shown in response versus partial response versus partial response
Online Supplementary Figure S2A,B. Notably, there was no with lymphocytosis versus stable disease versus progres-
significant difference in progression-free survival by sive disease) appeared to correlate with a longer progres-
front-line versus relapsed/refractory use (P=0.27, log-rank sion-free survival (Figure 2E). We also stratified progres-
test) (Figure 2A) or at first, second or third relapse sion-free survival by deletion 17p status and complex
(P=0.45) (Online Supplementary Figure S3). Progression-free karyotype status (≥3 abnormalities) in CLL patients treat-
survival was similar when stratified by ibrutinib use in ed in the relapsed/refractory setting. Progression-free sur-
the clinical practice setting as compared to the clinical vival was not significantly different in patients with dele-
trial setting (P=0.14, log-rank test) (Figure 2B). Patients tion 17p (P=0.70), but was significantly shorter in patients
who discontinued due to toxicity had significantly longer with a complex karyotype (hazard ratio=1.8, 95% confi-
progression-free survival and overall survival than those dence interval: 1.1-3.0, P=0.01). The Kaplan Meier curves
who discontinued due to disease progression (P=0.01 and for these analyses are shown in Online Supplementary
P=0.02, respectively, log-rank test) (Figure 2C,D). Figure S4A-C.

A B

C D

Figure 2. Outcomes stratified by line of therapy, clinical trial participation,


reason for discontinuation and depth of response. Kaplan Meier curves
showing outcomes stratified by (A) line of therapy (progression-free survival),
(B) clinical trial participation (progression-free survival), (C) reason for dis-
continuation (progression-free survival), (D) reason for discontinuation (over-
all survival), and (E) depth of response. CR: complete response; PR: partial
response; PR-L: partial response with lymphocytosis; SD: stable disease; PD:
progressive disease.

haematologica | 2018; 103(5) 877


A.R. Mato et al.

Discussion Table 2. Reasons for Ibrutinib discontinuation.


Reason for ibrutinib Ibrutinib in Ibrutinib in relapse
In the largest reported series of ibrutinib-treated CLL discontinuation front-line (n=19) (n=231)
patients so far, we found that the median progression-free
Toxicity 63.1% (n=12) 50.2% (n=116)
survival was 35 months. Interestingly, this outcome was
comparable between previously untreated and CLL progression 15.8% (n=3) 20.9% (n=49)
relapsed/refractory patients. Our observed progression- Other/unrelated death 5.3% (n=1) 12.1% (n=28)
free survival of 35 months is shorter than that previously Physician’s or patient’s preference 10.5% (n=2) 6.7% (n=15)
described in clinical trials in which the median progres- RT DLBCL 5.3% (n=1) 4.6% (n=10)
sion-free survival was 52 months in relapsed/refractory
disease.9 These observations suggest that outcomes vary Stem cell transplantation/CAR T-cell 0 3.3% (n=8)
when comparing clinical trial patients and those treated in Financial concerns 0 0.8% (n=2)
clinical practice, underscoring the need to better under- Secondary malignancy 0 0.8% (n=2)
stand outcomes and toxicities in a real-world setting. RT Hodgkin lymphoma 0 0.4% (n=1)
Patient-specific factors, including molecular prognostic
CLL: chronic lymphocytic leukemia; RT DLBCL: Richter transformation to diffuse large
markers, performance status and prior therapies, may par- B-cell lymphoma; CAR T-cell: chimeric antigen receptor T-cell); RT: Richter transforma-
tially account for these discrepancies. Our front-line tion.
cohort of patients had a greater number of molecular
abnormalities than typically seen in the treatment-naïve
population; 37% of patients had del17p and 40% had a periods of time on treatment,24 these data may be of value
complex karyotype. Both features are associated with an in developing monitoring strategies for specific toxicities
inferior progression-free survival.22 For the relapsed and educational material related to ibrutinib toxicities.
cohort, patients may have received prior idelalisib which Reasons for discontinuation also varied by line of thera-
could have affected their subsequent response to ibrutinib; py. Our analysis revealed a strikingly higher number of
notably, this was prohibited in the earliest clinical trials of discontinuations due to toxicity in the front-line setting;
ibrutinib.11 63% compared to the previously reported 9% in RES-
At a median follow-up of 17 months, overall discontin- ONATE-2.4 Similar findings, although not as marked,
uation in our study was high at 41%, suggesting that ibru- were noted in the relapsed setting; 50% of patients in our
tinib discontinuation is an emerging issue in clinical prac- analysis discontinued therapy due to intolerance com-
tice. This mirrors the high overall discontinuation patterns pared to 12% discontinuing due to adverse events in the
seen in longer follow-up studies of clinical trial patients,6-9 4-year follow-up of RESONATE data.6 In line with prior
particularly the 51% estimated discontinuations seen at a reports, we found that atrial fibrillation and pulmonary
median follow-up of 3.4 years by Woyach et al.8 In addi- complications were common reasons for discontinuation.
tion, 15% of front-line and 20% of relapsed ibrutinib- Arthralgias and rash were frequently noted as well, partic-
treated patients required a dose reduction, which is signif- ularly in treatment-naïve patients. The higher discontinu-
icantly higher than the 4% of patients requiring dose ation rate for toxicity may reflect lack of physicians’ com-
reduction due to adverse events in the RESONATE trial fort in toxicity management, a higher incidence of toxicity
due to adverse events (4%).2 in clinical practice, differences in patients’ comorbidities
Early ibrutinib trials suggested that progressive disease and age, or a lower threshold for discontinuation given an
was the cause of the majority of cases of discontinuation,2 increasing number of available alternative treatment
a pattern that has persisted in many,6,8,9 but not all,5,7,10 stud- choices. This may be in contrast to the limited number of
ies including longer follow-up periods. Surprisingly, our therapies available to ibrutinib-treated patients in early
results demonstrate that intolerance (50.2% in the clinical trials. In a recent series by Lampson et al. of treat-
relapsed/refractory setting, 63% in the front-line setting), ment-naïve patients treated with idelalisib plus ofatu-
rather than progressive disease (21% in the mumab adverse events (particularly liver toxicity) were
relapsed/refractory setting, 16% in the front-line setting), the most common reasons for discontinuation.25 This
accounts for the majority of cases of discontinuation. study suggested that younger patients’ age and intact
Similar findings were previously noted in a smaller series immunity may lead to autoimmune treatment-related tox-
reported by our group.11 With respect to adverse events icities in treatment-naïve patients.25
leading to discontinuation, these values are greater than Progression of disease was the second most common
those reported in a pooled analysis from the phase III indication for ibrutinib discontinuation. Sixteen percent of
relapsed RESONATE and front-line RESONATE-2 studies previously untreated patients experienced progression
in which the discontinuation rate was 10%.23 Because at compared to 10% progressions/deaths at an 18-month fol-
least 50% of discontinuations are due to intolerance rather low-up in the front-line RESONATE-2 study.4 This differ-
than progression of disease, it is unlikely that these ence may be related to the exclusion of patients with
patients harbor ibrutinib resistance mutations and, there- del17p from the RESONATE-2 study.4 Rates of discontin-
fore, should likely be sensitive to alternate kinase uation due to progression in the relapsed population in our
inhibitors with different side effect profiles. Two clinical cohort were more comparable with the 27% found in the
trials are being conducted at this time studying umbralisib 4-year follow-up of the RESONATE study.6 Outcomes of
(NCT02742090) and acalabrutinib (NCT02717611) in patients in our series who discontinued therapy due to
kinase inhibitor-intolerant patients. toxicity were superior to those of patients who discontin-
For the first time we have established a timeline associ- ued due to progressive disease.
ated with time to discontinuation due to specific ibrutinib- We also considered limitations in the study design.
related toxicities. Similar to the experience with idelalisib Conducted by physicians and research coordinators from
where specific toxicities appear to occur after different several institutions in a retrospective manner, data collec-

878 haematologica | 2018; 103(5)


Ibrutinib: treatment toxicities and outcomes

tion may have been affected by inconsistencies in chart small subset of patients who achieve complete remission.
interpretation, as well as clinical experience and practice This strategy has been successfully demonstrated in
style. There were a disproportionate number of patients receiving venetoclax who were able to achieve
relapsed/refractory patients compared to front-line minimal residual disease negativity.26 For example, the
patients. incorporation of BCL-2 inhibitors and/or anti-CD20 mon-
It is vital to develop strategies to mitigate ibrutinib intol- oclonal antibody therapies in combination with ibrutinib
erance so that efficacy can be further maximized. may enable patients to experience minimal residual dis-
Examples include the creation of guidelines for the evalu- ease-negative responses that may translate into shorter
ation and management of problematic side effects such as durations of treatment.27,28
atrial fibrillation, rash, and arthralgias. An educational
forum focused on oncologists, physician educators, and Acknowledgments
nurses should be implemented. In addition, the design of The authors thank Joseph and Cindy Riggs for their ongoing
future clinical trials should allow for cessation of therapy support of this work. They also thank the Center for CLL,
in order to minimize ibrutinib exposure, particularly in the University of Pensylvania.

References 10. Jain P, Thompson P, Keating M, et al. Long- points in chronic lymphocytic leukemia. J
term outcomes for patients with chronic Clin Oncol. 2012;30(23):2820-2822.
lymphocytic leukemia who discontinue 20. Matthews D, Farewell V. 7 The log-rank or
1. Byrd J, Furman R, Coutre S, et al. Targeting ibrutinib. Cancer. 2017;123(12): 2268-2273. Mantel-Haenszel test for the comparison of
BTK with ibrutinib in relapsed chronic lym- 11. Mato A, Nabhan C, Barr P, et al. Outcomes survival curves in: Basel S, Karger A, editors.
phocytic leukemia. N Engl J Med. of CLL patients treated with sequential Using and Understanding Medical Statistics
2013;369(1):32-34. kinase inhibitor therapy: a real world expe- 2007:67-75.
2. Byrd J, Brown J, O'Brien S, et al. Ibrutinib rience. Blood. 2016;128(18):2199-2205. 21. Anderson P, Gill R. Cox’s regression model
versus ofatumumab in previously treated 12. Parikh S, Chaffee K, Call T, et al. Ibrutinib for counting processes: a large sample study.
chronic lymphoid leukemia. N Engl J Med. therapy for chronic lymphocytic leukemia Ann Statist. 1982;10(4):1100-1120.
2014;371(3):213-223. (CLL): an analysis of a large cohort of 22. Thompson PA, O'Brien SM, Wierda WG, et
3. O'Brien S, Jones J, Coutre S, et al. Ibrutinib patients treated in routine clinical practice. al. Complex karyotype is a stronger predic-
for patients with relapsed or refractory Blood. 2015;126(23):2935. tor than del(17p) for an inferior outcome in
chronic lymphocytic leukaemia with 17p 13. Sandoval-Sus J, Chavez J, Dalia S, et al. relapsed or refractory chronic lymphocytic
deletion (RESONATE-17): a phase 2, open- Outcomes of patients with relapsed/refrac- leukemia patients treated with ibrutinib-
label, multicentre study. Lancet Oncol. tory chronic lymphocytic leukemia after based regimens. Cancer. 2015;121(20): 3612-
2016;17(10):1409-1418. ibrutinib discontinuation outside clinical tri- 3621.
4. Burger J, Tedeschi A, Barr P, et al. Ibrutinib as als: a single institution experience. Blood. 23. O'Brien SM, Byrd JC, Hillmen P, et al.
initial therapy for patients with chronic lym- 2015;126:2945. Outcomes with ibrutinib by line of therapy
phocytic leukemia N Engl J Med. 14. Winqvist M, Asklid A, Andersson P, et al. in patients with CLL: analyses from phase III
2015;373(25):2425-2437. Real-world results of ibrutinib in patients data. J Clin Oncol. 2016;34(15_suppl): 7520.
5. Chanan-Khan A, Cramer P, Demirkan F, et with relapsed or refractory chronic lympho- 24. Coutré SE, Barrientos JC, Brown JR, et al.
al. Ibrutinib combined with bendamustine cytic leukemia: data from 95 consecutive Management of adverse events associated
and rituximab compared with placebo, ben- patients treated in a compassionate use pro- with idelalisib treatment: expert panel opin-
damustine, and rituximab for previously gram. Haematologica. 2016;101(12): 1573- ion. Leuk Lymphoma. 2015;56(10): 2779-
treated chronic lymphocytic leukaemia or 1580. 2786.
small lymphocytic lymphoma (HELIOS): a 15. UK CLL Forum. Ibrutinib for 25. Lampson B, Kasar S, Matos T, et al. Idelalisib
randomised, double-blind, phase 3 study. relapsed/refractory chronic lymphocytic given front-line for treatment of chronic
Lancet Oncol. 2016;17(2):200-211. leukemia: a UK and Ireland analysis of out- lymphocytic leukemia causes frequent
6. Byrd J, Hillmen P, O’Brien S, et al. Long-term comes in 315 patients. Haematologica. immune-mediated hepatotoxicity. Blood
efficacy and safety with ibrutinib (ibr) in 2016;101(12):1563-1572. 2016;128(12):195-203.
previously treated chronic lymphocytic 16. Mato A, Nabhan C, Kay N, et al. Real-world 26. Seymour JF, Ma S, Brander DM, et al.
leukemia (CLL): up to four years follow-up clinical experience in the Connect chronic Venetoclax plus rituximab in relapsed or
of the RESONATE study. J Clin Oncol. lymphocytic leukaemia registry: a prospec- refractory chronic lymphocytic leukaemia: a
2017;35(suppl; abstr 7510). tive cohort study of 1494 patients across 199 phase 1b study. Lancet Oncol. 1017;18(2):
7. Maddocks K, Ruppert A, Lozanski G, et al. US centres. Br J Haematol. 2016;175(5):892- 230-240.
Etiology of ibrutinib therapy discontinua- 903. 27. Sharman J, Brander D, Mato A, et al.
tion and outcomes in patients with chronic 17. Bland J, Altman D. Survival probabilities Ublituximab and ibrutinib for previously
lymphocytic leukemia. JAMA Oncol. 2015; (the Kaplan-Meier method). BMJ. treated genetically high-risk chronic lym-
1(1):80-87. 1998;317(7172):1572. phocytic leukemia: results of the GENUINE
8. Woyach J, Ruppert A, Guinn D, et al. 18. Hallek M, Cheson B, Catovsky D, et al. phase 3 study. J Clin Oncol.
BTKC481S-mediated resistance to ibrutinib Guidelines for the diagnosis and treatment 2017;35(Supplemental, ASCO abstract
in chronic lymphocytic leukemia. J Clin of chronic lymphocytic leukemia: a report 7504).
Oncol. 2017;35(13):1437-1443. from the International Workshop on 28. Hillmen P, Rawstron A, Munir T, et al. The
9. O’Brien S, Furman R, Coutre S, et al. Five- Chronic Lymphocytic Leukemia updating initial report of bloodwise tap clarity study
year experience with single-agent ibrutinib the National Cancer Institute-Working combining ibrutinib and venetoclax in
in patients with previously untreated and Group 1996 guidelines. 2008;111(12):5445- relapsed, refractory CLL shows acceptable
relapsed/refractory chronic lymphocytic 5456. safety and promising early indications EHA
leukemia/small Lymphocytic Leukemia. 19. Cheson B, Byrd J, Rai K, et al. Novel targeted Oral Presentation June 22-25, Madrid Spain.
Blood. 2016; 128:233. agents and the need to refine clinical end 2017.

haematologica | 2018; 103(5) 879


ARTICLE Plasma Cell Disorders

A novel nano-immunoassay method for


Ferrata Storti
Foundation quantification of proteins from CD138-purified
myeloma cells: biological and clinical utility
Irena Misiewicz-Krzeminska,1,2,3 Luis Antonio Corchete,1,2 Elizabeta A. Rojas,1,2
Joaquín Martínez-López,4 Ramón García-Sanz,1,2,5 Albert Oriol,7 Joan Bladé,8
Juan-José Lahuerta,6 Jesús San Miguel,9 María-Victoria Mateos1,2,5
and Norma C. Gutiérrez1,2,5
Haematologica 2018 1
Cancer Research Center-IBMCC (USAL-CSIC), Salamanca, Spain, 2Institute of Biomedical
Research of Salamanca (IBSAL), Spain; 3National Medicines Institute, Warsaw, Poland;
Hematology Department, Hospital 12 de Octubre, CNIO, Complutense University,
Volume 103(5):880-889 4

CIBERONC, Madrid, Spain; 5Hospital Universitario de Salamanca, CIBERONC, Spain;


6
Hospital 12 de Octubre, Madrid, Spain; 7Hospital Germans Trias i Pujol, Barcelona, Spain;
8
Hospital Clinic, Barcelona, Spain and 9Clínica Universidad de Navarra, CIMA, IDISNA,
CIBERONC, Pamplona, Spain

ABSTRACT

P
rotein analysis in bone marrow samples from patients with multiple
myeloma has been limited by the low concentration of proteins
obtained after CD138+ cell selection. A novel approach based on
capillary nano-immunoassay could make it possible to quantify dozens
of proteins from each myeloma sample in an automated manner. Here
we present a method for the accurate and robust quantification of the
expression of multiple proteins extracted from CD138-purified multiple
myeloma samples frozen in RLT Plus buffer, which is commonly used for
nucleic acid preservation and isolation. Additionally, the biological and
clinical value of this analysis for a panel of 12 proteins essential to the
pathogenesis of multiple myeloma was evaluated in 63 patients with
Correspondence: newly diagnosed multiple myeloma. The analysis of the prognostic
normagu@usal.es impact of CRBN/Cereblon and IKZF1/Ikaros mRNA/protein showed
that only the protein levels were able to predict progression-free survival
of patients; mRNA levels were not associated with prognosis.
Received: September 27, 2017. Interestingly, high levels of Cereblon and Ikaros proteins were associated
Accepted: January 31, 2018. with longer progression-free survival only in patients who received
Pre-published: March 15, 2018. immunomodulatory drugs and not in those treated with other drugs. In
conclusion, the capillary nano-immunoassay platform provides a novel
opportunity for automated quantification of the expression of more than
doi:10.3324/haematol.2017.181628 20 proteins in CD138+ primary multiple myeloma samples.

Check the online version for the most updated


information on this article, online supplements,
and information on authorship & disclosures:
www.haematologica.org/content/103/5/880 Introduction

Genomics has come to dominate biomedical research in recent years. For exam-
©2018 Ferrata Storti Foundation ple, high-throughput genomic technologies have been used for the comprehensive
Material published in Haematologica is covered by copyright. analysis of multiple myeloma (MM). In particular, gene expression profiling has
All rights are reserved to the Ferrata Storti Foundation. Use of enabled the molecular classification of MM, which is widely used in biological
published material is allowed under the following terms and
conditions:
research.1 However, a knowledge of protein expression is essential for identifying
https://creativecommons.org/licenses/by-nc/4.0/legalcode. therapeutic targets, since proteins are the molecules through which most new
Copies of published material are allowed for personal or inter- drugs achieve their efficacy. The limited amount of sample remaining after plasma
nal use. Sharing published material for non-commercial pur- cell purification means that messenger RNA (mRNA) quantification is still used as
poses is subject to the following conditions:
an indirect measure of protein expression in most cases. However, several studies
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- have shown that protein levels cannot be predicted from mRNA measurements.2
mercial purposes is not allowed without permission in writing Immunohistochemistry and flow cytometry have been used to analyze expres-
from the publisher. sion at the protein level, although to a limited extent. These methods are of great
value and are of proven clinical utility, but they have some limitations that make
them less useful for studying intracellular protein levels. They mostly use directly

880 haematologica | 2018; 103(5)


Nano-scale protein quantification in multiple myeloma

marked antibodies that reduce the sensitivity of detection, extracted separately. After overnight incubation at -20ºC, the pro-
and fewer antibodies are available for these techniques, teins were centrifuged at 13,000 x g for 30 min at 4ºC, and washed
even when used in an indirect assay.3 twice with 70% ice-cold ethanol followed by centrifugation for a
Immunohistochemistry allows only semiquantitative further 10 min. The protein precipitate was dried at 39ºC and dis-
analysis of protein expression, and requires a well-trained solved with 50 μL 0.2 M NaOH for 10 min at room temperature
pathologist to interpret the results. Moreover, neither and 4x WB sample buffer for at least 15 min at room temperature.
technique is able to identify non-specific antibody binding Samples were then denatured at 95ºC for 5 min, cooled to room
to other proteins.3 temperature and stored in aliquots at -80ºC. Before any assay,
Western blotting (WB) remains the “gold standard” tech- samples were heated to room temperature, then kept at 37ºC for
nique for protein characterization in most laboratories. 30 min in order to re-dissolve any protein that had precipitated
However, WB consumes large quantities of reagents, has a during freezing.
low throughput, and requires a great deal of time and
effort involving many laborious manual processing steps. Capillary electrophoresis immunoassay
Moreover, WB only yields semiquantitative data of poor Capillary electrophoresis immunoassay or simple western
repeatability, making it a challenge to go beyond using the analysis was performed using the WESTM machine (ProteinSimple,
assay in discovery research to apply it reliably in the clin- San Jose, CA, USA) in accordance with the manufacturer’s proto-
ical setting.4–6 A further drawback is that it is not always cols. The Total Protein Assay (ProteinSimple) was used to quantify
possible to obtain the quantity of protein extract required the protein concentration. In brief, 5 μL of proteins were loaded on
for WB from primary cancer samples. the plate, separated by size, labeled with a biotin reagent and
MM is a clear prototype of a bone marrow-infiltrating detected by chemiluminescence using streptavidin-horseradish
tumor for which a relatively small quantity of sample is peroxidase. At the end of the run, the proportion of the protein of
available after the diagnostic procedure, which involves interest in the total protein in the sample was measured, in com-
morphological evaluation, immunophenotypic characteri- parison to a standard curve previously generated using JJN3 cell
zation by flow cytometry, and CD138+ plasma cell separa- line extracts of known protein concentrations.
tion for routine fluorescence in situ hybridization analysis. Primary antibodies used in the study and the optimized condi-
The recent development of a method based on the combi- tions for each antibody are presented in Table 1. Data were ana-
nation of capillary nano-electrophoresis with immunoas- lyzed using CompassTM software. Each protein peak was meas-
say (CNIA), also known as ‘simple western’, requires only ured automatically and normalized with respect to the GAPDH
very small amounts of sample to be able to measure pro- median area under the peak. Expression of each protein is present-
tein expression.3,7 This technical advance makes it possible ed as its abundance relative to GAPDH.
to analyze the expression of 50-100 proteins in a single
MM sample. Here we present the results of a pilot study
using this platform in MM patients. The main goal was to
quantify accurately and robustly the proteins extracted
from CD138-purified MM samples frozen in RLT Plus
buffer, which is commonly used as a method for RNA and
DNA preservation. Additionally, we attempted to estab-
lish the clinical value of this analysis using a panel of pro-
teins essential to MM pathogenesis, comparing it with
that of the corresponding RNA expression.

Methods
For more specific information see the Online Supplementary File.

Patients and multiple myeloma cell lines


Sixty-three samples from patients diagnosed with MM
between October 2013 and November 2015 were included in the
study (Online Supplementary Table S1). Forty-three had been
enrolled in two Spanish Myeloma Group clinical trials: GEM2010
[bortezomib/melphalan/prednisone and lenalidomide/dexam-
ethasone in a sequential or alternating manner; (n=24)] and
BenVelPres [bendamustine/bortezomib/prednisone; (n=19)]. The
other 20 patients were not treated as part of a clinical trial.
The impact of RNA and protein expression on patients’ survival
was evaluated only in the group of patients that took part in the
clinical trials (Online Supplementary Table S1). The scheme of the
study is presented in Figure 1.

Protein extraction from RLT Plus buffer


Proteins were extracted by ice-cold acetone precipitation from
RNA-column flow-through liquid. To increase the rate of protein
Figure 1. Scheme of the study.
precipitation 10 mM NaCl was added to the acetone at 80% (v/v).
For technical reasons, each sample was divided into two tubes and

haematologica | 2018; 103(5) 881


I. Misiewicz-Krzeminska et al.

DNA/RNA extraction and quantitative real-time tion and at double and half the indicated concentration. In
polymerase chain reaction analysis the event that the protein evaluated was not present in the
mRNA expression was evaluated by Taqman assay quantitative database, we performed a full optimization, which consist-
real-time polymerase chain reaction (qRT-PCR) analysis using the ed of running the assays in the cell line samples at two con-
respective GAPDH Taqman assay as a control, by the 2-ΔCt method. centrations (0.1 mg/mL and 0.2 mg/mL) with at least five
antibody dilutions in order to determine the optimal con-
Statistical analysis centration at which the antigen-antibody binding was sat-
Spearman correlations were calculated. Progression-free sur- urated and no change in antibody concentration influenced
vival (PFS) was calculated for each gene and protein. Survival the result. The optimized concentrations for each anti-
curves were plotted using the Kaplan–Meier method and statisti- body, the molecular weights at which the peaks were
cal significance was evaluated with the log-rank test. observed, and the coefficients of variation arising from the
validation of each protein are shown in Table 1.
Standard curves were produced for each protein to eval-
Results uate the range of linearity over which the expression of
each protein could be quantified. Briefly, each capillary
Protein extraction from RLT Plus buffer results contained the sample at a different dilution, and the pro-
in optimal quality and quantity tein detection was visualized as virtual blots, as exempli-
Firstly, we evaluated the amount and quality of the pro- fied by the use of Aiolos in Figure 3A. The peaks obtained
tein extracted with our protocol. The data generated by for each dilution, which were obtained automatically by
the WESTM system were visualized as virtual blots (Figure the program, have a distinct height and width, depending
2A) or peaks (Figure 2B) that were quantified as the area on the sample dilution (Figure 3B). Once they had been
under the curve using the inbuilt algorithm of the quantified the standard curve was generated (Figure 3C).
CompassTM software. Using JJN3 myeloma cell line lysates After protein quantification, we compared the value
of known concentration, we generated a protein standard obtained for each sample and each protein with the
curve that proved to be linear over the evaluated range of respective standard curve to ensure correct measurement.
concentrations (Figure 2C). The amount of protein The limit of quantitation was set as signal-to-noise ratio of
obtained from each sample ranged between 0.00 and 0.36 10:1 in accordance with the guidelines from the European
mg protein, with a median quantity of 0.06 mg per sam- Directorate for the Quality of Medicine set out in the
ple. Three of the 63 samples had insufficient material to European Union Pharmacopoeia.8
analyze protein expression (Figure 2D). We compared the The results of Aiolos quantification in six samples are
expression of the various proteins extracted from MM shown in Figure 3D, in which virtual blots for both Aiolos
cells stored in RLT Plus buffer with that obtained using the and GAPDH are presented.
standard RIPA protocol and found the signals to be similar
for the two protocols (Figure 2E,F). Analysis of mRNA and protein expression
We analyzed the expression of 12 genes and their
Optimization of protein quantification by capillary encoded proteins, together with GAPDH as a control
nano-electrophoresis with immunoassay (Figure 1). We decided to select proteins involved in MM
For each analyzed protein, we first searched in the or cancer pathogenesis: Cyclin D1 and Cyclin D2, whose
ProteinSimple antibody database for the optimized condi- overexpression is a unifying event for most MM;9–11 c-myc,
tions (http://www.proteinsimple.com/antibody/antibod- which is consistently found to be involved in the transfor-
ies.html). If the antibody was present, we re-evaluated it in mation of monoclonal gammopathy of undetermined sig-
our system, using the antibody at the indicated concentra- nificance into MM;12,13 HSP90, which is upregulated in

Table 1. Summary of proteins and antibodies used in the study.


Target protein Company Cat number Species Clonality Dilution Ab MW peak Intra-assay CV (%)
1 Aiolos Cell Signaling 12720 Rabbit Polyclonal 1:100 85 10.9
2 Calnexin Enzolifesciences ADI-SPA-860 Rabbit Polyclonal 1:250 119 4.9
3 Cereblon NovusBio NBP1-91810 Rabbit Polyclonal 1:80 59 9.7
4 c-myc Cell Signalling 5605 Rabbit Monoclonal 1:50 75 8.3
5 Cyclin D1 Abcam Ab134175 Rabbit Monoclonal 1:50 40 9.9
6 Cyclin D2 Cell Signaling 3741 Rabbit Monoclonal 1:100 38 7.8
7 DDX21 Abcam Ab182156 Rabbit Monoclonal 1:100 110 11.4
8 HSP90 Cell Signaling 4877 Rabbit Monoclonal 1:50 95 7.0
9 Ikaros Santa Cruz Sc-13039 Rabbit Polyclonal 1:50 70 10.8
10 PSME1 NovusBio NBP1-83121 Rabbit Polyclonal 1:50 37 10.2
11 RIPK1 Cell Signaling 3493 Rabbit Monoclonal 1:50 79 5.8
12 XAF1 Cell Signaling 13805 Rabbit Monoclonal 1:25 46, 110 15.5
13 GAPDH Cell Signaling 2118 Rabbit Monoclonal 1:50 42 8.6
Ab: antibody; MW: molecular weight; CV: coefficient of variation.

882 haematologica | 2018; 103(5)


Nano-scale protein quantification in multiple myeloma

many solid and hematologic malignancies, including median level of expression, and CRBN, RIPK1 and XAF1
MM;14 Calnexin, which forms endoplasmic reticulum and were the least expressed mRNA.
is upregulated in MM relative to normal plasma cells in In general, the expression of mRNA was more homoge-
genetically identical twin samples;15 and DDX21 or RIPK1, nous than that of proteins, as indicated by the higher coef-
with known involvement in several tumors.16–18 In addi- ficients of variation for the latter (Figure 4C). In fact, the
tion, proteins involved in the mechanism of action of coefficients of variation were significantly lower than
antimyeloma drugs were included: Cereblon, Ikaros, those for c-myc, DDX21, HSP90, IKZF1 and PSME1
Aiolos for immunomodulatory drugs; XAF1 for melpha- mRNA than for their respective encoded proteins. The
lan; and PSME1 for bortezomib.19–22 highest variability in expression, both at the mRNA and
At the protein level, PSME1 and Calnexin showed the protein levels, was observed for CCND2/Cyclin D2 and
highest median level of expression, while HSP90 was the CCND1/Cyclin D1, as well as for c-myc protein.
most strongly expressed mRNA (Figure 4A,B). Conversely, Next, we analyzed the correlation between the two lev-
proteins Cyclin D1, Cyclin D2 and c-myc had the lowest els of gene expression, mRNA and protein. Interestingly,

A B D

E F

Figure 2. Optimization of protein extraction from RLT Plus samples. Due to the various additives in the sample buffer there is marked incompatibility with most of
the normally used protein quantification methods. The Total Protein assay was therefore used, as it is insensitive to high SDS concentrations. The standard curve was
generated using JJN3 cell line extracts at 0.25 mg/mL concentration, and serial dilutions thereof. Each capillary contained one sample of a known concentration.
Results were visualized as virtual gels (A) and the numbers correspond to the areas under the curves of the peaks (B). A standard curve was generated, plotting the
result for each capillary quantification (C). Amount of protein obtained from each sample (D). Comparison of results from Calnexin, Cyclin D2, GAPDH and Ikaros quan-
tification in the U266 cell line, from which protein extracts were obtained by RLT Plus and RIPA extraction, and visualized as virtual blots or two distinct dilutions of
the sample (E), and one dilution extracted with both protocols visualized as peaks (F).

haematologica | 2018; 103(5) 883


I. Misiewicz-Krzeminska et al.

only Cyclin D1 and Cyclin D2 protein levels were strongly Influence of mRNA and protein levels on survival
correlated with the respective CCND1 and CCND2 mRNA of myeloma patients
levels (Figure 4D). We observed a modest correlation for Since clinical data were available for 43 MM patients, 24
Aiolos, Calnexin and DDX21 proteins with their respective enrolled in GEM 2010 and 19 in BenVelPres clinical trials,
mRNA. we also performed survival analysis for proteins and
Although the number of proteins analyzed was limited, mRNA using PFS as the endpoint (Table 2). Cereblon and
we examined the correlation between the levels of the dif- Ikaros were the only proteins able to predict PFS.
ferent proteins. A positive correlation was observed Interestingly, mRNA levels of CRBN and IKZF1 were not
between most of the protein pairs (Online Supplementary associated with prognosis (Figure 5). Accordingly, patients
Figure S2). We confirmed the previously described relation- with a high level of Cereblon protein had a longer PFS
ship between Ikaros/Aiolos and c-myc.19 Additionally, c- than those with a low level (50.4 versus 16.3 months,
myc protein expression was positively correlated with P<0.001). Similarly, high levels of Ikaros protein were
Cereblon, Calnexin, and RIPK1, and negatively correlated associated with longer PFS (45.1 versus 17.8 months,
with DDX21 (Online Supplementary Figure S2). We found P<0.01). The levels of two mRNA were associated with
that protein levels of Cereblon, Ikaros and Aiolos, all of longer PFS: a high level of PSME1 (50.4 versus 23.5
which are required for the activity of immunomodulatory months, P<0.05) and a low level of XAF1 (20.3 versus 45.1
drugs, were correlated with each other (Online months, P<0.05).
Supplementary Figure S2). Since Cereblon, Ikaros and Aiolos are involved in the
The potential association between the expression of pro- mechanism of action of immunomodulatory drugs, and
teins and mRNA tested in the study and chromosomal only GEM2010 patients were treated with lenalidomide,
abnormalities was also explored. We confirmed the well- we examined whether the prognostic value of these pro-
established pattern of CCND1/Cyclin D1 and teins was influenced by the type of treatment. Indeed,
CCND2/Cyclin D2 expression in t(11;14) and t(4;14) high levels of Cereblon and Ikaros were both associated
(Online Supplementary Figure S3). A lower level of expression with longer PFS only in patients who received
of PSME1 and RIPK1 proteins in MM with 1q gains, and a immunomodulatory drugs and not in those treated with
higher level of IKZF1 mRNA expression in MM with other drugs (Figure 6).
t(11;14) were also observed.

A B D

Figure 3. Optimization of protein expression quantification. For each protein the standard curve was generated using the sample with the strongest signal to prepare
the serial dilutions. Each dilution was run in a separate capillary. The sample result for Aiolos is visualized as a virtual blot (A) or as peaks (B). The standard curve
established the linear range for each protein (C). The sample result for Aiolos together with the respective GAPDH for each sample was run in separate capillaries
(D). Each peak area was quantified and Aiolos was normalized with respective to GAPDH, as shown in the example.

884 haematologica | 2018; 103(5)


Nano-scale protein quantification in multiple myeloma

Discussion ples after CD138+ separation are usually stored in buffers


such as TRIZOL or RLT Plus, which preserve nucleic acids
MM has been comprehensively studied at the DNA and for subsequent use in genomic studies. While it is techni-
RNA levels using high-throughput technologies such as cally possible to extract proteins from these buffers,23–25 the
microarrays for detecting copy number abnormalities, and quantity of protein would not be sufficient for multiple
gene expression profiling, and, more recently, next-gener- WB to be carried out. In the classic WB, the amount of the
ation sequencing for DNA mutation analysis. MM sam- purified plasma cells, even if all of it were available for the

A C

Figure 4. Two levels of analysis of each gene RNA and protein. mRNA expression of each gene was assessed by qRT-PCR and normalized relative to GAPDH and
expressed as 2-ΔCt (A). Abundance of each protein was assessed by CNIA and normalized relative to GAPDH abundance in each case (B). The Y axis of graphs (A) and
(B) are expressed on a log scale. The variability of each mRNA and protein measurement in the analyzed population of patients with MM, measured as percentage
coeffcient of variation (CV%). The threshold of statistical significance (*P<0.05) was determined as described in the Methods section (C). Spearman correlation coef-
ficient for each mRNA/protein pair ranked by increasing P value (D).

haematologica | 2018; 103(5) 885


I. Misiewicz-Krzeminska et al.

Table 2. Univariate analysis of progression-free survival.


mRNA HR Group n Median PFS (months) P-value Protein HR Group n Median PFS (months) P-value
(CI) (CI)
IKZF3 1.84 H 10 21.7 0.19 Aiolos 0.51 H 33 45.1 0.16
(0.74-4.6) L 33 45.1 (0.19-1.34) L 10 23.5
CANX1 0.44 H 21 50.4 0.074 Calnexin 1.82 H 22 30 0.2
(0.18-1.11) L 22 23.5 (0.72-4.6) L 18 43.8
CRBN 0.49 H 10 50.4 0.24 Cereblon 0.23 H 31 50.4 0.00034
(0.14-1.67) L 33 29.8 (0.09-0.55) L 12 16.3
CCND1 2.55 H 27 29.8 0.066 CyclinD1 1.55 H 23 30 0.33
(0.91-7.14) L 16 NR (0.64-3.78) L 20 NR
CCND2 0.38 H 17 NR 0.053 CyclinD2 0.24 H 11 NR 0.051
(0.13-1.05) L 26 29.8 (0.05-1.03) L 32 29.8
MYC 1.54 H 33 30 0.44 c-myc 0.63 H 27 45.1 0.31
(0.51-4.64) L 10 45.1 (0.26-1.56) L 13 29.8
DDX21 0.49 H 29 43.8 0.1 Ddx21 0.47 H 14 50.4 0.18
(0.2-1.17) L 14 23.5 (0.15-1.46) L 23 30
HSP90 0.52 H 24 50.4 0.14 Hsp90 1.5 H 12 29.8 0.44
(0.22-1.25) L 19 23.5 (0.53-4.27) L 25 50.4
IKZF1 1.93 H 25 29.8 0.15 Ikaros 0.31 H 32 45.1 0.0075
(0.77-4.83) L 18 43.8 (0.12-0.77) L 11 17.8
PSME1 0.33 H 18 50.4 0.018 Psme1 0.48 H 30 45.1 0.13
(0.12-0.86) L 25 23.5 (0.19-1.26) L 10 16.3
RIPK1 0.52 H 28 50.4 0.13 Ripk1 1.41 H 11 30 0.49
(0.22-1.24) L 15 23.5 (0.53-3.73) L 29 45.1
XAF1 2.39 H 11 20.3 0.048 Xaf1 1.56 H 15 30 0.34
(0.98-5.8) L 32 45.1 (0.63-3.85) L 25 45.1
HR: hazard ratio; CI: confidence interval; PFS: progression-free survival; H: high level; L: low level.

protein studies, would have allowed at most six proteins concentration of antibodies used by the CNIA platform is
to be evaluated, the median amount of sample being suf- higher than that used in WB, lower amounts of antibody
ficient to analyze two proteins. are required because of the small volume of antibody.
Here we present for the first time a method for quanti- Comparing the signal detected by each antibody when
fying the expression of multiple proteins from myeloma the sample was extracted by the RIPA method with that
cells stored in the buffer commonly used for nucleic acid obtained using our protocol revealed no significant differ-
preservation and isolation. We report a protocol for pro- ences for any of the proteins evaluated, which supports
tein extraction from MM cells stored in RLT Plus buffer the suitability of the present protocol for extracting pro-
based on a well-known acetone precipitation proce- teins from RLT Plus buffer. We observed differences
dure.26,27 We decided to add NaCl to the sample before pre- between the predicted molecular weight and that detected
cipitation, since a greater inorganic salt content is known by the CNIA system for some proteins, regardless of the
to improve protein yield.28 After testing several types of extraction method. The most probable explanation for
salts and concentrations, we chose the one with the best this phenomenon is that migration depends on the mobil-
performance. We also assessed several methods of protein ity in the matrix.31 In fact, each system provides a unique
pellet dissolution, finally settling for a 0.2 M NaOH and 4x molecular weight value that depends on the particular
WB sample buffer, since slight changes in the pH of the interactions between the matrix and protein, and the true
environment change protein solubility.26,29 molecular weight can only be determined by mass spec-
In contrast to the classic WB, which provides only semi- trometry or sequencing.32 In our assay, we analyzed only
quantitative (blot-based) results, CNIA quantifies the area the data obtained from the antibodies that detected peaks
under the curve of the signal in each capillary, enabling whose signal intensity was linear in the serial dilutions,
expression relative to the control protein to be calculat- enabling the frequent biases in WB to be eliminated.
ed.3,30 To determine whether the CNIA method is suitable Although such standard curves are not usually calculated
for evaluating the expression of multiple proteins, using as part of the WB method, this is highly recommendable.33
various antibodies, we tested the performance of each Even though, at first glance, the optimization step
protein in the WESTM system. We first optimized the con- required for each antibody seems laborious, one CNIA run
centration of the antibody to be employed using RIPA- allows 24 samples to be analyzed, so it would be possible
extracted proteins from MM cell lines, as suggested by the to optimize, for example, four antibodies in 3 hours.
system provider. The antibody dilution used has to be the Therefore, bearing in mind the subsequent possibility of
one that saturates the epitope-antibody binding, so that quantifying protein abundances, it is not such a time-con-
the additional increase in antibody concentration would suming process.
not have caused the increase in the signal. Although the After demonstrating that our approach accurately quan-

886 haematologica | 2018; 103(5)


Nano-scale protein quantification in multiple myeloma

A B

Figure 5. Progression-free sur-


vival according to levels of
MRNA and protein expression.
Progression-free survival in
patients with low and high lev-
els of mRNA (A) and protein (B)
expression. The log-rank test
was performed for each gene
and protein and Kaplan-Meier
curves represent the PFS of
MM patients depending on
mRNA and protein status.
Cutoff Finder software
(http://molpath.charite.de/cut
off) was used to obtain the
optimal cutoff, which was
defined as that producing the
most significant split that dis-
criminates between good and
poor survival by examining all
the possible cutoffs using the
log-rank test.

tified the proteins extracted at the same time as the DNA Aiolos have been quantified and the relationship between
and RNA from the RLT Plus buffer, we investigated the their expressions analyzed in MM. In T-cell acute lym-
applicability of the method to the analysis of the expres- phoblastic leukemia, for example, the levels of mRNA
sion of key proteins in MM biology, such as D Cyclins, c- encoding Ikaros and Aiolos were weakly, but significantly
myc, Cereblon, Ikaros, and Aiolos, among others. We also correlated.37
wanted to compare protein expression with the corre- Among the proteins included in our study, we observed
sponding mRNA level, since many basic studies have a significant association between protein level and PFS for
revealed that only 30-40% of protein abundance can be Cereblon and Ikaros, while this association was not
explained by the mRNA level.34 Our results showed a observed for the respective mRNA levels. Cereblon forms
moderate or low correlation between mRNA and protein an E3 ubiquitin ligase complex together with the damaged
levels of expression, and are consistent with the general DNA binding protein 1 (DDB1), Cullin4A (CUL4) and
observation that ~60% of the variation in protein concen- Roc1. Immunomodulatory drugs, such as lenalidomide or
tration cannot be explained by measuring mRNA alone.34 pomalidomide, bind to Cereblon in a region located at the
We also observed that the mRNA level was less variable C-terminus of this protein.38,39 Our results did not demon-
than protein expression among MM patients for all the strate a correlation between Cereblon protein and mRNA
mRNA and protein pairs analyzed. Indeed, protein abun- level, and showed that only high levels of Cereblon pro-
dance is regulated by a variety of complex mechanisms, tein were associated with a good prognosis in MM. These
such as post-transcriptional and post-translational modifi- findings are concordant with those of previous studies and
cations, and protein degradation regulation.34,35 By measur- support the usage of protein expression to evaluate
ing mRNA abundance, only the early steps in a long chain Cereblon levels.40
of regulatory events are considered.36 However, the Several independent groups have identified Ikaros and
mRNA level is still often employed as a proxy for protein Aiolos as the downstream targets of Cereblon after
abundance, mostly because of the lack of appropriate immunomodulatory drug activation.41–43 The role of the
technology to quantify proteins quickly and efficiently. level of Ikaros in MM survival is controversial. When
Our results reproduce the well-known pattern of Ikaros expression was investigated at the RNA level, a low
CCND1/Cyclin D1 and CCND2/Cyclin D2 expression in level of mRNA IKZF1 expression was associated with bet-
MM with t(11;14) and t(4;14).9 We also found a correla- ter prognosis in newly diagnosed patients treated with
tion between c-myc and Ikaros and Aiolos levels, analyz- immunomodulatory drugs.44 On the other hand, low
ing either mRNA or protein expression, consistent with IKZF1 levels were found to predict a lack of responsive-
the previously demonstrated regulation of c-myc by ness to immunomodulatory drugs and a shorter overall
Ikaros and Aiolos in MM.19 Interestingly, the correlation survival in refractory MM patients.45 We observed that a
between Ikaros and Aiolos levels was stronger for the high level of Ikaros protein was associated with longer
protein than for the mRNA. To our knowledge, this is the PFS, while no significant impact on prognosis was
first time that the protein levels of c-myc, Ikaros and observed when PFS was estimated from mRNA levels.

haematologica | 2018; 103(5) 887


I. Misiewicz-Krzeminska et al.

Figure 6. Progression-free sur-


vival in patients with low and
high Cereblon, Aiolos and
Ikaros protein levels, depend-
ing on the treatment scheme
(only patients treated accord-
ing to GEM2010 trial received
lenalidomide). The log-rank
test was performed for each
protein and Kaplan-Meier
curves represent progression-
free survival of MM patients
depending on protein status.
Cutoff Finder software
(http://molpath.charite.de/cut
off) was used to obtain the
optimal cutoff, which was
defined as that producing the
most significant split that dis-
criminates between good and
poor survival by examining all
the possible cutoffs using the
log-rank test.

These results are consistent with the longer survival dis- amount of material means that, for the first time, more
played by relapsed/refractory MM patients treated with than 20 proteins can be analyzed using the same sample
lenalidomide who expressed high levels of IKZF1/3 pro- frozen for DNA and RNA analysis. This makes the CNIA
tein, as evaluated by immunohistochemical staining.46 platform a fast, effective and accurate tool for exploring
Although the number of patients analyzed in this the impact of different proteins on the survival of patients
study is relatively small, the survival analysis carried out with MM and for investigating new protein biomarkers
dividing patients according to drug therapy showed that that could help to predict the response to new drugs that
high levels of Cereblon and Ikaros proteins are associat- directly target specific proteins. These encouraging results
ed with a longer PFS only in patients who receive require further validation in a larger cohort of patients
immunomodulatory drugs and not in those who are with MM or other hematologic malignancies.
treated with other drugs. Other studies reached the same
conclusion that the level of Cereblon can predict the out- Acknowledgments
come of patients with MM mainly in those treated with The authors thank Isabel Isidro, Teresa Prieto and Vanesa
immunomodulatory drugs.47–50 By contrast, in the present Gutierrez for their technical assistance.
series of MM patients, the level of Aiolos did not influ-
ence the outcome of the patients treated with Funding
immunomodulatory drugs. This work was funded by a grant from the International
In summary, we present the implementation of a novel Myeloma Foundation's Black Swan Research Initiative® and
technique based on capillary nano-immunoassay for “Gerencia Regional de Salud, Junta de Castilla y León”
quantifying protein expression in MM samples in the clin- (BIO/SA35/14). WES™ platform was acquired thanks to
ical setting. The requirement for only a relatively small INNOCAMPUS Program (CEI10-1-0010).

888 haematologica | 2018; 103(5)


Nano-scale protein quantification in multiple myeloma

Bortezomib treatment sensitizes oncolytic 36. Payne SH. The utility of protein and mRNA
References HSV-1-treated tumors to NK cell correlation. Trends Biochem Sci. 2015;40
immunotherapy. Clin Cancer Res. 2016;22 (1):1–3.
1. Broyl A, Hose D, Lokhorst H, et al. Gene (21):5265–5276. 37. Mitchell JL, Yankee TM. Variations in
expression profiling for molecular classifica- 19. Bjorklund CC, Lu L, Kang J, et al. Rate of mRNA and protein levels of Ikaros family
tion of multiple myeloma in newly diag- CRL4(CRBN) substrate Ikaros and Aiolos members in pediatric T cell acute lym-
nosed patients. Blood. 2010;116(14):2543– degradation underlies differential activity of phoblastic leukemia. Ann Transl Med.
2553. lenalidomide and pomalidomide in multiple 2016;4(19):363–363.
2. Tian Q, Stepaniants SB, Mao M, et al. myeloma cells by regulation of c-Myc and 38. Ito T, Ando H, Suzuki T, et al. Identification
Integrated genomic and proteomic analyses IRF4. Blood Cancer J. 2015;5e354. of a primary target of thalidomide terato-
of gene expression in mammalian cells. Mol 20. Kortüm KM, Zhu YX, Shi CX, Jedlowski P, genicity. Science. 2010;327(5971):1345–
Cell Proteomics. 2004;3(10):960–969. Stewart AK. Cereblon binding molecules in 1350.
3. Chen J-Q, Wakefield LM, Goldstein DJ. multiple myeloma. Blood Rev. 39. Ito T, Ando H, Handa H. Discovery of the
Capillary nano-immunoassays: advancing 2015;29(5):329–334. target for immunomodulatory drugs
quantitative proteomics analysis, biomarker 21. Park GB, Kim Y-S, Kim D, et al. Melphalan- (IMiDs). Rinsho Ketsueki. 2016;57(5):556–
assessment, and molecular diagnostics. J induced apoptosis of EBV-transformed B 562.
Transl Med. 2015;13:182. cells through upregulation of TAp73 and 40. Gandhi AK, Mendy D, Waldman M, et al.
4. Chen J-Q, Heldman MR, Herrmann MA, et XAF1 and nuclear import of XPA. J Measuring cereblon as a biomarker of
al. Absolute quantitation of endogenous Immunol. 2013;191(12):6281–6291. response or resistance to lenalidomide and
proteins with precision and accuracy using a 22. Dytfeld D, Luczak M, Wrobel T, et al. pomalidomide requires use of standardized
capillary western system. Anal Biochem. Comparative proteomic profiling of refrac- reagents and understanding of gene com-
2013;442(1):97–103. tory/relapsed multiple myeloma reveals bio- plexity. Br J Haematol. 2014;164(2):233–244.
5. Ghosh R, Gilda JE, Gomes AV. The necessity markers involved in resistance to borte- 41. Lu G, Middleton RE, Sun H, et al. The
of and strategies for improving confidence in zomib-based therapy. Oncotarget. myeloma drug lenalidomide promotes the
the accuracy of western blots. Expert Rev 2016;7(35):56726-56736. cereblon-dependent destruction of Ikaros
Proteomics. 2014;11(5):549–560. 23. Chomczynski P. A reagent for the single- proteins. Science. 2014;343(6168):305–309.
6. Gassmann M, Grenacher B, Rohde B, Vogel step simultaneous isolation of RNA, DNA 42. Krönke J, Udeshi ND, Narla A, et al.
J. Quantifying western blots: pitfalls of den- and proteins from cell and tissue samples. Lenalidomide causes selective degradation
sitometry. Electrophoresis. 2009;30(11): BioTechniques. 1993;15(3):532–534, 536– of IKZF1 and IKZF3 in multiple myeloma
1845–1855. 537. cells. Science. 2014;343(6168):301–305.
7. Fan AC, Deb-Basu D, Orban MW, et al. 24. Vorreiter F, Richter S, Peter M, Baumann S, 43. Zhu YX, Braggio E, Shi C-X, et al. Cereblon
Nanofluidic proteomic assay for serial analy- von Bergen M, Tomm JM. Comparison and expression is required for the antimyeloma
sis of oncoprotein activation in clinical spec- optimization of methods for the simultane- activity of lenalidomide and pomalidomide.
imens. Nat Med. 2009;15(5):566–571. ous extraction of DNA, RNA, proteins, and Blood. 2011;118(18):4771–4779.
8. Technical guide for the eleboration of mono- metabolites. Anal Biochem. 2016;50825–33. 44. Krönke J, Kuchenbauer F, Kull M, et al.
graphs. 25. Mathieson W, Thomas GA. Simultaneously IKZF1 expression is a prognostic marker in
9. Bergsagel PL, Kuehl WM, Zhan F, Sawyer J, extracting DNA, RNA, and protein using newly diagnosed standard-risk multiple
Barlogie B, Shaughnessy J. Cyclin D dysreg- kits: is sample quantity or quality preju- myeloma treated with lenalidomide and
ulation: an early and unifying pathogenic diced? Anal Biochem. 2013;433(1):10–18. intensive chemotherapy: a study of the
event in multiple myeloma. Blood. 26. Pace CN, Treviño S, Prabhakaran E, Scholtz German Myeloma Study Group (DSMM).
2005;106(1):296–303. JM. Protein structure, stability and solubility Leukemia. 2017;31(6):1363-1367.
10. Chesi M, Bergsagel PL. Molecular pathogen- in water and other solvents. Philos Trans R 45. Zhu YX, Braggio E, Shi C-X, et al.
esis of multiple myeloma: basic and clinical Soc Lond B Biol Sci. 2004;359(1448):1225- Identification of cereblon-binding proteins
updates. Int J Hematol. 2013;97(3):313–323. 1234; discussion 1234-1235. and relationship with response and survival
11. Misiewicz-Krzeminska I, Sarasquete ME, 27. Race J. The determination of blood-proteins after IMiDs in multiple myeloma. Blood.
Vicente-Dueñas C, et al. Post-transcriptional by acid-acetone. Biochem J. 2014;124(4):536–545.
modifications contribute to the upregulation 1932;26(5):1571–1584. 46. Pourabdollah M, Bahmanyar M, Atenafu
of Cyclin D2 in multiple myeloma. Clin 28. Crowell AMJ, Wall MJ, Doucette AA. EG, Reece D, Hou J, Chang H. High IKZF1/3
Cancer Res. 2016;22(1):207–217. Maximizing recovery of water-soluble pro- protein expression is a favorable prognostic
12. Chiecchio L, Dagrada GP, Protheroe RKM, teins through acetone precipitation. Anal factor for survival of relapsed/refractory
et al. Loss of 1p and rearrangement of MYC Chim Acta. 2013;79648–54. multiple myeloma patients treated with
are associated with progression of smoulder- 29. Cutler P, editor. Protein Purification lenalidomide. J Hematol Oncol. 2016;9
ing myeloma to myeloma: sequential analy- Protocols. 2nd. Totowa, NJ: Humana Press; (1):123.
sis of a single case. Haematologica. 2004. 484 p. 47. Bila J, Sretenovic A, Jelicic J, et al. Prognostic
2009;94(7):1024–1028. 30. Rustandi RR, Loughney JW, Hamm M, et al. significance of cereblon expression in
13. Chng W-J, Huang GF, Chung TH, et al. Qualitative and quantitative evaluation of patients with multiple myeloma. Clin
Clinical and biological implications of MYC SimonTM, a new CE-based automated Lymphoma Myeloma Leuk. 2016;16(11):
activation: a common difference between Western blot system as applied to vaccine 610–615.
MGUS and newly diagnosed multiple development. Electrophoresis. 2012;33(17): 48. Jung S-H, Choi H-J, Shin M-G, et al.
myeloma. Leukemia 2011;25(6):1026–1035. 2790–2797. Thalidomide-based induction regimens are
14. Zhang L, Fok JHL, Davies FE. Heat shock 31. Copeland RA. Methods for Protein Analysis. as effective as bortezomib-based regimens
proteins in multiple myeloma. Oncotarget. Boston, MA: Springer US. in elderly patients with multiple myeloma
2014;5(5):1132–1148. 32. Strupat K. Molecular weight determination with cereblon expression. Ann Hematol.
15. Munshi NC, Hideshima T, Carrasco D, et al. of peptides and proteins by ESI and MALDI. 2016;95(10):1645–1651.
Identification of genes modulated in multi- Meth Enzymol. 2005;405:1–36. 49. Huang S-Y, Lin C-W, Lin H-H, et al.
ple myeloma using genetically identical twin 33. McDonough AA, Veiras LC, Minas JN, Ralph Expression of cereblon protein assessed by
samples. Blood. 2004;103(5):1799–1806. DL. Considerations when quantitating pro- immunohistochemicalstaining in myeloma
16. Cimino D, Fuso L, Sfiligoi C, et al. tein abundance by immunoblot. Am J Physiol cells is associated with superior response of
Identification of new genes associated with Cell Physiol. 2015;308(6):C426-433. thalidomide- and lenalidomide-based treat-
breast cancer progression by gene expres- 34. Vogel C, Marcotte EM. Insights into the reg- ment, but not bortezomib-based treatment,
sion analysis of predefined sets of neoplastic ulation of protein abundance from proteom- in patients with multiple myeloma. Ann
tissues. Int J Cancer. 2008;123(6):1327–1338. ic and transcriptomic analyses. Nat Rev Hematol. 2014;93(8):1371–1380.
17. Schneider AT, Gautheron J, Feoktistova M, Genet. 2012;13(4):227–232. 50. Broyl A, Kuiper R, van Duin M, et al. High
et al. RIPK1 suppresses a TRAF2-dependent 35. Liu Y, Beyer A, Aebersold R. On the cereblon expression is associated with better
pathway to liver cancer. Cancer Cell. Dependency of cellular protein levels on survival in patients with newly diagnosed
2017;31(1):94–109. mRNA abundance. Cell. 2016;165(3):535– multiple myeloma treated with thalidomide
18. Yoo JY, Jaime-Ramirez AC, Bolyard C, et al. 550. maintenance. Blood. 2013;121(4):624–627.

haematologica | 2018; 103(5) 889


ARTICLE Plasma Cell Disorders

Impact of extramedullary disease in patients


Ferrata Storti
Foundation with newly diagnosed multiple myeloma
undergoing autologous stem cell
transplantation: a study from the Chronic
Malignancies Working Party of the EBMT
Nico Gagelmann,1 Diderik-Jan Eikema,2 Simona Iacobelli,3 Linda Koster,2
Hareth Nahi,4 Anne-Marie Stoppa,5 Tamás Masszi,6 Denis Caillot,7
Haematologica 2018 Stig Lenhoff,8 Miklos Udvardy,9 Charles Crawley,10 William Arcese,11
Clara Mariette,12 Ann Hunter,13 Xavier Leleu,14 Martin Schipperus,15
Michel Delforge,16 Pietro Pioltelli,17 John A. Snowden,18 Maija Itälä-Remes,19
Volume 103(5):890-897
Maurizio Musso,20 Anja van Biezen,2 Laurent Garderet21 and Nicolaus Kröger1

1
Department of Stem Cell Transplantation, University Medical Center Hamburg-
Eppendorf, Hamburg, Germany; 2EBMT Data Office, Leiden, the Netherlands;
3
Dipartimento di Biologia, Università degli Study di Roma “Tor Vergata”, Italy; 4Center for
Hematology and Regenerative Medicine, Karolinska Institutet, Stockholm, Sweden;
5
Institut Paoli Calmettes, Marseille, France; 6St. István and St. László Hospital,
Budapest, Hungary; 7Hématologie Clinique, Dijon University Hospital, Dijon, France;
8
Department of Hematology, Skane University Hospital Lund, Sweden; 9Department of
Hematology, Bone Marrow Transplant Unit, Debrecen Medical University, Hungary;
10
Department of Haematology, Cambridge University Hospitals, UK; 11University Tor
Vergata, Roma, Italy; 12Department of Hematology, Grenoble University Hospital, France;
13
Leicester Royal Infirmary, Leicester; 14Hematology, Hôpital La Mileterie, Poitiers,
France; 15Haga Teaching Hospital, the Hague, the Netherlands; 16Department of
Hematology, UZ Leuven, Belgium; 17Hematology, Ospedale San Gerardo ASST Monza-
Università degli Studi di Milano Bicocca, Monza, Italy; 18Department of Haematology,
Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK; 19Turku University
Hospital, Finland; 20Division of Hematology, Casa di Cura "La Maddalena", Palermo, Italy
and 21Hopital St Antoine, Paris, France

ABSTRACT
Correspondence:

W
e investigated extramedullary disease in newly diagnosed mul-
nkroeger@uke.uni-hamburg.de tiple myeloma patients and its impact on outcome following
first-line autologous stem cell transplantation. We identified
3744 adult myeloma patients who received up-front single (n=3391) or
tandem transplantation (n=353) between 2005 and 2014 with available
Received: August 15, 2017. data on extramedullary involvement at diagnosis. The overall incidence
Accepted: January 26, 2018. of extramedullary disease was 18.2% (n=682) and increased per year
Pre-published: February 1, 2018. from 6.5% (2005) to 23.7% (2014). Paraskeletal involvement was found
in 543 (14.5%) and extramedullary organ involvement in 139 (3.7%).
More patients with extramedullary organ involvement had multiple
doi:10.3324/haematol.2017.178434 involved sites (≥2; P<0.001). In a comparison of patients with single sites
with patients without the disease, up-front transplantation resulted in at
Check the online version for the most updated least similar 3-year progression-free survival (paraskeletal: P=0.86, and
information on this article, online supplements, extramedullary organ: P=0.88). In single paraskeletal involvement, this
and information on authorship & disclosures: translated less clearly into worse 3-year overall survival (P=0.07) while
www.haematologica.org/content/103/5890 single organ involvement was significantly worse (P=0.001). Multiple
organ sites were associated with worse outcome (P<0.001 and P=0.01).
First-line treatment with tandem compared with single transplantation
©2018 Ferrata Storti Foundation resulted in similar survival in patients with extramedullary disease at
Material published in Haematologica is covered by copyright. diagnosis (P=0.13 for both).
All rights are reserved to the Ferrata Storti Foundation. Use of
published material is allowed under the following terms and
conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode. Introduction
Copies of published material are allowed for personal or inter-
nal use. Sharing published material for non-commercial pur- Multiple myeloma (MM) accounts for approximately 2% of all new cancer cases
poses is subject to the following conditions:
and 13% of hematologic cancers with an age-adjusted incidence of 6 per 100,000
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- per year in the USA and Europe.1 Autologous stem cell transplantation (ASCT) and
mercial purposes is not allowed without permission in writing the development of new agents have considerably increased the median survival of
from the publisher. MM patients.2 The disease is characterized by a clonal proliferation of malignant
plasma cells with a strong dependence on the bone marrow (BM) microenviron-
ment.3

890 haematologica | 2018; 103(5)


Autologous SCT for extramedullary myeloma

However, in some MM patients, myeloma cells escape and response. Overall survival was defined as the time from
the BM, resulting in extramedullary disease (EMD), ASCT to death from any cause or last follow up. Non-relapse
which can be further characterized by two different types mortality was defined as death without evidence of relapse or
of involvement: 1) paraskeletal (PS), consisting of masses progression, with relapse or progression as competing events.
that arose from bone lesions; and 2) extramedullary organ Remission, progression and relapse were defined according to
involvement (EM), resulting from hematogenous spread standard EBMT criteria.19
into different organs, skin and lymph nodes.4,5 At the time On the basis of type of extramedullary involvement, we
of MM diagnosis, the incidence of EM involvement in defined three groups of myeloma patients: 1) without EMD
observational studies ranges from 1.7% to 4.5 using a (MM group); 2) with paraskeletal (PS group); and 3)
baseline staging that includes whole-body magnetic reso- extramedullary organ involvement (EM group). In addition, we
nance imaging (MRI) or positron emission tomography- determined and analyzed the impact of the number of involved
computed tomography (PET-CT).6 Paraskeletal involve- sites as one or multiple (≥ 2) sites. Disease stage at diagnosis was
ment is more frequent and varies from 7% to 34.2% due determined according to the International Staging System (ISS; I-
to different definitions and access of sensitive imaging III),20 Salmon and Durie stages I, II or III, and also according to
techniques.7-10 Rates are also considered to be higher at renal function A or B.21 Performance status at ASCT was
relapse or after surgery.11,12 Several studies reported that assessed with the Karnofsky score (≤80 indicating poor and >80
EMD was associated with shorter survival rates, and thus good status).22 Categorical variables were compared with the use
considered EMD as a high-risk feature. However, the evi- of the Fisher’s exact test or the χ² test. Continuous variables
dence of the effect of EMD at diagnosis is limited due to were analyzed using the Kruskal-Wallis test for independent
small populations, heterogenous patient or intervention samples.
selection, and relapse settings.13-16 Therefore, very limited Survival probabilities were estimated by the Kaplan-Meier
data are available to assess the role of EMD at diagnosis method,23 and the Log-Rank test was used for univariate com-
of MM patients after up-front ASCT. This lack of evi- parison. Median follow up was calculated according to the
dence is striking, since ASCT is standard therapy in first- reverse Kaplan-Meier method.24 Outcomes were artificially cen-
line therapy in eligible patients.17,18 sored at three years. We used cumulative incidence analysis to
Therefore, the objective of this study was to determine assess NRM, and labeled death from relapse as a competing
the demographic and clinical characteristics of EMD in event.25,26 The proportional hazards assumption was verified
MM patients at diagnosis and to evaluate its impact on using graphical methods. Scaled Schoenfeld27 residuals and
outcome after up-front ASCT as first-line therapy. For graphical checks proposed by Klein and Moeschberger28 were
this purpose, we analyzed 3744 patients with or without performed to find evidence of violations. To minimize the effect
EMD at diagnosis after up-front single or tandem ASCT of selection bias, we used a landmark analysis at six months
who had been reported to the European Society for Blood whenever single and tandem ASCT were compared.
and Marrow Transplantation (EBMT) registry between To assess the multivariate effect of factors on each end point,
2005 and 2014. we used the Cox proportional hazards model to estimate hazard
ratios (HR).29 Only complete cases were included in the analysis.
All tests were two-sided, with the type I error rate fixed at
Methods a=0.05. All analyses were performed using the statistical soft-
ware R, v.3.1.0 (R Foundation for Statistical Computing, Vienna,
Study design and data collection Austria) and SPSS Statistics 23 (SPSS, IBM Corp, Armonk, NY,
We included adult patients with MM who had available data USA).
on extramedullary involvement at time of diagnosis who
received an up-front single ASCT within 12 months of diagnosis
or a tandem ASCT within six months from first ASCT as first- Results
line therapy and who had been reported to the EBMT registry
between January 2005 and December 2014. Patients were con- Incidence and sites
sidered eligible for analysis if there were full data available on Among the 3744 patients identified in the registry,
extramedullary involvement (yes or no) at time of diagnosis, its 14.5% (n=543) had paraskeletal involvement (PS group)
location, and the number of sites. This study was performed in and 3.7% (n=139) extramedullary organ involvement (EM
accordance with the principles of the Declaration of Helsinki group), while 81.8% (n=3062) had no EMD (MM group).
and was approved by the Chronic Malignancies Working Party Between 2005 and 2014, the EMD incidence per year
of the EBMT. The EBMT is a non-profit, scientific society repre- increased from 6.5% to 23.7%.
senting more than 600 transplant centers, mainly in Europe. Within the EM group, the involved sites were: kidney
Data are entered, managed, and maintained in a central database (27.3%, n=38), skin (23.0%, n=32), lymph nodes (17.3%,
with internet access. Audits are routinely performed to deter- n=24), central nervous system (CNS; 10.1%, n=14), lung
mine the accuracy of the data. Data on extramedullary involve- and respiratory tract (6.5%, n=9), gastrointestinal tract
ment were extracted from the database using Med-B forms. (GI) and liver (5.8%, n=8), pleura and heart (5.0%, n=7),
Patients whose transplant data are reported provided informed and spleen, ovaries and testes (5.0%, n=7). Most patients
consent to use the information for research purposes and data with EMD (93.5%, n=639) presented with one involved
are anonymized. site (PS1 and EM1), 5.7% (n=36) had two sites, 0.7%
(n=5) had three sites, while four and five sites were pres-
Definitions and statistical analysis ent in 0.1% (n=1) of patients, respectively. Notably, with-
The primary end point was 3-year progression-free survival in the PS group, all 19 patients with multiple (≥2) sites
(PFS), which was defined as the time from ASCT to disease pro- had only additional paraskeletal involvement (PS2), while
gression or death from any cause. The secondary end points further involvement in all 24 EM patients was also
were 3-year overall survival (OS), non-relapse mortality (NRM) restricted to other organs (EM2).

haematologica | 2018; 103(5) 891


N. Gagelmann et al.

Table 1. Patients’, disease and transplantation characteristics.


Patients without EMD Patients with EMD
Characteristic MM group PS group EM group Total P
N. of patients (%) 3062 (81.8) 543 (14.5) 139 (3.7) 3744
Sex, n. (%)
Female 1279 (41.8) 240 (44.2) 57 (41.0) 1576 (42.1) 0.55
Male 1783 (58.2) 303 (55.8) 82 (59.0) 2168 (57.9)
Age at diagnosis in years
Median 59.8 59.8 59.0 0.59
Range 27.4 to 77.7 26.8 to 76.8 31.8 to 72.8
ISS, n. (%)
I 781 (36.9) 158 (38.6) 29 (30.5) 968 (36.9)
II 759 (35.8) 148 (36.2) 29 (30.5) 936 (35.7) 0.11
III 578 (27.3) 103 (25.2) 37 (38.9) 781 (27.4)
Unknown 944 134 44 1122
Renal function, n. (%)
A 2188 (82.7) 410 (83.2) 85 (65.9) 2683 (82.1) <0.001
B 458 (17.3) 83 (16.8) 44 (34.1) 585 (17.9)
Unknown 416 50 10 476
Karnofsky score, n. (%)
Good 1877 (67.3) 344 (67.7) 81 (62.8) 2302 (67.2)
Poor 914 (32.7) 164 (32.3) 48 (37.2) 1126 (32.8) 0.55
Unknown 271 35 10 316
Status at ASCT, n. (%)
CR 580 (19.1) 115 (21.5) 16 (11.7) 711 (19.2)
PR 2262 (74.7) 389 (72.6) 109 (79.6) 2760 (74.6) 0.10
< PR 187 (6.2) 32 (6.0) 12 (8.8) 231 (6.2)
Unknown 33 7 2 42
Type of myeloma, n. (%)
Light chain only 672 (22.1) 122 (22.5) 39 (28.3) 833 (22.4)
Non-secretory 74 (2.4) 29 (5.4) 3 (2.2) 106 (2.9) 0.002
Heavy and light chain 2292 (75.4) 389 (72.0) 96 (69.6) 2777 (74.7)
Unknown 24 3 1 28
Ig-type, n. (%)
G 1648 (70.8) 282 (72.1) 72 (73.5) 2002 (71.1)
A 634 (27.2) 101 (25.8) 22 (22.4) 757 (26.9) 0.51
D/E/M 45 (1.9) 8 (2.0) 4 (4.1) 57 (2.0)
Unknown 735 152 41 928
Light chain type, n. (%)
Kappa 1853 (63.7) 327 (65.1) 73 (55.7) 2253 (63.6) 0.13
Lambda 1054 (36.3) 175 (34.9) 58 (44.3) 1287 (36.4)
Unknown 155 41 8 204
Years of ASCT, n. (%)
0 3062 (100) 3062 (81.8) <0.001
1 524 (96.5) 115 (82.7) 639 (17.1)
≥2 19 (3.5) 24 (17.3) 43 (1.1)
Years of ASCT, n. (%)
< 2009 518 (16.9) 82 (15.1) 25 (18.0) 625 (16.7) 0.001
2009-11 1400 (45.7) 204 (37.6) 62 (44.6) 1666 (44.5)
> 2011 1144 (37.4) 257 (47.3) 52 (37.4) 1453 (38.8)
Time to 1st ASCT in months
Median 6.2 6.2 6.1 0.81
Range 1.1 to 11.9 2.1 to 11.9 3.9 to 11.9
Type of ASCT, n.
Tandem 249 89 15 353
Single 2813 454 124 3391
EMD: extramedullary disease; MM: patients without extramedullary disease; PS: paraskeletal involvement; EM: extramedullary organ involvement; N.: number; ISS: International
Staging System; ASCT: autologous stem cell transplantation; CR: complete remission; PR: partial remission.

892 haematologica | 2018; 103(5)


Autologous SCT for extramedullary myeloma

Figure 1. Progression-free survival (A) and overall survival (B) with numbers at risk of myeloma patients following up-front autologous stem cell transplantation
according to presence of involvement. MM: no extramedullary disease; PS: paraskeletal involvement; EM: extramedullary organ involvement; N: number.

Patients’ and disease characteristics GI/liver (22.5%), and spleen, ovaries and testes (60.0%)
Median age at diagnosis was 59.8 years in both MM and (Table 2).
PS, and 59.0 years in EM patients (P=0.59). In all groups Comparing the MM group without EMD to those with
there were more males (57.9%) than females (42.1%). EMD, one involved site resulted in a similar 3-year PFS of
More EM patients (34.1%) had worse renal function (stage 49.4% (95%CI: 44.6-54.3; P=0.36) while multiple involved
B) in comparison to PS (16.8%) and MM patients (17.3%; sites showed a worse PFS of 22.7% (95%CI: 5.2-40.2;
P<0.001). Patients with EM involvement (28.3%) were P=0.001) (Figure 2A). Both one and multiple involved sites
more likely to have light chain disease compared to PS showed worse 3-year OS rates of 73.5% (95%CI: 69.2-
(22.5%) and MM patients (22.1%; P=0.002). More detailed 77.7; P<0.001) and 71.4% (95%CI: 55.1-87.7; P=0.05) in
patients’ characteristics are listed in Table 1. comparison to patients without EMD (80.1%) (Figure 2B).
After stratification of EMD groups according to one
Transplantation characteristics and responses versus multiple involved sites (PS1 vs. PS2, and EM1 vs.
The source of stem cells for all patients was peripheral EM2), PS patients showed no significant difference in 3-
blood. A total of 3391 patients underwent up-front single year PFS of 50.5% (95%CI: 45.0-55.9%) versus 36.0%
ASCT and 353 patients up-front tandem ASCT; there was (95%CI: 5.2-66.8%; P=0.71), and OS of 77.2% (95%CI:
no difference in time to first ASCT between the two 72.7-81.7%) versus 91.7% (95%CI: 76.0-100; P=0.27). In
groups (P=0.81). Complete remission (CR) before the first EM patients, this comparison resulted in a significantly
ASCT was reported in 21.5% PS, 11.7% EM and 19.1% worse 3-year PFS of multiple sites in the univariate analy-
MM patients; partial remission (PR) was achieved by sis: 44.7% (95%CI: 34.1-55.3%) versus 13.9% (95%CI: 0-
72.6% PS, 79.6% EM and 74.7% MM patients (P=0.1) 35.5%; P=0.03) (Figure 3A). In contrast, 3-year OS was
(Table 1). After ASCT, complete response was achieved by 58.7% (95%CI: 47.9-69.5%) for EM1 versus 57.5%
41.6% PS, 36.1% EM and 43.9% MM patients, while (95%CI: 34.2-80.8%; P=0.51) (Figure 3B).
54.0% PS, 51.9% EM and 49.8% MM patients showed
partial response (P=0.001). Tandem transplantation and survival
A landmark analysis was used to compare tandem and
EMD and survival single ASCT, considering a total of 3139 patients who were
Median follow up was 36.3 months (range: 1-118.9 alive at six months. In patients without EMD, the compar-
months) after the date of ASCT. In the univariate analysis, ison of tandem versus single ASCT resulted in similar 3-
the MM and PS groups showed similar 3-year PFS of year PFS, 53.8% (95%CI: 46.7-60.9) versus 51.3% (95%CI:
47.9% (95%CI: 45.8-50.1) versus 50.0% (95%CI: 44.6-55.3; 48.9-53.7; P=0.37), and similar 3-year OS: 84.7% (95%CI:
P=0.78) and similar 3-year OS of 80.1% (95%CI: 78.4-81.8) 79.6-89.8) versus 81.6% (95%CI: 79.8-83.4; P=0.26).
versus 77.7% (95%CI: 73.3-82.1; P=0.09) (Figure 1A and B). Patients with EMD showed a 3-year PFS of 59.0%
In contrast, EM patients had a significantly worse 3-year (95%CI: 46.3-71.8) after tandem versus 53.0% (95%CI:
PFS of 39.9% (95%CI: 30.3-49.5) in comparison to MM 47.5-58.6) after single (P=0.43) ASCT, while 3-year OS was
(P=0.001) and PS patients (P=0.007), and a significantly 77.0% (95%CI: 66.1-87.9) versus 76.9% (95%CI: 72.4-81.4;
worse 3-year OS of 58.0% (95%CI: 48.1-67.9) compared P=0.91).
to MM and PS patients (P<0.001, respectively). Within the Within each EMD group, PS patients showed a similar 3-
EM group, 3-year PFS differed according to involved year PFS of 59.4% (95%CI: 45.3-73.6) after tandem
organs: kidney (59.5%), skin (20.1%), lymph nodes versus 54.3% (95%CI: 48.0-60.5; P=0.44) after single ASCT
(37.6%), CNS (47.9%), lung/respiratory tract (44.4%), and similar 3-year OS of 82.6% (95%CI: 72.3-92.8) versus

haematologica | 2018; 103(5) 893


N. Gagelmann et al.

80.3% (95%CI: 75.6-85.1; P=0.88). Patients with EM and Salmon and Durie (P=0.02), and lower remission status
involvement showed no significant difference in both 3- at transplantation (P<0.001).
year PFS and OS after tandem versus single transplantation: Non-relapse mortality at three years occurred in 3.0%
56.2% (95%CI: 27.2-85.3) versus 48.3% (95%CI: 36.6-60.1; (95%CI: 2.0-4.0) of MM, 3.0% (95%CI: 2.0-5.0) of PS
P=0.98), and 52.0% (95%CI: 20.0-84.0) versus 64.9% patients, and 7.0% (95%CI: 2.0-12.0) of EM patients
(95%CI: 54.2-75.7; P=0.39). (P=0.05). Main causes of death were relapse or progression
(86.3%), infection (7.1%), secondary malignancy or post-
Role of other factors on survival and causes of death transplant lymphoproliferative disorder (3.6%), organ
All patients in CR before first ASCT showed a signifi- damage or failure (1.8%), toxicity (0.4%), and unknown in
cantly better 3-year PFS of 59.8% (95%CI: 55.3-64.3) com- 83 patients.
pared to 30.7% (95%CI: 28.2-33.2) in PR and 24.7%
(95%CI: 17.6-31.8; P<0.001) in less than PR. There was Multivariate analyses
also a significant difference in 3-year OS, with patients in A multivariable model was constructed to examine the
CR showing 83.6% (95%CI: 80.2-87.0) compared to effect of EMD on 3-year PFS and OS after adjusting for
78.8% (95%CI: 76.9-80.6) in patients with PR and 27.8% possible prognostic factors. All factors and covariates
(95%CI: 20.8-34.9) in patients with less than PR (P<0.001). including corresponding references are listed in Table 3. To
Other factors associated with worse PFS in patients with avoid linearly dependent covariates, we merged the dis-
EMD were: older age (P=0.04), transplantation before 2011 ease group and the new variable of the number of involved
(P=0.01), higher disease stage according to ISS (P=0.01) and sites into a 5-level variable consisting of patients without
Salmon and Durie (P=0.02), and lower remission status at EMD (MM group) and patients with EMD according to
transplantation (P<0.001). Factors associated with worse number of involved sites (PS1, PS2, EM1 and EM2). Cox
OS in EMD patients were: transplantation before 2011 proportional hazards regression considering independent
(P=0.02), higher disease stage according to ISS (P=0.002) factors for worse PFS yielded significant results for EM2

Table 2. Involved sites in extramedullary organ involvement (EM) group and survival after autologous stem cell transplantation (ASCT).
Site N. of N. of deaths 3-year PFS 3-year OS
patients (%) in % (95% CI) in % (95% CI)
Kidney 38 (27.3) 7 59.5 (41.1 to 77.9) 75.3 (59.0 to 91.7)
CNS 14 (10.1) 4 47.9 (18.3 to 77.4) 64.3 (35.5 to 93.1)
Lung / respiratory tract 9 (6.5) 3 44.4 (7.4 to 81.5) 41.7 (0 to 85.1)
GI tract / liver 8 (5.8) 3 22.5 (0 to 58.8) 58.3 (22.0 to 94.7)
Pleura / heart 7 (5.0) 5 NE NE
Spleen / ovaries / testes 7 (5.0) 2 60.0 (17.1 to 100) 60.0 (17.1 to 100)
Skin 32 (23.0) 10 20.1 (3.4 to 36.7) 53.3 (30.5 to 76.0)
Lymph nodes 24 (17.3) 10 37.6 (16.4 to 58.7) 48.2 (25.1 to 71.3)
PFS: progression-free survival; OS: overall survival; N.: number; CI: Confidence Interval; CNS: central nervous system; GI: gastrointestinal; NE: not estimable.

Figure 2. Progression-free survival (A) and overall survival (B) with numbers at risk of myeloma patients following up-front autologous stem cell transplantation
according to number of involvements: 0, 1 and ≥ 2. N: number.

894 haematologica | 2018; 103(5)


Autologous SCT for extramedullary myeloma

with an HR of 3.40 (95%CI: 1.74-6.61; P<0.001). Discussion


Interestingly, there was no difference in PFS in EM1 com-
pared to MM with an HR of 1.03 (95%CI: 0.66-1.62; Extramedullary disease in patients with MM is consid-
P=0.88). Comparison of PFS between PS and MM showed ered a poor prognostic factor. This EBMT registry study
no difference for PS1, with an HR of 1.02 (95%CI: 0.83- including 682 EMD patients identified an increase per year
1.27; P=0.86), and a less clear HR of 2.46 (95%CI: 0.92- of EMD incidence at diagnosis from 2005 to 2014. We
6.62; P=0.07) for PS2. demonstrated that first-line ASCT resulted in at least simi-
In the OS analysis, EM1 and EM2 were associated with lar 3-year PFS in patients with single sites of EMD com-
worse outcome, showing HRs of 2.30 (95%CI: 1.43-3.70; pared to patients without EMD. Another finding, even
P=0.001) and 3.64 (95%CI: 1.48-8.94; P=0.01). The differ- though this was less clear, was that this translated into
ence between patients with one site of PS involvement and worse 3-year OS in single PS involvement while single sites
those without EMD was less clear, with an HR of 1.33 of EM were significantly associated with worse outcome,
(95%CI: 0.98-1.83; P=0.07), while PS2 resulted in a similar which worsened still further when multiple sites of organs
outcome with an HR of 0.74 (95%CI: 0.10-5.32; P=0.77). were involved. As far as treatment options for EMD at
Tandem ASCT showed similar 3-year PFS and OS com- diagnosis are concerned, we found both first-line tandem
pared to single ASCT, with HRs of 0.83 (95%CI: 0.66-1.06; and single ASCT resulted in similar 3-year PFS and OS.
P=0.13) and 0.74 (95%CI: 0.51-1.09; P=0.13). However, Evidence on the role of EMD at diagnosis after first-line
other factors made a significant contribution to an ASCT is still limited. A retrospective single center study30
increased risk of worse outcome (Table 3). For PFS, these of 27 patients concluded that ASCT might overcome poor
factors were: ISS stage II and III, PR and less than PR at prognosis at onset compared to patients without EMD,
ASCT. OS was significantly influenced by ISS stage II and while another study showed extramedullary organ
III, male sex, PR and less than PR at ASCT, and the pres- involvement in only 4 patients and that its impact on out-
ence of heavy and light chains. come could be under-estimated.5 A prospective study31 of

Table 3. Multivariate analysis.


3-year PFS 3-year OS
Factors – reference Hazard ratio (95% CI) P Hazard ratio (95% CI) P
Group – MM without EMD < 0.001 < 0.001
PS1 1.02 (0.82 to 1.27) 0.86 1.33 (0.98 to 1.83) 0.07
PS2 2.46 (0.92 to 6.62) 0.07 0.74 (0.10 to 5.32) 0.77
EM1 1.03 (0.66 to 1.62) 0.88 2.30 (1.43 to 3.70) 0.001
EM2 3.40 (1.74 to 6.61) < 0.001 3.64 (1.48 to 8.94) 0.01
Sex – male
Female 0.86 (0.74 to 1.01) 0.06 0.71 (0.56 to 0.91) 0.01
Age in years, > 60 0.22 0.45
< 50 0.81 (0.74 to 1.03) 0.08 1.04 (0.73 to 1.48) 0.85
50 to 60 0.95 (0.80 to 1.11) 0.50 1.17 (0.91 to 1.50) 0.21
ISS – I < 0.001 < 0.001
II 1.48 (1.23 to 1.77) < 0.001 1.75 (1.29 to 2.37) < 0.001
III 1.81 (1.46 to 2.24) < 0.001 2.68 (1.92 to 3.74) < 0.001
Renal function – A
B 0.99 (0.79 to 1.24) 0.93 1.25 (0.92 to 1.69) 0.16
Status at ASCT – CR < 0.001 0.02
PR 1.58 (1.26 to 1.97) < 0.001 1.48 (1.05 to 2.08) 0.03
< PR 2.18 (1.54 to 3.10) < 0.001 2.08 (1.22 to 3.54) 0.01
Type of myeloma - light chain 0.10 0.09
Non-secretory 0.77 (0.42 to 1.43) 0.41 1.70 (0.80 to 3.61) 0.17
Heavy and light 1.19 (0.98 to 1.46) 0.08 1.38 (1.01 to 1.88) 0.04
Year of ASCT - > 2011 0.21 0.91
< 2009 1.22 (0.97 to 1.53) 0.09 1.07 (0.75 to 1.53) 0.71
2009 to 2011 1.05 (0.88 to 1.26) 0.61 1.00 (0.76 to 1.33) 0.98
Type of ASCT - single
Tandem 0.83 (0.66 to 1.06) 0.13 0.74 (0.51 to 1.09) 0.13
PFS: progression-free survival; OS: overall survival; CI: Confidence Interval; MM: patients without extramedullary disease; PS: patients with paraskeletal involvement arising from
bone lesions; PS1: patients with paraskeletal involvement having one involved site; PS2: patients with paraskeletal involvement and multiple involved sites; EM1: patients with
extramedullary organ involvement having one involved site; EM2: patients with extramedullary organ involvement and multiple involved sites; ISS: International Staging System;
CR: complete remission; PR: partial remission; ASCT: autologous stem cell transplantation.

haematologica | 2018; 103(5) 895


N. Gagelmann et al.

Figure 3. Progression-free survival (A) and overall survival (B) with numbers at risk of myeloma patients with extramedullary organ involvement following up-front
autologous stem cell transplantation according to number of involvements: 1 and ≥ 2. EM: patients with extramedullary organ involvement; EM1: patients with one
site of EM; EM2: patients with two or more sites of EM; N: number.

patients in relapse with either soft-tissue or bone-related of response.42 With regard to these analyses outside trans-
involvement at a single institution found that bone-related plantation settings, we investigated survival according to
relapses were associated with better OS. However, treat- involved sites in EM patients, finding most of the patients
ments before diagnosis of extramedullary relapse signifi- had kidney, skin or lymph node involvement. After up-
cantly differed between groups. Since different types of front ASCT, best outcomes were found in kidney and CNS
involvement were reported, this variable was examined involvement while skin and lymph node involvement
closely. showed worse outcome. Interestingly, our CNS cohort
In our study, especially EM involvement in 139 MM showed higher rates of OS compared to previous reports,
patients was associated with lower rate of CR before and which might be due to the selection of patients with CNS
after ASCT, a higher frequency of ISS stage III, and worse involvement at diagnosis, while most reports evaluated
renal function. Importantly, the impact of the number of patients at later phases of the disease.41,42
involved sites on outcome in EMD at diagnosis had not A pooled analysis of prospective studies regarding trans-
previously been described. We found 20% of all EMD plantation strategies suggested the superiority of tandem
patients having multiple sites of involvement, which is in ASCT in patients with poor prognostic features at diagno-
line with previous reports (16%).13 Notably, the location sis.4,43 Our landmark analyses of EMD patients who
of further involvement was only paraskeletal in the PS received either tandem or single ASCT as first-line therapy
group and was also restricted to other organs in the EM found no difference in PFS and OS. However, this analysis
group.32 was conducted with the use of retrospective data and is,
The use of radiation therapy might contribute to the dif- therefore, subject to the attendant limitations. Regression
ference in PFS and OS of patients with single sites of EMD modeling and landmark analyses were performed as a
compared to patients without EMD, because it is consid- means of controlling for differences between the patients,
ered effective in reducing progression in patients with soli- but such adjustment cannot account for all discrepancies in
tary osseous and extraosseous involvement,33,34 in particu- clinical and diagnostic characteristics between groups. The
lar because reports about the efficacy of novel agents in increasing incidence of EMD might be caused by a more
these cohorts at diagnosis are very limited. Some results frequent use of whole-body MRI or PET-CT in recent
would suggest an induction bortezomib-based regimen years. However, although recent evidence promotes the
followed by high-dose melphalan/ASCT for patients with use of more sensitive imaging techniques,44 data are not
paraskeletal rather than extramedullary involvement.14,35-37 routinely documented, and they are still not part of routine
In a retrospective study38 investigating carfilzomib alone or diagnostics and were thus not available in our study.45,46 A
in combination as salvage therapy in relapse, presence of randomized trial is the only way to overcome these chal-
extramedullary involvement resulted in shorter duration of lenges and to assess the definite impact of EMD in newly
response compared to absent EMD, suggesting limited diagnosed MM patients after ASCT.
treatment effect. Smaller reports on the possible impact of In conclusion, this EBMT study identified an increase in
immunomodulatory drugs showed partial efficacy regard- incidence per year of EMD in newly diagnosed MM
ing response rates in EMD patients.10,39,40 patients from 2005 to 2014. We revealed that first-line
Retrospective studies highlighted an extremely poor ASCT in patients with single sites of EMD (PS or EM)
prognosis for CNS involvement with a median OS of less resulted in at least similar 3-year PFS compared to patients
than six months.41,42 However, in addition to systemic anti- without EMD. Nevertheless, single EM involvement was
MM therapy, CNS irradiation and the use of novel combi- associated with worse 3-year OS, which worsened still fur-
nation therapies have been shown to improve the duration ther when multiple sites of organs were involved.

896 haematologica | 2018; 103(5)


Autologous SCT for extramedullary myeloma

risk cytogenetics: a consensus of the Recommendations for the Treatment of


References International Myeloma Working Group. Multiple Myeloma–Related Bone Disease. J
1. National Cancer Institute. Surveillance epi- Blood. 2016;127(24):2955-2962. Clin Oncol. 2013;31(18):2347-2357.
demiology and end results program. SEER 17. Cavo M, Rajkumar SV, Palumbo A, et al. 35. Paubelle E, Coppo P, Garderet L, et al.
stat fact sheets: Myeloma. 2013; International Myeloma Working Group con- Complete remission with bortezomib on
http://seer.cancer.gov/statfacts/html/mulmy sensus approach to the treatment of multi- plasmocytomas in an end-stage patient
.html [Last accessed 14 April 2017]. ple myeloma patients who are candidates with refractory multiple myeloma who
2. Kumar SK, Rajkumar SV, Dispenzieri A, et for autologous stem cell transplantation. failed all other therapies including
al. Improved survival in multiple myeloma Blood. 2011;117(23):6063-6073. hematopoietic stem cell transplantation:
and the impact of novel therapies. Blood. 18. Bianchi G, Richardson PG, and Anderson possible enhancement of graft-vs-tumor
2008;111(5):2516-2520. KC. Best Treatment Strategies in High-Risk effect. Leukemia. 2005;19(9):1702-1704.
3. Palumbo A, Anderson K. Multiple myeloma. Multiple Myeloma: Navigating a Gray Area. 36. Sonneveld P, Goldschmidt H, Rosiñol L, et
N Engl J Med. 2011; 364(11):1046-1060. J Clin Oncol. 2014;32(20):2125-2132. al. Bortezomib-based versus nonborte-
4. Touzeau C, Moreau P. How I treat 19. Bladé J, Samson D, Reece D, et al. Criteria zomib-based induction treatment before
extramedullary myeloma. Blood. 2016; for evaluating disease response and progres- autologous stem-cell transplantation in
127(8):971-976. sion in patients with multiple myeloma patients with previously untreated multiple
5. Bladé J, Fernández de Larrea C, Rosiñol L, treated by high-dose therapy and myeloma: a meta-analysis of phase III ran-
Cibeira MT, Jiménez R, Powles R. Soft-tis- haemopoietic stem cell transplantation. domized, controlled trials. J Clin Oncol.
sue plasmacytomas in multiple myeloma: Myeloma Subcommittee of the EBMT. 2013;31(26):3279-3287.
incidence, mechanisms of extramedullary European Group for Blood and Marrow 37. Mateos MV, Oriol A, Matínez-López J, et al.
spread, and treatment approach. J Clin Transplant. Br J Haematol. 1998; Bortezomib, melphalan, and prednisone
Oncol. 2011;29(28):3805-3812. 102(5):1115-1123. versus bortezomib, thalidomide, and pred-
6. Weinstock M, Aljawai Y, Morgan EA, et al. 20. Greipp PR, San Miguel J, Durie BG, et al. nisone as induction therapy followed by
Incidence and clinical features of International staging system for multiple maintenance treatment with bortezomib
extramedullary multiple myeloma in myeloma. J Clin Oncol. 2005;23(15):3412- and thalidomide versus bortezomib and
patients who underwent stem cell transplan- 3420. prednisone in elderly patients with untreat-
tation. Br J Haematol. 2015;169(6):851-858. 21. Durie BG, Salmon SE. A clinical staging sys- ed multiple myeloma: a randomised trial.
7. Cavo M, Terpos E, Nanni C, et al. Role of tem for multiple myeloma. Correlation of Lancet Oncol. 2010;11(10):934-941.
18F-FDG PET/CT in the diagnosis and man- measured myeloma cell mass with present- 38. Muchtar E, Gatt ME, Rouvio O, et al.
agement of multiple myeloma and other ing clinical features, response to treatment, Efficacy and safety of salvage therapy using
plasma cell disorders: a consensus statement and survival. Cancer. 1975;36(3):842-854. Carfilzomib for relapsed or refractory mul-
by the International Myeloma Working 22. Karnofsky DA, Abelmann WH, Craver LF, tiple myeloma patients: a multicentre retro-
Group. Lancet Oncol. 2017;18(4):e206-e217. Burchenal JH. The Use of the Nitrogen spective observational study. Br J Haematol.
8. Bartel TB, Haessler J, Brown TL, et al. F18- Mustards in the Palliative Treatment of 2016;172(1):89-96.
fluorodeoxyglucose positron emission Carcinoma – with Particular Reference to 39. Calvo-Villas JM, Alegre A, Calle C, et al.
tomography in the context of other imaging Bronchogenic Carcinoma. Cancer. Lenalidomide is effective for
techniques and prognostic factors in multi- 1948;1(4):634-56. extramedullary disease in relapsed or refrac-
ple myeloma. Blood. 2009; 114(10):2068- 23. Kaplan E, Meier P. Nonparametric estima- tory multiple myeloma. Eur J Haematol.
2076. tion from incomplete observations. J Am 2011;87(3):281-284.
9. Usmani SZ, Heuck C, Mitchell A, et al. Stat Assoc. 1958;53(282):457-481. 40. Rosiñol L, Cibeira MT, Bladé J, et al.
Extramedullary disease portends poor prog- 24. Schemper M, Smith TL. A note on quantify- Extramedullary multiple myeloma escapes
nosis in multiple myeloma and is overrepre- ing follow-up in studies of failure time. the effect of thalidomide. Haematologica.
sented in high-risk disease even in the era of Control Clin Trials. 1996;17(4):343-346. 2004;89(7):832-836.
novel agents. Haematologica. 25. Fine JP, Gray RJ. A proportional hazards 41. Nieuwenhuizen L, Biesma DH. Central
2012;97(11):4761-4767. model for the subdistribution of a compet- nervous system myelomatosis: review of
10. Short KD1, Rajkumar SV, Larson D, et al. ing risk. J Am Stat Assoc. 1999;94(446):496- the literature. Eur J Haematol. 2008;80(1):1-
Incidence of extramedullary disease in 509. 9.
patients with multiple myeloma in the era of 26. Gray RJ. A class of k-sample tests for com- 42. Chen CI, Masih-Khan E, Jiang H, et al.
novel therapy and the activity of pomalido- paring the cumulative incidence of a com- Central nervous system involvement with
mide in extramedullary myeloma. peting risk. Ann Stat. 1988;16(3):1141-1154. multiple myeloma: long term survival can
Leukemia. 2011;25(6):906-908. 27. Iacobelli S, EBMT Statistical Committee. be achieved with radiation, intrathecal
11. Rosiñol L, Fernández de Larrea C, Bladé J. Suggestions on the use of statistical method- chemotherapy, and immunomodulatory
Extramedullary myeloma spread triggered ologies in studies of the European Group for agents. Br J Haematol. 2013;162(4):483-488.
by surgical procedures: an emerging entity? Blood and Marrow Transplantation. Bone 43. Cavo M, Salwender H, Rosiñol L, et al.
Acta Haematol. 2014;132(1):36-38. Marrow Transplant. 2013;48(Suppl 1):S1-37. Double vs single autologous stem cell trans-
12. Pérez-Simón JA, Sureda A, Fernández-Aviles 28. Klein JP, Moeschberger ML (eds): Survival plantation after bortezomib-based induc-
F, et al. Reduced-intensity conditioning allo- Analysis: Techniques for Censored and tion regimens for multiple myeloma: an
geneic transplantation is associated with a Truncated Data, 2nd edn. Springer-Verlag: integrated analysis of patient-level data
high incidence of extramedullary relapses in New York, USA, 2003. from phase European III studies. Blood.
multiple myeloma patients. Leukemia. 29. Cox DR: Regression models and life tables. J 2013;122:767.
2006;20(3):542-545. R Stat Soc B. 1972;34(2):187-220. 44. Moreau P, Attal M, Caillot D, et al.
13. Varettoni M1, Corso A, Pica G, 30. Lee SE, Kim JH, Jeon YW, et al. Impact of Prospective evaluation of magnetic reso-
Mangiacavalli S, Pascutto C, Lazzarino M. extramedullary plasmacytomas on out- nance imaging and [18F]
Incidence, presenting features and outcome comes according to treatment approach in Fluorodeoxyglucose positron emission
of extramedullary disease in multiple myelo- newly diagnosed symptomatic multiple tomography-computed tomography at
ma: a longitudinal study on 1003 consecu- myeloma. Ann Hematol. 2015; 94(3):445- diagnosis and before Maintenance Therapy
tive patients. Ann Oncol. 2010;21(2):325- 452. in Symptomatic Patients With Multiple
330. 31. Pour L, Sevcikova S, Greslikova H, et al. Myeloma Included in the IFM/DFCI 2009
14. Rosiñol L, Oriol A, Teruel AI, et al. Soft-tissue extramedullary multiple myelo- Trial: Results of the IMAJEM Study. J Clin
Superiority of bortezomib, thalidomide, and ma prognosis is significantly worse in com- Oncol. 2017; 35(25):2911-2918.
dexamethasone (VTD) as induction pre- parison to bone-related extramedullary 45. Kumar S, Paiva B, Anderson KC, et al.
transplantation therapy in multiple myelo- relapse. Haematologica. 2014;99(2):360- International Myeloma Working Group con-
ma: a randomized phase 3 364. sensus criteria for response and minimal
PETHEMA/GEMstudy. Blood. 32. Ghobrial IM. Myeloma as a model for the residual disease assessment in multiple
2012;120(8):1589-1596. process of metastasis: implications for ther- myeloma. Lancet Oncol. 2016;17(8):e328-
15. Usmani SZ, Rodriguez-Otero P, Bhutani M, apy. Blood. 2012;120(1):20-30. 346.
Mateos MV, Miguel JS. Defining and treat- 33. Hu K, Yahalom J. Radiotherapy in the man- 46. Pawlyn C, Fowkes L, Otero S, et al. Whole-
ing high-risk myeloma. Leukemia. agement of plasma cell tumors. Oncology body diffusion-weighted MRI: a new gold
2015;29(11):2119-2125. (Williston Park). 2000;14(1):101-108. standard for assessing disease burden in
16. Sonneveld P, Avet-Loiseau H, Lonial S, et al. 34. Terpos E, Morgan G, Dimopoulos MA, et patients with multiple myeloma?
Treatment of multiple myeloma with high- al. International Myeloma Working Group Leukemia. 2016;30(6):1446-1448.

haematologica | 2018; 103(5) 897


ARTICLE Hemostasis

Immobilized fibrinogen activates human


Ferrata Storti
Foundation platelets through glycoprotein VI
Pierre H Mangin,1 Marie-Blanche Onselaer,2 Nicolas Receveur,1 Nicolas Le
Lay,3 Alexander T Hardy,2 Clare Wilson,4 Ximena Sanchez,5 Stéphane Loyau,3
Arnaud Dupuis,1 Amir K Babar,6 Jeanette LC Miller,6 Helen Philippou,4 Craig E
Hughes,2,8 Andrew B Herr,6 Robert AS Ariëns,4 Diego Mezzano,5 Martine
Jandrot-Perrus,3,7 Christian Gachet1 and Steve P. Watson2,9
1
Université de Strasbourg, INSERM, EFS Grand-Est, BPPS UMR-S 1255, FMTS, France;
Haematologica 2018 2
Institute of Cardiovascular Sciences, IBR Building, College of Medical and Dental
Sciences, University of Birmingham, UK; 3Université de Paris Diderot, INSERM
UMR_S1148, Hôpital Bichat, Paris, France ; 4Thrombosis and Tissue Repair Group, Institute
Volume 103(5):898-907
of Cardiovascular and Metabolic Medicine, University of Leeds, UK; 5Laboratorio de
Hemostasia, Pontificia Universidad Catolica de Chile, Santiago, Chile; 6Division of
Immunobiology, Center for Systems Immunology & Division of Infectious Diseases,
Cincinnati, OH, USA; 7Acticor Biotech, Hôpital Bichat, INSERM, UMR-S 1148, Paris, France;
8
Institute for Cardiovascular and Metabolic Research, Harborne Building, University of
Reading, UK and 9Centre of Membrane Proteins and Receptors (COMPARE), Universities of
Birmingham and Nottingham, Midlands, UK

ABSTRACT

G
lycoprotein VI, a major platelet activation receptor for collagen
and fibrin, is considered a particularly promising, safe antithrom-
botic target. In this study, we show that human glycoprotein VI
signals upon platelet adhesion to fibrinogen. Full spreading of human
platelets on fibrinogen was abolished in platelets from glycoprotein VI-
deficient patients suggesting that fibrinogen activates platelets through
glycoprotein VI. While mouse platelets failed to spread on fibrinogen,
human-glycoprotein VI-transgenic mouse platelets showed full spreading
Correspondence: and increased Ca2+ signaling through the tyrosine kinase Syk. Direct bind-
pierre.mangin@efs.sante.fr or ing of fibrinogen to human glycoprotein VI was shown by surface plas-
s.p.watson@bham.ac.uk mon resonance and by increased adhesion to fibrinogen of human glyco-
protein VI-transfected RBL-2H3 cells relative to mock-transfected cells.
Received: October 20, 2017. Blockade of human glycoprotein VI with the Fab of the monoclonal anti-
Accepted: February 13, 2018.
body 9O12 impaired platelet aggregation on preformed platelet aggre-
gates in flowing blood independent of collagen and fibrin exposure.
Pre-published: February 22, 2018.
These results demonstrate that human glycoprotein VI binds to immobi-
lized fibrinogen and show that this contributes to platelet spreading and
doi:10.3324/haematol.2017.182972 platelet aggregation under flow.

Check the online version for the most updated


information on this article, online supplements,
and information on authorship & disclosures:
www.haematologica.org/content/103/5/898 Introduction

The immunoglobulin receptor glycoprotein (GP) VI is expressed on megakary-


©2018 Ferrata Storti Foundation ocytes and platelets. GPVI associates with the Fc receptor (FcR) γ-chain in the
Material published in Haematologica is covered by copyright. membrane, and with the Src family kinases (SFK) Lyn and Fyn through its cytosolic
All rights are reserved to the Ferrata Storti Foundation. Use of tail.1 Ligand binding clusters GPVI at the platelet surface promoting phosphoryla-
tion of the immunoreceptor tyrosine-based motif (ITAM) of the FcR γ-chain by
published material is allowed under the following terms and
conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode. SFK.2–4 This results in the recruitment of Syk and formation of a LAT-based signalo-
Copies of published material are allowed for personal or inter- some that activates PLCγ2 leading to an increase in Ca2+, activation of integrin and
nal use. Sharing published material for non-commercial pur- secretion of granules.5
poses is subject to the following conditions:
GPVI is widely known as a platelet activation receptor for fibrillar collagen.5
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- However, in recent years, GPVI has been shown to bind to additional ligands,
mercial purposes is not allowed without permission in writing including subendothelial and plasma adhesive proteins such as laminins and fib-
from the publisher. rin,6–8 the hormone adiponectin and the transmembrane protein emmprin.9,10
Several of these interactions are relatively weak and of unclear significance. For
example, GPVI supports adhesion and efficient activation of platelets to collagen

898 haematologica | 2018; 103(5)


Fibrinogen, GPVI and platelet activation

and fibrin but is only involved in post-adhesive events on Mice


laminin.6,11 Determining the importance of the interaction Wild-type mice were generated from breeding of heterozy-
of GPVI with each ligand in mediating platelet activation gotes or purchased from Harlan Laboratories (Hillcrest, UK) or
in vivo will require development of selective inhibitors. Charles River (Lyon, France). GPVI-/- mice were provided by Dr
GPVI is involved in arterial thrombosis and in several of Jerry Ware.31 Mouse platelets expressing human GPVI have
the more recently discovered roles of platelets, including been described and characterized previously.32 Syk chimera
maintenance of vascular integrity at sites of inflammatory mice have been described previously.27 Ethical approval for ani-
challenge.12 We and others have reported that the absence mal experimentation was obtained from the French Ministry of
of GPVI reduces experimental thrombosis in mouse mod- Research and UK Home Office in accordance with the
els of atherosclerotic plaque rupture13,14 and abolishes European Union guidelines, the Guide for the Care and Use of
occlusive thrombus formation following FeCl3 injury.15 In Laboratory Animals.
contrast, the absence or blockade of GPVI has a relatively
minor impact on hemostasis in mice16 and patients defi- Reagents
cient in GPVI have a relatively mild bleeding diathesis.17–19 PRT-060318 was obtained from Caltag Medsystems
These results highlight that GPVI is a promising anti- (Buckingham, UK), REOPRO from E. Lilly (Indianapolis, IN,
thrombotic target with inhibitors predicted to have a USA). Recombinant GPVI was made as described elsewhere.33
minor effect on hemostasis.20 Fibrinogen was from Kabi (Bad Homburg, Germany) or from
Following ligand binding, GPVI stimulates signals that ERL (South Bend, IN, USA). RAM.1 (anti-GPIbβ) was generated
convert integrin aIIbβ3 from a low to a high affinity state in U949.34 The blocking Fab fragment of monoclonal antibody
for fibrinogen and other physiological ligands.21 Ligand directed against human GPVI, 9O12.2, and its humanized ver-
engagement of integrin aIIbβ3 has been reported to gen- sion are referred to as 9O12 in this manuscript.35,36 All other
erate outside-in signals that are similar to those of GPVI, reagents were from previously described sources.11,37 The anti-
including activation of Src and Syk kinases, PLCγ2 and fibrin antibody 59D8 was obtained from CT Esmon (Oklahoma
Ca2+ mobilization.22–24 Paradoxically, however, human but Medical Research Foundation, OK, USA).38
not mouse platelets generate extensive lamellipodial
sheets and stress fibers on fibrinogen whereas on colla- Generation and characterization of RBL-2H3 cells
gen, which stimulates similar signals, full spreading of The cDNA of WT human GPVI33 was inserted in pSRαNeo
platelets is seen in both species.25 One explanation for between the 5’-XhoI and 3’-BamHI restriction sites. Rat
this difference is the presence of the low affinity immune basophilic leukemia cells, RBL-2H3, were cultured in Dulbecco
receptor, FcγRIIA, in the human but not the rodent modified Eagle medium supplemented with 10% fetal bovine
genome, as FcγRIIA-transfected transgenic mouse serum albumin and stably transfected with 1 μg of DNA corre-
platelets exhibit increased spreading and Syk activation sponding to the empty vector or WThGPVI vector mixed with
upon adhesion to fibrinogen, although the increase in FuGENE6 (Roche, Boulogne-Billancourt, France) and selected in
spreading is only partial.26–28 Outside-in signaling by growth medium containing G418 0.7%; 1 mg/mL geneticin
aIIbβ3 is also mediated by two conserved tyrosines pres- (GibcoBRL, Invitrogen, Cergy Pontoise). Cell surface expression
ent in a NxxY motif in the integrin β3 intracytoplasmic of recombinant GPVI and constitutively expressed integrin
domain independent of Src and Syk activation. Mutation aIIbβ3 was confirmed by flow cytometry and immunoblot
of these two tyrosine residues to phenylalanine leads to (data not shown).
a re-bleeding diathesis that has been attributed to a
defect in clot retraction.29 This shows that engagement of Cell adhesion to fibrinogen
integrin aIIbβ3 leads to activation of multiple signaling LAB TEK 4 wells were coated with 400 μL/well of collagen
pathways. (50 µg/mL) or fibrinogen (100 μg/mL) overnight at 37°C. Wells
In the present study, we showed that full spreading of were saturated with human serum albumin (10 mg/mL) for 1 h
human platelets on fibrinogen is abolished in patients at 37°C. Trypsinised RBL cells (3x105 cells/mL) were incubated
deficient in GPVI and that transgenic mouse platelets with phosphate-buffered saline or 9O12 (50 μg/mL) and/or
expressing human GPVI, in contrast to wild-type REOPRO (40 μg/mL) for 15 min at 37°C. Subsequently, 100,000
platelets, spread fully on fibrinogen. Direct binding of fib- cells were added to the wells (300 μL) for 1 h at 37°C. After
rinogen to human GPVI was demonstrated using human three washing steps, cells were fixed with 400 μL paraformalde-
GPVI-transfected cell lines and recombinant GPVI. hyde 4% for 20 min. Pictures were taken with an EVOS optic
Inhibiting the binding of fibrinogen to GPVI limited microscope (x10). Actin was stained with Alexa-488-phalloidin
platelet aggregation under conditions that excluded and the nucleus with DAPI.
involvement of collagen and fibrin.
Washed platelets
Human blood was taken from patients or from healthy
Methods donors using 3.8% (v/v) sodium citrate (1:9) as the anticoagu-
lant. Human and mouse washed platelets were obtained by
Patients centrifugation using prostacyclin (2.8 μmol/L) and resuspended
Family 1 and family 2 are two families who are heterozygous or in modified Tyrode-HEPES buffer as described elsewhere.37,39
homozygous for an adenine insertion in exon 6 of GP6 that gen-
erates a premature stop codon in position 242 of the protein.17 Platelet spreading
They have been described previously.30 Patient 3 is a 10-year old Platelet adhesion to immobilized fibrinogen was achieved as
boy suffering from an autoimmune disease with anti-GPVI anti- described previously.37 Images of the platelets were obtained
bodies. The platelets of this patient do not aggregate to collagen with a Zeiss Axiovert 200 mol/L microscope or with a Leica
and GPVI is not detected at the platelet surface using flow cytom- DMI400 microscope. Platelet surface area was analyzed using
etry and western blot (data not shown). ImageJ software (NIH, Bethesda, USA).

haematologica | 2018; 103(5) 899


P.H. Mangin et al.

Western blotting The values are indicated as mean ± standard error of the mean
For stimulation on fibrinogen, washed platelets were pre-treat- (SEM). The statistical analysis is described in the Figure legends.
ed with 10 μmol/L indomethacin and 2 U/mL apyrase. Platelets
(1.5 mL containing 5x108/mL) were allowed to adhere to 10 cm
dishes coated with 100 μg/mL fibrinogen or heat-inactivated Results
bovine serum albumin for 45 min at 37°C. Non-adherent platelets
were removed and lysed by addition of 2X lysis buffer (150 Abolition of spreading on fibrinogen in glycoprotein
mmol/L NaCl, 10 mmol/L Tris, 1 mmol/L EGTA, 1 mmol/L EDTA, VI-deficient human platelets
1% NP-40; pH 7.4, plus 1.25 mmol/L Na3VO4, 50 μg/mL AEBSF, Human platelets undergo robust spreading on immobi-
2.5 μg/mL leupeptin, 2.5 μg/mL aprotinin and 0.25 μg/mL pep- lized fibrinogen, generating lamellipodial sheets and stress
statin). Adherent platelets were washed twice with Tyrode buffer fibers.41 This is illustrated in Figure 1A with over 90% of
then lysed with 1X lysis buffer on ice for 15 min before scraping. platelets from a control donor undergoing full spreading
Proteins were immunoprecipitated with a-Syk antibody and pro- on fibrinogen over 30 min; the small number of partially-
tein A-sepharose beads for 2 h. The beads were washed, proteins spread platelets most likely represent newly adhered cells.
eluted in sodium dodecyl sulfate (SDS) sample buffer, separated In 2013, Matus et al. described four unrelated families with
by SDS-polyacrylamide gel electrophoresis (PAGE), electro-trans- index cases who are homozygous for an adenine insertion
ferred, and western blotted with the stated antibodies. For whole in exon 6 of human GP6, which leads to a premature ‘stop
platelet lysates, washed platelets (5x108/mL) were lysed directly codon’ in position 242 prior to the transmembrane
with an equal volume of 2xSDS sample buffer, separated by SDS- domain.17 All four homozygous patients lack expression of
PAGE, electro-transferred, and western blotted with the stated GPVI on their platelets and heterozygous relatives express
antibodies. approximately 50% of the receptor. Since then, two fur-
ther unrelated families with the same mutation have been
Ca2+ assay and in vitro perfusion assay identified by the same group and also been shown to lack
Intraplatelet Ca2+ concentrations following platelet adhesion to surface expression of GPVI with absent platelet aggrega-
fibrinogen were measured using a dual-dye ratiometric method tion to collagen.30 Unexpectedly, in studying platelets from
and hirudinated blood perfusion was performed as previously two unrelated index cases in these families, we observed
described.40 Three-dimensionsal reconstructed images were reduced adhesion on immobilized fibrinogen and a failure
obtained using the 3D module of Leica LAS X software. to form lamellipodial sheets and stress fibers (Figure 1A).
The absence of GPVI was confirmed by flow cytometry
Solid-based binding assay and by abolition of aggregation to collagen but not to
Binding studies were performed with the recombinant proteins, other agonists in both cases30 (data not shown). In contrast,
GPVI-Fc fusion (dimer) and GPVI-His tagged (monomer). Cover spreading and adhesion of platelets from heterozygote
slips were coated with collagen or fibrinogen overnight at 4°C. carriers from each family and platelets from a control were
The plates were blocked with 3% bovine serum albumin – phos- similar (Figure 1A). The same result was also seen in a
phate-buffered saline for 1 h and washed prior to addition of patient with an auto-immune thrombocytopenia associat-
monomeric or dimeric GPVI at a concentration of 100 nmol/L for ed with the absence of GPVI expression (Figure 1Bi).
1 h. After washing, 4 μg/mL of secondary antibodies, horseradish Adhesion of platelets was blocked by the aIIbβ3 receptor
peroxidase (HRP)-conjugated goat anti-human IgG Fc or HRP-con- antagonist, REOPRO (Figure 1B), as previously shown in
jugated anti-His Tag, were added for 1 h. GPVI binding was controls. These results demonstrate that adhesion of
detected using 3,3′,5,5′-tetramethylbenzidine. The reaction was human platelets on fibrinogen is critically dependent on
stopped with H2SO4 (2 mol/L) and absorbance was measured at integrin aIIbβ3 with a minor contribution from GPVI, but
450 nm with a spectrofluorometer. that full spreading requires GPVI.

Surface plasmon resonance Mouse platelets expressing human glycoprotein VI


Surface plasmon resonance was performed on a Pioneer plat- undergo full spreading on fibrinogen
form from PALL® FortéBio® (Portsmouth, UK). IF-1 purified fib- Mouse platelets adhere and undergo limited spreading
rinogen was diluted to 100 μg/mL using 10 mmol/L NaAc pH 5.0. on human or mouse fibrinogen, forming filopodia and
IF-1 fibrinogen was adsorbed to the chip surface via amine cou- limited lamellipodia but not stress fibers (Figure 2A). A
pling to a level of 3825 resonance units (RU) at flow-cell 1 and similar response is seen in platelets deficient in GPVI
3423 RU on flow-cell 3. Flow-cell 2 was activated using amine (Figure 2A), whereas adhesion is abolished in the absence
coupling and blocked using 1 mol/L ethanolamine and was the of the integrin β3-subunit (Figure 2B). A similar level of
designated reference channel. GPVI analytes were dialysed and adhesion is seen in human GPVI transgenic mouse
diluted to 1 µmol/L using the same batch of running buffer as used platelets but is associated with the formation of lamellipo-
for the blanks. Analytes were injected using the OneStep® titration dial sheets and stress fibers (Figure 2C). These results
function at a flow rate of 30 μL/min with a 100% loop-inject and demonstrate that full spreading but not adhesion of mouse
400 s dissociation. The chip surface was regenerated by flushing platelets is dependent on human GPVI and not mouse
with 1 mol/L NaCl at 60 μL/min for 10 s, followed by a further 400 GPVI. One potential explanation for these results is that
s dissociation. Qdat data analysis software (PALL® FortéBio®, UK) human but not mouse GPVI is able to bind to fibrinogen
was used to analyze the data. Binding data were fitted using a one and mediate platelet activation.
site KA/KD model and analyte aggregation parameters adjusted per
binding curve according to goodness of fit and curve type. Fibrinogen binds to monomeric human glycoprotein VI
To test whether fibrinogen is able to bind to GPVI,
Statistics increasing concentrations of recombinant soluble GPVI
The statistical analyses were performed using the GraphPad extracellular domain, expressed either as a monomer
Prism program, version 5.0 (Prism, GraphPad, LaJolla, CA, USA). (GPVI-ex) or dimer (GPVI-Fc), was flowed over immobi-

900 haematologica | 2018; 103(5)


Fibrinogen, GPVI and platelet activation

lized fibrinogen and binding monitored by surface plas- antibody 9012 Fab blocked the increase in adhesion.
mon resonance. As shown in Figure 3A, clear binding of Blocking the integrin aIIbβ3 with REOPRO reduced cell
monomeric GPVI (ka = 1.17 ± 0.01 x 104 M-1s-1) was adhesion to immobilized fibrinogen to the same level as
observed with a kd of 3.94 ± 0.01 x 10-3 s-1. Binding was fit- 9O12 Fab, with no further inhibition in the presence of
ted to a single site with an equilibrium dissociation con- both inhibitors (data not shown), indicating the presence of
stant (KD) of 336 ± 1 nmol/L. In contrast, binding of dimer- additional binding proteins for fibrinogen in the adherent
ic GPVI to fibrinogen was not detected at concentrations cell line although binding to these was not sufficient to
up to 1 μmol/L (Figure 3Ai). In a second approach, fibrino- induce spreading (Figure 3Biii and not shown). These
gen was immobilized on a plastic surface and a solid results demonstrate that GPVI binds to immobilized fib-
phase binding assay was performed. There was increased rinogen and is able to contribute to cell adhesion.
binding of monomeric GPVI, but not dimeric GPVI, which
was inhibited by D-dimer (Figure 3Aii) where the binding Spreading of human platelets but not mouse platelets
motif in fibrin resides.30 To further investigate the ability is dependent on Syk
of GPVI to bind to fibrinogen, we transfected rat RBL-2H3 The formation of lamellipodial sheets and stress fibers
basophilic cells, which constitutively express integrin in human platelets on fibrinogen and collagen is blocked
aIIbβ3 at low levels, with human GPVI and studied adhe- by the inhibitors of Src and Syk tyrosine kinases, PP2 and
sion to immobilized fibrinogen. We observed a 3-fold PRT060318, respectively (Figure 4Ai & ii). Adhesion of
increase in adhesion of GPVI-transfected cells to fibrino- human platelets to fibrinogen induces phosphorylation of
gen and to collagen relative to the adhesion of mock-trans- Syk which co-precipitates with the phosphorylated FcR γ-
fected control cells (Figure 3Bi, ii). RBL-2H3 cells express- chain (Figure 4Aiii). These results provide further evidence
ing human GPVI also formed stress fibers upon adhesion of GPVI activation in human platelets by immobilized fib-
to fibrinogen. The human GPVI-blocking monoclonal rinogen. In contrast, the morphological modifications of

Figure 1. Glycoprotein VI supports


platelet adhesion and spreading on
immobilized fibrinogen. (A, B) Washed
human platelets were allowed to
adhere to immobilized fibrinogen (10
μg/mL) for 30 min. (A)(i).
Representative epifluorescence
images of fibrinogen-adherent
platelets from healthy donors (Control)
or members of a family with a mutation
in the GP6 gene (Heterozygotes: Family
1 +/-; homozygotes: Family 1 -/-). Scale
bars represent 10 μm. (A)(ii). Bar graph
representing the percentage of
platelets spreading on fibrinogen.
Spreading is expressed as the
mean±SEM in five random fields, in
two separate experiments (one-way
ANOVA, Kruskal-Wallis post-hoc test,
***P<0.0002; ****P<0.0001).
(A)(iii). Bar graph representing the
number of platelets adhering to immo-
bilized fibrinogen per mm² of a control
(Control) and two families with a muta-
tion in the GP6 gene. Adhesion is
expressed as mean±SEM in five ran-
dom fields, in two separate experi-
ments (one-way ANOVA, Kruskal-Wallis
post-hoc test, **P<0.002;
****P<0.0001). (B). Washed platelets
from a control or a patient with an
immune thrombocytopenic purpura
presenting with undetectable levels of
B GPVI on platelets (Patient 3) were
allowed to adhere to fibrinogen (100
μg/mL). (B)(i). Representative epifluo-
rescence images of washed platelets
from patient 3 adhering to immobilized
fibrinogen for 30 min, in the presence
or absence of REOPRO (40 μg/mL).
Scale bars represent 10 μm. (B)(ii). Bar
graph represents the number of
platelets adhering to fibrinogen per
mm2. Adhesion is expressed as
mean±SEM in eight random fields, in
two separate experiments.

haematologica | 2018; 103(5) 901


P.H. Mangin et al.

mouse platelets on fibrinogen is blocked by the Src kinase fibers in human GPVI transgenic mouse platelets was
inhibitor PP2 but not by the Syk kinase inhibitor blocked by PRT060318 (Figure 4C). The ability of Src and
PRT060318 (Figure 4B). Morphological changes of mouse Syk inhibitors to block spreading of human platelets and
platelets on fibrinogen are also not altered in platelets human GPVI transgenic mouse platelets on fibrinogen is
from irradiated mice transplanted with Syk-deficient fetal consistent with platelet activation by GPVI. This is sup-
liver (Figure 4B) or from PF4.Cre-Sykfl/fl mice (Online ported by demonstration of phosphorylation of the FcR γ-
Supplementary Figure S1). Western blotting for Syk con- chain. The limited spreading of mouse platelets on fibrino-
firmed lack of expression of the tyrosine kinase in the two gen is mediated through a Src-dependent but Syk-inde-
transgenic models (Figure 4B and not shown). Thrombin pendent pathway. Together, these results support a model
stimulated full spreading of wild-type and Syk-deficient in which immobilized fibrinogen activates human but not
platelets (Figure 4B). Formation of lamellipodia and stress mouse platelets through GPVI.

A
Figure 2. Human but not mouse glyco-
protein VI supports platelet adhesion
and spreading on immobilized fibrino-
gen. (A). Washed platelets from wild-
type mice (WT mice), GPVI-deficient
mice (GPVI-/- mice) or from healthy
donors (Human) were allowed to adhere
to human or mouse fibrinogen (FGN) for
30 or 45 min, respectively, and fixed
with PFA and stained with Alex-488-
phalloidin (4 μg/mL). (A)(i).
Representative epifluorescence images
of washed platelets adhering to fibrino-
gen. Scale bars represent 10 µm. (A)(ii).
Bar graph representing the number of
platelets adhering to immobilized fib-
rinogen per mm². Adhesion is expressed
as mean±SEM in five random fields, in
three separate experiments (two-way
ANOVA, Bonferroni post-hoc test:
P>0.05). (A)(iii). Bar graph representing
the percentage of platelets spreading
on immobilized fibrinogen. Spreading is
expressed as the mean±SEM in five ran-
dom fields, in six separate experiments.
Significance was attained using a two-
way ANOVA, Bonferroni post-hoc test:
****P<0.001. (B). Washed control (WT)
or β3-deficient (β3-/-) platelets were
allowed to adhere to fibrinogen for 60
min, fixed with PFA and stained with
TRITC-phalloidin (2 μg/mL). (B)(i).
Representative epifluorescence images
of washed mouse platelets adhering to
fibrinogen. Scale bars represent 10 μm.
(B)(ii). Bar graph representing the num-
ber of platelets adhering to immobilized
B fibrinogen per mm². Adhesion is
expressed as the mean±SEM in eight
random fields, in four separate experi-
ments (Mann-Whitney test,
**P<0.001). (C). Washed platelets from
wild-type mice (WT mice) or mice
expressing human GPVI (hGPVI mice)
were allowed to adhere to fibrinogen for
60 min, fixed with PFA and stained with
TRITC-phalloidin (2 μg/mL). (C)(i).
Representative epifluorescence images
of washed platelets adhering to fibrino-
gen. Scale bars represent 10 μm. (C)(ii).
Bar graph (left) representing the num-
ber of platelets adhering to immobilized
C fibrinogen per mm². Adhesion is
expressed as the mean±SEM in eight
random fields, in four separate experi-
ments (Mann-Whitney test, P>0.05).
Bar graph (right) representing the per-
centage of platelets spreading on immo-
bilized fibrinogen. Spreading is
expressed as the mean±SEM in eight
random fields, in four separate experi-
ments (Mann-Whitney test,
**P<0.001).

902 haematologica | 2018; 103(5)


Fibrinogen, GPVI and platelet activation

Fibrinogen stimulates an increase of Ca2+ in human platelets and was blocked in the presence of PRT-060318
glycoprotein VI-transgenic mouse platelets (Figure 5Ai-ii) highlighting the critical role of Syk in Ca2+
The observation that platelets expressing human but mobilization. These results demonstrate that human GPVI
not mouse GPVI undergo full spreading suggests that sig- stimulates Ca2+ signaling in fibrinogen-adherent mouse
nals from human GPVI are of significance. To investigate platelets.
this, a dual-dye Ca2+ assay was used to monitor cytoplas-
mic Ca2+ levels as a marker of PLCγ2 activation. Analysis Fab 9O12 blocks aggregate growth of humanized
of single platelet Ca2+ profiles by confocal microscopy glycoprotein VI mouse platelets
highlighted that signals generated on fibrinogen are com- Fibrinogen plays a critical role in hemostasis and arterial
posed of Ca2+ spikes (Figure 5A). The number of Ca2+ thrombosis through crosslinking of platelets in the grow-
spikes in mouse platelets expressing human GPVI was sig- ing thrombus. In addition, we now show that fibrinogen
nificantly increased relative to the number in wild-type induces platlet activation by GPVI. To establish whether

Figure 3. Monomeric but not dimeric GPVI binds to


immobilized fibrinogen and supports cellular adhe-
sion. (A)(i). IF-1 purified fibrinogen was immobilised
to a COOH-V chip surface using amine coupling
covalent linkage, to a level of 3,825 RU. Monomeric
or dimeric GPVI was titrated over three to four orders
of magnitude across the chip surface to a maximum
of 1 µmol/L, generating a single binding curve (1 RU
represents the binding of approximately 1 pg/mm2
protein). The graphs shown are representative of
three repeats and the KD is expressed as mean ±
SEM. (A)(ii). Solid-phase binding assays were per-
formed in Nunc maxisorb 96-well plates coated
overnight with BSA and fibrinogen (FGN).
Monomeric or dimeric GPVI (100 nmol/L) was incu-
bated as described. Bound GPVI was detected using
HRP-coupled to an anti-6×His monoclonal antibody
for monomeric GPVI or an anti-human IgG for dimer-
ic GPVI. The histogram (mean ± SD) shows the
results from five independent experiments.
Significance was determined using a one-way
ANOVA, Dunnett post-hoc test : *P<0.05. (B). RBL-
2H3 cells (3x105 cells/mL; 300 μL) transduced with
an empty vector (RBL-2H3 Ctrl) or with human full-
length GP6 cDNA (RBL-2H3 huGPVI) were pre-incu-
bated with PBS, REOPRO (20 μg/mL), and 9O12 (50
μg/mL) and allowed to adhere to immobilized fib-
rinogen for 1 h at 37°C, in 5% CO2. After three gentle
washing steps, cells were permeabilised with TRI-
TON x100 0.2% and stained with Alexa-568 phal-
loidin (1.5 U/mL). (B)(i). Representative epifluores-
cence images of RBL cells adhering to immobilized
fibrinogen. (B)(ii). and (B)(iii). Cells were manually
counted in six to eight different experiments
(*P<0.05 Mann Whitney t-test in (ii) and one-way
ANOVA was followed by the Bonferroni multiple com-
parison test).

haematologica | 2018; 103(5) 903


P.H. Mangin et al.

activation of GPVI by platelet-bound fibrinogen partici- then perfused additional blood from the same donor over
pates in platelet aggregation we performed an in vitro flow the aggregate at a wall shear rate of 300 s-1 in the presence
adhesion assay under conditions that prevent activation of or absence of the Fab fragment of the GPVI blocking mon-
GPVI by collagen and by fibrin. To achieve this, we gener- oclonal antibody 9O12. As expected, we were unable to
ated a platelet aggregate over type I fibrillar collagen using detect the presence of fibrin in the aggregate using a spe-
hirudin-treated blood to prevent formation of fibrin. We cific antibody (data not shown). The aggregate continued to

Figure 4. Syk promotes platelet spreading


on fibrinogen downstream of glycoprotein
VI but not in integrin aIIbβ3 outside-in sig-
naling. Washed human platelets, or
washed platelets from wild-type mice (WT
mice), Syk chimera mice (Syk chimera mice)
and mice expressing human GPVI (hGPVI
mice) were allowed to adhere to fibrinogen
or to collagen in the presence of either the
Src inhibitor PP2 (20 μmol/L), the Syk
inhibitor (5 μmol/L), thrombin (0.1 U/mL),
or vehicle control, for 30 min (human
platelets) or 45 min (mouse platelets) at
37°C followed by fixation with PFA. (A)(i).
Representative DIC images of human
platelets adhering to fibrinogen or collagen.
Scale bars represent 5 μm. (A)(ii). Bar graph
B
representing the surface area of platelets
spreading on fibrinogen. Spreading is
expressed as the mean±SEM in five or more
random fields, in three separate experi-
ments. Significance was determined by one-
way ANOVA and Bonferroni multiple compar-
ison test: *P<0.01. (A)(iii) Representative
western blot from human platelets following
adhesion to fibrinogen. Following immuno-
precipitation of Syk, proteins were separat-
ed by sodium dodecyl sulfate-polyacry-
lamide gel electrophoresis and western blot
for phosphotyrosine. Membranes were then
stripped and reprobed for Syk to confirm
equal protein loading. Blots are representa-
tive of three separate experiments. (B)(i).
Representative DIC images of mouse
platelets adhering to fibrinogen. Scale bars
represent 5 μm. (B)(ii). Bar graph represent-
ing the surface area of platelets spreading
on fibrinogen. Spreading is expressed as
the mean±SEM in five or more random
fields, in three separate experiments.
Significance was determined using one-way
ANOVA, with the Bonferroni post-hoc test:
*P<0.05, **P<0.001. (B)(iii). Expression of
Syk was measured by western blotting
platelet whole-cell lysates with an a-Syk
antibody. Membranes were then stripped
and reprobed with an anti-a-tubulin anti-
C body to confirm equal protein loading. Blots
are representative of three separate experi-
ments. (C)(i). Representative epifluores-
cence images of mouse platelets adhering
to fibrinogen. Scale bars represent 10 µm.
(C)(ii). Bar graph representing the surface
area of platelets spreading on fibrinogen.
Spreading is expressed as the mean±SEM
in eight random fields, in five separate
experiments (Mann-Whitney test,
**P<0.01).

904 haematologica | 2018; 103(5)


Fibrinogen, GPVI and platelet activation

grow in the presence of a Fab control but was dramatically gen to bind GPVI in suspension due to conformational dif-
inhibited in the presence of Fab 9O12 (Figure 6A and ferences between circulating and immobilized fibrinogen.
Online Supplementary Figure S2). In contrast, and as previ- Alternatively it may be due to the inability of the dimeric
ously shown, blockade of GPVI did not impair aggrega- fibrinogen to cluster GPVI on the platelet surface in sus-
tion measured by light transmission aggregometry in pension or because of a dependency on binding to integrin
response to ADP, U46619 and thrombin.35 These results aIIbβ3. While the affinity of fibrinogen for GPVI is in the
demonstrate a critical role for GPVI in aggregate growth range of that for collagen for GPVI,44,45 we have shown that
under flow when the roles of collagen and fibrin are negat- fibrinogen (and fibrin) bind selectively to monomeric
ed. In contrast, fibrinogen does not induce activation of GPVI and this would not be sufficient to induce activation
platelets in suspension either because the interaction is because of the absence of crosslinking. The reason why
dependent on activation of integrin aIIbβ3 or because it human platelets, but not mouse platelets, spread on fib-
cannot crosslink GPVI. rinogen is unclear. Based on the fact that human and
mouse GPVI share 64% homology,33 one could imagine
that only human GPVI binds to fibrinogen or that both
Discussion bind to this adhesive protein but only human GPVI is able
to promote activation.
In this study we show that human GPVI binds to immo- GPVI is primarily known as the major signaling receptor
bilized fibrinogen and that this leads to intracellular sig- for collagen. However, in recent years, GPVI has been
nals, which drive the formation of lamellipodial sheets shown to bind to other ligands including laminin, the
and stress fibers in human platelets and in human GPVI- transmembrane protein emmprin, adiponectin, histones
expressing mouse platelets. This explains the previously and fibrin.6-8,10,47 The physiological significance of many of
paradoxical observation that only human platelets form these interactions is uncertain, in part because of their low
lamellipodial sheets and stress fibers on a fibrinogen sur- affinity or because of whether they acutally occur in vivo.
face, despite mouse platelets being able to form both actin The interaction that has received the greatest attention is
structures in the presence of G protein-coupled receptor that with fibrin which lies at the interface of the core and
agonists such as thrombin. We also show that the interac- shell of the growing platelet aggregate.7,8 This interaction
tion of fibrinogen with GPVI is important for aggregate
growth providing a new understanding of hemostasis and
thrombosis.
We recently identified fibrin as a novel ligand for
GPVI7,8,42 and have shown that binding resides in the D- A
dimer region.30 The observation that fibrinogen also acti-
vates GPVI should not, therefore, be a surprise.
Nevertheless, this was unexpected and came from the
observation that human platelets deficient in GPVI adhere
to but do not spread on fibrinogen. This raises the ques-
tion as to why this has been previously overlooked. One
reason is that mouse platelets do not spread on fibrinogen
and thus there is no defect in the absence of GPVI. A sec-
ond reason is the low level of phosphorylation of the FcR
γ-chain induced by fibrinogen in human platelets relative
to that by collagen and other GPVI-agonists. This may
reflect the extent to which each ligand is able to cluster
GPVI and, in the case, of fibrinogen, the dependency on
the interaction with integrin aIIbβ3. A third reason is that
fibrinogen binds selectively to monomeric GPVI whereas
the original binding studies were performed with dimeric
GPVI.7,8 It is worth noting that we have reported a reduced
number of dimers on immobilized fibrinogen relative to
collagen.42
Adhesion of human platelets to fibrinogen is dependent
on integrin aIIbβ3. At present, it is not known whether
binding to integrin aIIbβ3 is critical for activation of GPVI
or simply to promote adhesion such that activation of
GPVI can occur. As a dimer, fibrinogen should be able to
Figure 5. Human glycoprotein VI, but not integrin aIIbβ3 plays a major role in
the regulation of the calcium signaling after platelet adhesion to fibrinogen.
bind two GPVI monomers, but alternatively the interac-
tion with integrin aIIbβ3 may be required to support acti- Washed platelets from wild-type (WT) or mice expressing human GPVI (hGPVI)
vation of monomeric GPVI. A similar role for an integrin were loaded with Oregon-green Bapta-1-AM and Calcein red orange and
in the activation of an ITAM receptor has been reported in deposited on immobilized fibrinogen (100 μg/mL). Modifications in fluores-
cence of individual adherent platelets were monitored for 7 min by confocal
microscopy and the Ca2+ concentrations were determined as detailed in the
other hematopoietic cells with the postulate that the inte-
grin and the ITAM receptor would be associated via a link- Methods section. (A)(i). Typical time-course Ca2+ profile of one representative
er protein.43 platelet adhering to fibrinogen. (A)(ii). Dot plot representing the number of cal-
cium spikes over a period of 5 min. Each point represents an individual
platelet. The results are presented as the mean±SEM of five independent
Fibrinogen is present in whole blood at a concentration
of 2 - 4 mg/mL but does not induce platelet activation. experiments (one-way ANOVA, Bonferroni post-hoc test, ***P<0.001).
This may be explained by an inability of soluble fibrino-

haematologica | 2018; 103(5) 905


P.H. Mangin et al.

Figure 6. Blockade of human glycoprotein VI limits platelet accumulation to a growing aggregate. (A)(i). Hirudinated human whole blood labeled with DIOC6 (1
μmol/L) was perfused over immobilized collagen (200 μg/mL) to preform aggregates for 2 min 30 s, before perfusing hirudinated blood from the same donor in the
presence of the Alexa Fluor 647-conjugated monoclonal antibody against GPIbβ (5 μg/mL) and with a Fab control (Control) or the blocking anti-GPVI antibody 9O12
(50 μg/mL). (A)(i). Representative 3D reconstructions from confocal images of aggregates obtained after 7 min 30 s of blood perfusion at 300 s-1. Preformed aggre-
gates are represented in gray, aggregates formed in the presence of a Fab control are represented in red and aggregates formed in the presence of the Fab 9O12
are depicted in orange. (A)(ii). Bar graph representing the volume of the platelet aggregates (mean±SEM) in eight random fields, in six separate experiments per-
formed with different blood donors (Mann-Whitney test, ***P<0.001). The gray, red and orange colors represent the volume of the preformed aggregates, the aggre-
gates formed in the presence of a Fab control and the aggregates formed in the presence of the Fab 9O12, respectively.

takes place at a critical checkpoint in aggregate consolida- can be explained by redundancy in pathways of platelet
tion and aggregate growth. Thus, GPVI has the potential activation, with the GPIb-von Willebrand factor axis ini-
to both initiate and propagate thrombus formation tiating hemostasis, and ADP, thromboxane and thrombin
through interactions with collagen and fibrin and, it inducing powerful activation. Additionally, the reactive
appears now, also with fibrinogen. Moreover, while colla- fibrillar type I and III collagen present in deeper layers of
gen and fibrin are localized at the base of the thrombus the vessels would limit the role of GPVI in the hemosta-
and in the core, respectively, fibrinogen is found through- sic response following superficial injury. On the other
out the aggregate. This suggests a model in which throm- hand, the discovery that fibrin and immobilized fibrino-
bus growth could be sustained by GPVI/fibrinogen, poten- gen activate GPVI may be of significance at sites of fib-
tially in association with other adhesive proteins. Indeed, rinogen deposition or fibrin formation in diseased vessels
in addition to fibrinogen other adhesive proteins such as following inflammation or loss of vascular integrity. The
von Willebrand factor and fibronectin have been shown to ability of fibrin and immobilized fibrinogen to activate
support thrombus growth.47–50 Whether these proteins par- GPVI may also reflect yet-to-be-discovered new roles for
ticipate in GPVI-mediated platelet aggregation is unclear GPVI.
since von Willebrand factor is not known to be a ligand of In conclusion, in the present study, we have identified
GPVI and fibronectin does not directly promote platelet immobilized fibrinogen as a novel activator of human but
adhesion and activation through GPVI.51 Selective inhibi- not mouse GPVI and have shown that this interaction
tion of the interaction of GPVI with collagen, fibrinogen supports platelet aggregation under flow. This further
and fibrin is required to establish their respective contribu- emphasizes the contribution of GPVI to platelet activation
tions to platelet activation in hemostasis and thrombosis. in thrombosis.
The discovery that GPVI initiates and propagates
platelet aggregation at sites of vessel injury suggests a Acknowledgments
major role in hemostasis and thrombosis. Paradoxically, This work was supported by the British Heart Foundation
however, mice and humans deficient in GPVI only have (RG/13/18/30563); SPW holds a BHF Chair (CH03/003) and
at most a mild bleeding diathesis, which in the case of ATH holds a BHF Studentship (FS/15/71/31677). NLL’s con-
humans may be due to additional confounders such as a tract was funded by the Agence Nationale pour la Recherche
low platelet count as seen in patients with immune- ANR-14-CE35-0022-02. The authors would like to thank Victor
induced thrombocytopenia caused by antibodies to Tybulewicz and Edina Schweighoffer for providing critical
GPVI. The relatively minor role of GPVI in hemostasis reagents.

2. Ezumi Y, Shindoh K, Tsuji M, Takayama H. chain downstream of glycoprotein VI in


References Physical and functional association of the Src murine platelets, and Lyn regulates a novel
family kinases Fyn and Lyn with the colla- feedback pathway. Blood. 2000;96(13):4246-
1. Suzuki-Inoue K, Tulasne D, Shen Y, et al. gen receptor glycoprotein VI-Fc receptor γ 4253.
Association of Fyn and Lyn with the proline- chain complex on human platelets. J Exp 4. Severin S, Nash CA, Mori J, et al. Distinct
rich domain of glycoprotein VI regulates Med. 1998;188(2):267-276. and overlapping functional roles of Src fam-
intracellular signaling. J Biol Chem. 3. Quek LS, Pasquet JM, Hers I, et al. Fyn and ily kinases in mouse platelets. J Thromb
2002;277(24):21561-21566. Lyn phosphorylate the Fc receptor gamma Haemost. 2012;10(8):1631-1645.

906 haematologica | 2018; 103(5)


Fibrinogen, GPVI and platelet activation

5. Nieswandt B, Watson SP. Platelet-collagen 22. Obergfell A, Judd BA, Del Pozo MA, O’Callaghan CA, Watson SP. Critical role for
interaction: is GPVI the central receptor? Schwartz MA, Koretzky GA, Shattil SJ. The an acidic amino acid region in platelet signal-
Blood. 2003;102(2):449-461. molecular adapter SLP-76 relays signals from ing by the HemITAM (hemi-immunorecep-
6. Inoue O, Suzuki-Inoue K, McCarty OJ, et al. platelet integrin aIIbβ3 to the actin tor tyrosine-based activation motif) contain-
Laminin stimulates spreading of platelets cytoskeleton. J Biol Chem. 2001;276(8): ing receptor CLEC-2 (C-type lectin receptor-
through integrin alpha6beta1-dependent 5916-5923. 2). J Biol Chem. 2013;288(7):5127-5135.
activation of GPVI. Blood. 2006;107(4):1405- 23. Goncalves I, Hughan SC, Schoenwaelder 38. Hui KY, Haber E, Matsueda GP,Monoclonal
1412. SM, Yap CL, Yuan Y, Jackson SP. Integrin antibodies to a synthetic fibrin-like peptide
7. Mammadova-bach E, Ollivier V, Loyau S, et aIIbβ3-dependent calcium signals regulate bind to human fibrin but not fibrinogen.
al. Platelet glycoprotein VI binds to polymer- platelet-fibrinogen interactions under flow: Science 1983,222(4628):1129-1132.
ized fibrin and promotes thrombin genera- Involvement of phospholipase Cγ2. J Biol 39. Cazenave J-P, Ohlmann P, Cassel D, Eckly A,
tion. Blood. 2016;126(5):683-692. Chem. 2003;278(37):34812-34822. Hechler B, Gachet C. Preparation of washed
8. Alshehri OM, Hughes CE, Montague S, et 24. Wonerow P, Pearce AC, Vaux DJ, Watson SP. platelet suspensions from human and rodent
al. Fibrin activates GPVI in human and A critical role for phospholipase Cγ2 in blood. Methods Mol Biol. 2004;272:13-28.
mouse platelets. Blood. 2015;126(13):1601- aIIbβ3-mediated platelet spreading. J Biol 40. Schaff M, Receveur N, Bourdon C, et al.
1608. Chem. 2003;278(39):37520-37529. Novel function of tenascin-C, a matrix pro-
9. Riba R, Hughes CE, Graham A, Watson SP, 25. Watson SP, Auger JM, McCarty OJ, Pearce tein relevant to atherosclerosis, in platelet
Naseem KM. Globular adiponectin induces AC. GPVI and integrin aIIbβ3 signaling in recruitment and activation under flow.
platelet activation through the collagen platelets. J Thromb Haemost. 2005;3(8): Arterioscler Thromb Vasc Biol. 2011;31(1):
receptor GPVI-Fc receptor chain complex. J 1752-1762. 117-124.
Thromb Haemost. 2008;6(6):1012-1020. 26. Boylan B, Gao C, Rathore V, Gill JC, 41. Haimovich B, Lipfert L, Brugge JS, Shattil SJ.
10. Seizer P, Borst O, Langer HF, et al. EMM- Newman DK, Newman PJ. Identification of Tyrosine phosphorylation and cytoskeletal
PRIN (CD147) is a novel receptor for platelet FcgammaRIIa as the ITAM-bearing receptor reorganization in platelets are triggered by
GPVI and mediates platelet rolling via GPVI- mediating alphaIIbbeta3 outside-in integrin interaction of integrin receptors with their
EMMPRIN interaction. Thromb Haemost. signaling in human platelets. Blood. immobilized ligands. J Biol Chem.
2009;101(4):682-686. 2008;112(7):2780-2786. 1993;268(21):15868-15877.
11. Schaff M, Tang CJ, Maurer E, et al. Integrin 27. Hughes CE, Finney BA, Koentgen F, Lowe 42. Poulter NS, Pollitt AY, Owen DM, et al.
6 1 is the main receptor for vascular KL, Watson SP. The N-terminal SH2 domain Clustering of glycoprotein VI (GPVI) dimers
laminins and plays a role in platelet adhe- of Syk is required for (hem) ITAM, but not upon adhesion to collagen as a mechanism
sion, activation, and arterial thrombosis. integrin, signaling in mouse platelets. Blood. to regulate GPVI signaling in platelets. J
Circulation. 2013;128(5):541-552. 2015;125(1):144-155. Thromb Haemost. 2017;15(3):549-564.
12. Boulaftali Y, Hess PR, Getz TM, et al. 28. Zhi H, Rauova L, Hayes V, et al. Cooperative 43. Evans R, Patzak I, Svensson L, et al. Integrins
Platelet ITAM signaling is critical for vascu- integrin/ITAM signaling in platelets in immunity. J Cell Sci. 2009;122(2):215-225.
lar integrity in inflammation. J Clin Invest. enhances thrombus formation in vitro and 44. Miura Y, Takahashi T, Jung SM, Moroi M.
2013;123(2):908-916. in vivo. Thromb Haemost. 2013;121(10): Analysis of the interaction of platelet colla-
13. Kuijpers MJE, Gilio K, Reitsma S, et al. 1858-1867. gen receptor glycoprotein VI (GPVI) with
Complementary roles of platelets and coag- 29. Law DA, DeGuzman FR, Heiser P, collagen: a dimeric form of GPVI, but not
ulation in thrombus formation on plaques Ministri-Madrid K, Killeen N, Phillips DR. the monomeric form, shows affinity to
acutely ruptured by targeted ultrasound Integrin cytoplasmic tyrosine motif is fibrous collagen. J Biol Chem. 2002;277
treatment: a novel intravital model. J required for outside-in aIIbβ3 signalling (48):46197-46204.
Thromb Haemost. 2009;7(1):152-161. and platelet function. Nature. 1999;401 45. Loyau S, Dumont B, Ollivier V, et al. Platelet
14. Hechler B, Gachet C. Comparison of two (6755):808-811. glycoprotein VI dimerization, an active
murine models of thrombosis induced by 30. Onselaer M-B, Hardy AT, Wilson C, et al. process inducing receptor competence, is an
atherosclerotic plaque injury. Thromb Fibrin and D-dimer bind to monomeric indicator of platelet reactivity. Arterioscler
Haemost. 2011;105 (Suppl):S3-12. GPVI. Blood Adv. 2017; 1(19):1495-1504. Thromb Vasc Biol. 2012;32(3):778-785.
15. Bender M, Hagedorn I, Nieswandt B. 31. Kato K, Kanaji T, Russell S, et al. The contri- 46. Aslan JE, Phillips KG, Healy LD, et al.
Genetic and antibody-induced glycoprotein bution of glycoprotein VI to stable platelet Histone deacetylase 6-mediated deacetyla-
VI deficiency equally protects mice from adhesion and thrombus formation illustrat- tion of a-tubulin coordinates cytoskeletal
mechanically and FeCl3-induced thrombo- ed by targeted gene deletion. Blood. and signaling events during platelet activa-
sis. J Thromb Haemost. 2011;9(7):1423- 2003;102(5):1701-1707. tion. AJP Cell Physiol. 2013;305(12):C1211-
1426. 32. Mangin PH, Tang C, Bourdon C, et al. A C1213.
16. Nieswandt B, Schulte V, Bergmeier W, et al. humanized glycoprotein VI (GPVI) mouse 47. Kulkarni S, Dopheide SM, Yap CL, et al. A
Long-term antithrombotic protection by in model to assess the antithrombotic effica- revised model of platelet aggregation. J Clin
vivo depletion of platelet glycoprotein VI in cies of anti-GPVI agents. J Pharmacol Exp Invest. 2000;105(6):783-791.
mice. J Exp Med. 2001;193(4):459-469. Ther. 2012;341(1):156-163. 48. Yang H, Reheman A, Chen P, Zhu G.
17. Matus V, Valenzuela G, Sáez CG, et al. An 33. Jandrot-Perrus M, Busfield S, Lagrue AH, et Fibrinogen and von Willebrand factor-inde-
adenine insertion in exon 6 of human GP6 al. Cloning, characterization, and functional pendent platelet aggregation in vitro and in
generates a truncated protein associated studies of human and mouse glycoprotein vivo. J Thromb Haemost. 2006;4(10):2230-
with a bleeding disorder in four Chilean VI: a platelet-specific collagen receptor from 2237.
families. J Thromb Haemost. the immunoglobulin superfamily. Blood. 49. Reheman A, Yang H, Zhu G, et al. Plasma
2013;11(9):1751-1759. 2000;96(5):1798-1807. fibronectin depletion enhances platelet
18. Dumont B, Lasne D, Rothschild C, et al. 34. Perrault C, Moog S, Rubinstein E, et al. A aggregation and thrombus formation in
Absence of collagen-induced platelet activa- novel monoclonal antibody against the mice lacking fibrinogen and von Willebrand
tion caused by compound heterozygous extracellular domain of GPIbbeta modulates factor plasma fibronectin depletion
GPVI mutations. Blood. 2009;114(9):1900- vWF mediated platelet adhesion. Thromb enhances platelet aggregation and thrombus
1903. Haemost. 2001;86(5):1238-1248. formation in mice lacking fibrinogen and
19. Hermans C, Wittevrongel C, Thys C, 35. Lecut C, Feeney LA, Kingsbury G, et al. von Willebrand factor. 2012;113(8):1809-
Smethurst PA, Van Geet C, Freson K. A com- Human platelet glycoprotein VI function is 1817.
pound heterozygous mutation in glycopro- antagonized by monoclonal antibody- 50. Cho J, Mosher DF. Impact of fibronectin
tein VI in a patient with a bleeding disorder. derived Fab fragments. J Thromb Haemost. assembly on platelet thrombus formation in
J Thromb Haemost. 2009;7(8):1356-1363. 2003;1(12):2653-2662. response to type I collagen and von
20. Zahid M, Mangin P, Loyau S, et al. The 36. Lebozec K, Jandrot-Perrus M, Avenard G, Willebrand factor. Blood. 2006;108(7):2229-
future of glycoprotein VI as an antithrom- Favre-Bulle O, Billiald P. Design, develop- 2236.
botic target. J Thromb Haemost. ment and characterization of ACT017, a 51. Maurer E, Schaff M, Receveur N, Bourdon
2012;10(12):2418-2427. humanized Fab that blocks platelet’s glyco- C, Mercier L, Nieswandt B. Fibrillar cellular
21. Bennett JS, Berger BW, Billings PC. The protein VI function without causing bleed- fibronectin supports efficient platelet func-
structure and function of platelet integrins. J ing risks. MAbs. 2017:9(6):945-958. tion and procoagulant activity. Thromb
Thromb Haemost. 2009;7(Suppl 1):200-205. 37. Hughes CE, Sinha U, Pandey A, Eble JA, Haemost. 2015;114(6):1175-1188.

haematologica | 2018; 103(5) 907


ARTICLE Coagulation & its Disorders

Circulating microRNAs as biomarkers


Ferrata Storti
Foundation of disease and typification of the
atherothrombotic status in antiphospholipid
syndrome
Carlos Pérez-Sánchez,1* Iván Arias-de la Rosa,1* María Ángeles Aguirre,1,2
María Luque-Tévar,1 Patricia Ruiz-Limón,1 Nuria Barbarroja,1
Yolanda Jiménez-Gómez,1 María Carmen Ábalos-Aguilera,1
Eduardo Collantes-Estévez,1,2,3 Pedro Segui,1,4 Francisco Velasco,5
María Teresa Herranz,6 Jesús Lozano-Herrero,6 María Julia Hernandez-Vidal,6
Haematologica 2018
Volume 103(5):908-918 Constantino Martínez,7 Rocío González-Conejero,7 Massimo Radin,8
Savino Sciascia,8 Irene Cecchi,8 María José Cuadrado9** and Chary López-
Pedrera1,2**

1
Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Spain; 2Unidad
de Gestión Clínica Reumatología, Hospital Universitario Reina Sofía, Córdoba, Spain;
3
Departamento de Medicina (Medicina, Dermatología y Otorrinolaringología),
Universidad de Córdoba, Spain; 4Unidad de Gestión Clínica Radiología, Hospital
Universitario Reina Sofía, Córdoba, Spain; 5Unidad de Gestión Clínica Hematología,
Hospital Universitario Reina Sofía, Córdoba, Spain; 6Servicio de Medicina Interna,
Hospital Morales Meseguer, Murcia, Spain; 7Centro Regional de Hemodonación,
Universidad de Murcia, IMIB-Arrixaca, Spain; 8Department of Clinical and Biological
Sciences, Center of Research of Immunopathology and Rare Diseases, Torino, Italy and
9
Lupus Research Unit and St. Thomas’ Hospital, London, UK
*CP-S and IA-R shared first authorship and contributed equally to this work. **MJC and CL-P shared last author-
ship and contributed equally to this work.

ABSTRACT

W
e aimed to identify the plasma miRNA profile of antiphospho-
lipid syndrome (APS) patients and to investigate the potential
role of specific circulating miRNAs as non-invasive disease bio-
Correspondence: markers. Ninety APS patients and 42 healthy donors were recruited.
rosario.lopez.exts@juntadeandalucia.es Profiling of miRNAs by PCR-array in plasma of APS patients identified
a set of miRNAs differentially expressed and collectively involved in
clinical features. Logistic regression and ROC analysis identified a signa-
ture of 10 miRNA ratios as biomarkers of disease. In addition, miRNA
Received: November 10, 2017.
signature was related to fetal loss, atherosclerosis, and type of thrombo-
Accepted: February 22, 2018. sis, and correlated with parameters linked to inflammation, thrombosis,
Pre-published: March 15, 2018. and autoimmunity. Hard clustering analysis differentiated 3 clusters rep-
resenting different thrombotic risk profile groups. Significant differences
between groups for several miRNA ratios were found. Moreover,
doi:10.3324/haematol.2017.184416 miRNA signature remained stable over time, demonstrated by their
analysis three months after the first sample collection. Parallel analysis
Check the online version for the most updated in two additional cohorts of patients, including thrombosis without
information on this article, online supplements, autoimmune disease, and systemic lupus erythematosus without
and information on authorship & disclosures: antiphospholipid antibodies, each displayed specific miRNA profiles
www.haematologica.org/content/103/5/908 that were distinct from those of APS patients. In vitro, antiphospholipid
antibodies of IgG isotype promoted deregulation in selected miRNAs
and their potential atherothrombotic protein targets in monocytes and
©2018 Ferrata Storti Foundation endothelial cells. Taken together, differentially expressed circulating
Material published in Haematologica is covered by copyright. miRNAs in APS patients, modulated at least partially by antiphospho-
All rights are reserved to the Ferrata Storti Foundation. Use of lipid antibodies of IgG isotype, might have the potential to serve as
published material is allowed under the following terms and
conditions:
novel biomarkers of disease features and to typify patients’
https://creativecommons.org/licenses/by-nc/4.0/legalcode. atherothrombotic status, thus constituting a useful tool in the manage-
Copies of published material are allowed for personal or inter- ment of the disease.
nal use. Sharing published material for non-commercial pur-
poses is subject to the following conditions:
https://creativecommons.org/licenses/by-nc/4.0/legalcode,
sect. 3. Reproducing and sharing published material for com- Introduction
mercial purposes is not allowed without permission in writing
from the publisher. Antiphospholipid syndrome (APS) is a clinical disorder characterized by the
occurrence of thrombosis and/or pregnancy morbidity associated with the persist-
ent presence of antiphospholipid antibodies (aPL), including anti-cardiolipin anti-

908 haematologica | 2018; 103(5)


MicroRNAs as antiphospholipid syndrome biomarkers

Table 1. Clinical and laboratory parameters of the antiphospholipid syndrome (APS) patients and the healthy donors (HDs).
APS HDs P
(total n. 90) (total n. 42)
Females/males 48/42 22/20
Age, years 51.2 ± 13.1 46.2 ± 13.4 NS
Arterial thrombosis 35/90 0/42
Venous thrombosis 55/90 0/42
Recurrences 37/90 0/42
Pregnancy morbidity 23/90 0/42
Pathological CIMT 24/90 6/42 0.00
ABI – left* 1.3 ± 0.11 1.2 ± 0.09 0.02
ABI – right* 1.27 ± 0.11 1.2 ± 0.09 0.02
LA positivity 85/90 0/42 0.00
aCL-IgG# GPL 23.4 (0.5-448) 1.3 (0.5-5) 0.00
aCL-IgM# MPL 21.8 (0-354) 4.9 (0.8-17) 0.00
Anti-β2GPI IgG# SGU 23.9 (0-361) 1 (1-2) 0.02
Anti-β2GPI IgM# SMU 14.8 (0-289) 1.2 (1-2.6) 0.02
Antiplatelet agents† 30/90 0/42
Anticoagulant agents‡ 62/90 0/42
Total cholesterol level,* mg/dL 191.7 ± 32.06 190 ± 41.8 NS
Cholesterol HDL level,* mg/dL 51.09 ± 12.4 55.4 ± 13.1 NS
Cholesterol LDL level,* mg/dL 112.1 ± 33.2 118.4 ± 26.9 NS
Triglycerides level,* mg/dL 155.1 ± 163.2 88.3 ± 50.07 NS
ESR,* mm/h 13.5 ± 13.6 6.6 ± 4 0.05
n: number; NS: not significant; CIMT: carotid intima-media thickness; ABI: ankle brachial index; LA: lupus anticoagulant; aCL: anti-cardiolipin antibodies; GPL: IgG phospholipid
units; MPL: IgM phospholipid units; anti-β2GPI: anti-β2 glycoprotein 1 antibodies; SGU: stantard IgG units; SMU: standard IgM units; HDL: high-density lipoprotein; LDL: low-density
lipoprotein; ESR: erythrocyte sedimentation rate. *Results expressed as mean±Standard Deviation. #Results expressed in mean and values range. †Antiplatelet agents include
acetylsalicylic acid and clopidogrel. ‡Anticoagulant agents indicate vitamin K antagonists, including warfarin and acenocumarol.

bodies (aCL), anti-β2-glycoprotein 1 antibodies (anti- In the setting of APS, a previous study by our group
β2GPI) and/or lupus anticoagulant (LA). Cardiac, cerebral recognized that aPL modulate the expression of 2
and vascular strokes in these patients are responsible for a miRNAs in monocytes (miR-19b and miR-20a) that con-
significant reduction in life expectancy.1 The course of car- trol the expression of key proteins involved in the
diovascular disease (CVD) in APS patients may rapidly pathology of the disease, such as tissue factor (TF).8
change from asymptomatic to severe life-threatening Moreover, we recently demonstrated that both aPL and
manifestations that are difficult to deal with. Timely diag- the anti-double stranded DNA antibodies (anti-dsDNA)
nosis and accurate monitoring of the course of APS are promote specific changes in the expression of proteins
essential to improve the quality of therapy and avoid related to the biogenesis of miRNAs in leukocytes of APS
approaches based on medical empirical protocols. In the and systemic lupus erythematosus (SLE) patients, which
same way, like many other autoimmune diseases, APS is are translated in the altered expression of the miRNAs
a heterogeneous entity, and this has a dramatic impact on profile and that of their protein targets (related to CVD)
diagnosis and treatment.2 in these disorders.9
Understanding of the pathophysiological mechanisms Extensive analyses have shown that miRNAs are
explaining how atherosclerosis and CVD are associated to released into the circulation where they are present in con-
APS has been greatly broadened with the application of centration levels that differ between healthy subjects and
genomic technologies.3 One emerging and important patients. Although little is known about the origin and
mechanism controlling gene expression is epigenetics, function of such circulating miRNAs, these molecules are
which regulates gene packaging and independent expres- increasingly recognized as non-invasive and readily acces-
sion of alterations in the DNA sequence. Epigenetics, sible biomarkers for risk stratification, diagnosis and prog-
which comprises DNA methylation, histone modifica- nosis of multiple forms of CVD.10
tions, and microRNAs (miRNAs) activity, is providing Specific profiles of circulating miRNAs are also associat-
new directions linking genomics and environmental fac- ed to the pathophysiology of different systemic autoim-
tors.4 miRNAs are small, non-coding RNAs that, depend- mune diseases, including SLE, systemic sclerosis, and
ing upon base pairing to messenger RNA (mRNA), medi- rheumatoid arthritis (RA), and some of them appear to be
ate mRNA cleavage, translational repression or mRNA of diagnostic and, possibly, of prognostic value.11 To date,
destabilization. miRNAs are known to be involved in cru- in the context of APS, no study has analyzed the potential
cial cellular processes and their dysregulation has been role of the circulating miRNAs as biomarkers of the dis-
described in many cell types and fluids in a broad range of ease. Therefore, the present study was designed to deter-
diseases.5-7 mine the plasma miRNA specific profile of APS patients,

haematologica | 2018; 103(5) 909


C. Perez-Sánchez et al.

A B

Figure 1. Antiphospholipid syndrome (APS) patients showed a specific circulating miRNAs profile related to clinical features of this autoimmune disorder. (A) To
identify the changes that occurred in the expression levels of microRNAs (miR) in plasma from antiphospholipid syndrome versus controls, Human Serum & Plasma
miRNA PCR-array (Qiagen) was performed in the study cohort. Expression levels of 19 miRNAs were found up-regulated in antiphospholipid syndrome, while 20
miRNAs were down-regulated. (B) Ingenuity Pathway Analysis (IPA) uncovered the main enriched biological functions and pathways in which these microRNAs are
involved. The analysis included only the functions and pathways with average IPA score >2 [indicated as -log (P value)]. (C) Validation of selected miRNAs by RT-PCR
in the whole cohort of APS patients and healthy donors. *P<0.05.

their modulation by autoantibodies, and their potential ical parameters, B-Mode Ultrasound IMT and Ankle Brachial
role as non-invasive biomarkers of disease features. Index measurements see the Online Supplementary Appendix.

Isolation of miRNAs and analysis of miRNAs expression


Methods profiling
Total RNA, including the miRNA fraction, was extracted from
Patients both plasma and supernatants obtained from in vitro studies by
Ninety patients with primary APS and 42 healthy donors (HDs) using the QIAzol miRNeasy kit (Qiagen, Valencia, CA, USA) fol-
were included in this study over a period of 24 months (see Online lowing the manufacturer's instructions13 (Online Supplementary
Supplementary Appendix). All experimental protocols were Appendix). To identify the changes that occurred in the expression
approved by the ethics committee of the Reina Sofia Hospital, levels of miRNAs in plasma from APS patients and HDs, a Human
Cordoba, Spain, and written informed consent was obtained. The Serum & Plasma miRNA PCR-array (Qiagen) was performed
characteristics of patients and HDs are shown in Table 1. (Online Supplementary Appendix) on an exploratory cohort (Online
The adjusted global anti-phospholipid syndrome score Supplementary Table S2).
(aGAPSS) was calculated for each APS patient, as previously
described.12 Briefly, aGAPSS was calculated by adding the points Quantitative real-time PCR
corresponding to both the cardiovascular and thrombotic risk fac- A fixed volume of 3 μl of RNA solution from the 14 μl-eluate
tors, based on a linear transformation derived from the β regres- from RNA isolation of 200 μl plasma sample was used as input
sion coefficient as follows: 3 for hyperlipidemia, 1 for arterial into the reverse transcription. Input RNA was reverse transcribed
hypertension, 5 for aCL IgG/IgM, 4 for anti-β2GPI IgG/IgM, and 4 using the TaqMan miRNA Reverse Transcription kit and miRNA-
for LA. specific stem-loop primers (Life Technologies, Madrid, Spain)
Two additional cohorts of patients were further analyzed as dis- (Online Supplementary Appendix). The expression levels of miRNAs
ease control, including 23 patients with thrombosis in the absence were calculated by using 2-Ct and reciprocal ratios were performed
of an associated autoimmune disease [12 non-pregnant women [Ratio miR-A/miR-B = log2 (2-Ct miR-A/2-Ct miR-B)], as previously
and 11 men; mean age 44 (range: 21-73 years), including patients described.14-19 Reciprocal ratios analysis is an approach that
with objectively verified thrombotic events: 14 deep venous bypasses the controversial issue of data normalization of miRNAs
thrombosis and 9 thrombosis in intra-cerebral vessels], and 25 SLE in plasma (self-normalization). Furthermore, miRNAs whose con-
patients without aPLs (Online Supplementary Table S1). centrations are changed because of a pathology in opposite direc-
For details of blood sample collection and assessment of biolog- tions can be effective in differentiating investigated populations.

910 haematologica | 2018; 103(5)


MicroRNAs as antiphospholipid syndrome biomarkers

Figure 2. Interaction network of microRNAs identified potential mRNA targets involved in clinical features of antiphospholipid syndrome. Using microRNA Target
Filter of QIAGEN’s Ingenuity Pathway Analysis (IPA, QIAGEN Redwood City, CA, USA, www.qiagen.com/ingenuity), the software generated a network including the
selected microRNAs (miRNAs or miR) and their mRNA targets, filtered by coronary artery disease, thrombosis, abortion and cerebrovascular dysfunction. Only targets
experimentally observed and predicted with high confidence are shown and related by direct interactions to their specific miRNA regulators.

Target gene prediction and integrated analysis by Bioinformatic identification and analysis of
Ingenuity Pathway Analysis deregulated miRNAs related to the pathophysiology of
The altered miRNAs were further analyzed to obtain informa- APS and analysis of potential protein targets
tion about biological functions, pathways and networks by using In silico studies were performed to identify the altered
the web-based bioinformatics tool QIAGEN’s Ingenuity Pathway miRNAs that might have as potential targets a number of
Analysis (IPA; Ingenuity Systems, http://www.INGENUITY.com). genes/proteins involved in the development of clinical
For this purpose, all differentially regulated miRNAs and fold manifestations related to APS, such as coronary artery dis-
changes were imported into IPA20 (Online Supplementary Appendix). ease, thrombosis, abortion, and cerebrovascular dysfunc-
Details of purification of IgG, in vitro exposure of monocytes and tion. IPA identified 11 altered miRNAs as the main regula-
endothelial cells to aPL antibodies, and the statistical analysis are tors of proteins involved in the pathology of APS, includ-
available in the Online Supplementary Appendix. ing miRNA 34a-5p, 15a-5p, 145a-5p, 133b-3p, 124-3p,
206, 20a-5p, 19b-3p, 210-3p, 296-5p and 374a-5p. This set
Results of 11 miRNAs included, among others, the top 5 up-regu-
lated miRNAs and 3 out of the top 5 down-regulated
Differentially expressed miRNAs in the plasma of APS miRNAs in the PCR-array. The expression levels of the 11
patients and HDs selected miRNAs were analyzed in all study subjects by
In the discovery phase (exploratory cohort), we identi- RT-PCR (Figure 1B). MiR-124 and miR-34a were found
fied 39 miRNAs that were differentially expressed increased in APS patients in relation to healthy donors,
between APS patients and HDs (cut off: 1.7-fold change), while miR-20a, miR-19b and miR145a were found
including 19 up-regulated and 20 down-regulated (Figure reduced. The remaining microRNAs were also found to be
1A). Using the IPA software, the functional analysis of the altered, showing a trend to either increase or reduction as
altered miRNAs in APS patients showed that a large num- observed in the discovery phase, thus validating the data
ber of them had validated and putative target mRNAs obtained by PCR-array.
mainly involved in connective tissue disorders, inflamma- We further developed a network that defined the inter-
tory response, reproductive system disease, CVD or skele- action between miRNA-mRNA targets (Figure 2). Key pro-
tal and muscular disorders (Figure 1C). teins involved in the pathophysiology of APS, and identi-

haematologica | 2018; 103(5) 911


C. Perez-Sánchez et al.

Figure 3. A circulating miRNA signature in antiphospholipid syndrome (APS) might have potential value as biomarkers of disease. (A) Selected microRNAs (miRNAs
or miR) were analyzed in the whole cohort, including 90 APS patients and 42 healthy donors, and reciprocal ratios were performed. Beeswarm plot of each differen-
tially expressed miR ratio is shown, along with mean, Standard Deviation, and P-value. For statistical analysis, after normality and equality of variance tests, compar-
isons were made by paired Student t-test or a non-parametric test (Mann-Whitney rank sum test). (B) A combination of the 10 miRNA ratios as a panel was carried
out by using logistic regression on the data set. ROC curve of miRNA panel and cut off were generated based on the predicted probability (P) for each subject as a
single score. The equation used in our model was: “Combined miRNA-ratio panel [Logit(p)] = - 0.64 + 0.034x(miR-19b/miR-34a) + 1.061x(miR-19b/miR-15a) +
0.248x(miR-19b/miR-124) – 1.704x(miR-19b/miR-145) + 2.34x(miR-20a/miR-145) – 0.729x(miR-20a/miR-374a) – 0.624x(miR-20a/miR-210) + 0.088x(miR-
20a/miR-133b) + 0.166x(miR-206/miR-34a) + 0.056x(mir-124/miR-296)”. The area under the curve (AUC), sensitivity and specificity are displayed, and a cut-off
value with higher specificity was selected.

fied as potential mRNA targets of those miRNAs, were APS patients, a combination of the 10 miRNA ratios as a
quantified in the plasma of APS patients and HDs. As pre- panel was carried out by using a logistic regression on the
viously reported,20-23 APS patients showed significantly data set, as previously described.24 Thus, all miRNA-ratios
increased plasma levels of TF, PAI-1, MCP-1, VEGF-A and were integrated into a single model or equation, which
VEGFR-1 (Online Supplementary Figure S1). provided a single ‘score’ that allowed us to perform the
ROC analysis and establish the cut off for prediction. The
Circulating miRNA signature as potential biomarkers ROC curve for the 10 miRNA ratios signature revealed a
of disease in APS marked accuracy, evidenced by an AUC of 0.81. At the
It has been shown that the combination of miRNAs optimal cut-off value of 0.6, the sensitivity and specificity
improves their predictive potential to differentiate two of the combined miRNA panel for APS identification were
pathological conditions.14-19 Thus, to assess the potential of 78% and 80%, respectively (Figure 3B).
specific circulating miRNAs in APS patients as biomarkers
of disease features, reciprocal ratios of the miRNAs ana- Stability of miRNA expression profile over time in APS
lyzed were performed by using statistical tools. By this Plasma from 21 APS patients included in the study was
approach, we identified 10 miRNA ratios, integrated by evaluated again three months after the first blood sample
the 11 selected miRNAs, and differentially expressed in collection to analyze the stability of the circulating
plasma of APS patients in comparison with HDs, includ- miRNA profile. Results demonstrated that miRNA
ing miR-19b/miR-34a, miR-19b/miR-15a, miR19b/miR- expression in the second sample collection did not
124, miR-19b/miR-145, miR-20a/miR-145, miR-20a/miR- change in relation to the first analysis (Online
374a, miR-20a/miR-210, miR-20a/miR-133b, miR- Supplementary Figure S2A). Moreover, the levels of
206/miR-34a and miR-124/miR-296 (Figure 3A). To fur- miRNA ratios at baseline correlated significantly with
ther explore the efficiency of these biomarkers to identify the levels of these ratios three months later (Online

912 haematologica | 2018; 103(5)


MicroRNAs as antiphospholipid syndrome biomarkers

Figure 4. Antiphospholipid syndrome (APS) patients show a specific miRNA profile distinct from both non-autoimmune patients with previous thrombotic events
and aPL-negative systemic lupus erythematosus (SLE) patients. Twenty-three thrombotic non-antiphospholipid syndrome patients (non-APS) and 25 aPL-negative
SLE patients were included, and the circulating microRNA (miRNA or miR) signatures of APS were compared. One-way ANOVA was used for statistical comparisons.
A Bonferroni correction was applied for multiple testing. P<0.05 was considered statistically significant. Beeswarm plot of each differentially expressed miRNA ratio
is shown along with mean, Standard Deviation and P-value. n.s.: no significant statistical differences.

Supplementary Figure S2B). Thus, our data support the ences in miRNA signature, except for the ratio miR-
theory that there is a specific circulating miRNA signa- 20a/374 (Online Supplementary Figure S3).
ture in APS which remains stable over time.
Circulating miRNAs are associated with clinical
APS patients show a specific miRNA profile different features of APS and show potential as biomarkers for
from both non-autoimmune patients with previous the development of atherosclerosis
thrombotic events and aPL-negative SLE patients The levels of some circulating miRNA ratios that inte-
To assess the specificity of the miRNA signature found grate the signature in APS were associated with the ocur-
in APS patients, and in order to analyze whether the rence of fetal losses in these patients, including elevated
altered expression of the circulating miRNAs evaluated levels of miR-19b/miR-124 and miR-20a/miR-374, and
was linked to their thrombophilic status, an additional dis- reduced levels of miR-124/miR-296 (Figure 5A).
ease group including 23 patients with thrombosis in the Associations between miRNA ratios and the type of
absence of an associated autoimmune disease was evalu- thrombosis suffered by APS patients were also identified.
ated. In these patients, the ratios formed by the expression Thus, elevated levels of ratios miR-20a/miR-145 and miR-
levels of the 11 selected miRNAs were significantly differ- 20a/miR-374 were significantly associated with the ocur-
ent from those described in APS patients, except for the rence of arterial thrombosis in APS patients (Figure 5B).
ratios miR-19b/miR-15a and miR-19b/miR-145 which Furthermore, the ratios of miR-19b/miR-124 and miR-
exhibited non-significant differences (Figure 4). To evalu- 124/miR-296 were also found to be associated with the
ate if the altered expression of the miRNA signature was presence of a pathological CIMT in these patients (Figure
a sign of an autoimmune status, an additional disease 5C). To accurately evaluate their relevance as biomarkers
group, including 25 SLE patients negative for aPL, was of early atherosclerosis, we conducted combined ROC
analyzed. In this SLE cohort, the ratios produced by the analyses of these miRNA ratios. The combination of both
selected circulating miRNAs were significantly different circulating miRNA ratios as a panel showed an evident
from those found in APS patients, except for the ratios accuracy, with an AUC of 0.76 at a sensitivity of 67% and
miR-19b/miR-34a, miR-20a/miR-374a and miR-124/miR- specificity of 78% from a cut-off value of 0.41 (Figure 5D).
296 which exhibited non-significant differences (Figure 4).
Cluster analysis
Potential influence of standard therapy on the profile Hard clustering analysis in the APS cohort differentiated
of circulating miRNAs in APS 3 clusters representing different thrombotic risk profile
APS patients were classified into two groups based on groups. Clinical and laboratory parameters of each cluster
the treatment received, including 30 primary APS patients are resumed (Figure 6A). Briefly, cluster 1 (50% of the clus-
treated with antiplatelet agents and 62 primary APS tered cohort) was characterized by lower prevalence of
patients treated with anticoagulant drugs. The statistical cardiovascular risk factors and aPL multiple positivity.
comparison between patients treated with antiplatelet Conversely, cluster 1 shows a higher rate of venous
and anticoagulant agents showed no significant differ- thrombotic event when compared to the other clusters.

haematologica | 2018; 103(5) 913


C. Perez-Sánchez et al.

A B

C D

Figure 5. Circulating miRNAs are related to clinical features of antiphospholipid syndrome (APS) and show potential as biomarkers for the development of ather-
osclerosis. Association studies of altered circulating microRNA (miRNA or miR) ratios and the occurrence of previous fetal loss (A), the type of thrombosis suffered
(B) and the presence of a pathological carotid intima-media thickness (CIMT) (C). Beeswarm plot of each miR ratio is shown, along with mean, Standard Deviation,
and P-value. (D) A combination as a panel of the 2 miRNA ratios associated to the pathological CIMT was carried out by using logistic regression on the data set and
receiver operator characteristics (ROC) curve analyses were performed. ROC curve of miRNA panel and cut off were generated based on the predicted probability
(P) for each patient as a single score. The equation used was: “Combined miRNA-ratio panel [Logit(p)] = 0.599 – 0.133x(miR-19b/miR-124) + 0.007x(miR-124/miR-
296)”. The area under the curve (AUC), sensitivity and specificity are shown, and a cut-off value with higher specificity was selected.

Cluster 2 (17.6% of the clustered cohort) was character- showed significant positive correlations with the expres-
ized by a higher rate of cardiovascular risk factors, arterial sion levels of various miRNA ratios and with levels of TF,
thrombotic events, recurrences and a low prevalence of PAI-1, VEGF-A, VEGF-R1 and MCP-1 (Online
multiple aPL positivity. Cluster 3 (32.4% of the clustered Supplementary Table S3). Some of these correlations were
cohort) was represented by a higher rate of multiple aPL also found among various miRNA ratios in plasma of APS
positivity, arterial thrombotic events, and lower rate of patients.
cardiovascular risk factors. When evaluating different
miRNA ratio expression among clusters, we found a sta- Antiphospholipid antibodies modulate the expression
tistically significant difference between groups for the fol- of both the circulating miRNAs that integrate the
lowing miRNA ratios: miR-19b/miR-124 (P<0.001, signature in APS and their potential protein targets
ANOVA), miR-20a/miR-374 (P<0.05, ANOVA), miR- The expression of the 11 selected miRNAs was signifi-
20a/miR-210 (P<0.001, ANOVA) and miR-124/miR-296 cantly altered in the supernatant of HUVECs treated with
(P<0.05, ANOVA). miRNA ratio expression in the differ- aPL-IgG in relation to those treated with a non-immune-
ent clusters are summarized in Figure 6C. IgG (Figure 7A), except for the miR-124 and miR-206.
When comparing the aGAPSS values among the differ- Accordingly, this treatment promoted in HUVECs the
ent clusters, we found a significant difference (P=0.008, secretion of atherothrombotic proteins, such as TF, PAI-1
t-test) between cluster 1 [mean aGAPSS 5.38; and VEGF-R1 (Figure 7B), potential targets of the miRNAs
1.628±Standard Deviation (SD)] and Cluster 2 (mean analyzed. On the other hand, the expression levels of sev-
aGAPSS 8,67; 3.67±SD). Similarly, we found a significant eral miRNAs were deregulated in the supernatant of
difference (P<0.001, t-test) between cluster 1 and cluster 3 monocytes treated with aPL-IgG, including miR-19b, miR-
(mean aGAPSS 10.82; 2.316±SD). aGAPSS values stratify- 20a, miR-145, miR-210 and miR-296 (Figure 7C).
ing for clusters are represented in Figure 6B. Concomitantly, aPL-IgG treatment promoted in mono-
cytes an increase in the secretion of TF, PAI-1 and MCP-1
Circulating miRNAs correlate with clinical and (Figure 7D).
serological parameters in APS
The miRNA ratios that integrate the signature in APS
were linked with clinical parameters, such as ABI, pres- Discussion
ence of elevated titers of aPL, particularly aCL and anti-
β2GPI antibodies, and erythrocyte sedimentation rate The present study identifies, for the first time, a specific
(Online Supplementary Table S3). Correlation analyses with signature of circulating miRNAs in APS patients that
serological markers related to atherothrombosis further might serve as potential biomarkers of clinical features of

914 haematologica | 2018; 103(5)


MicroRNAs as antiphospholipid syndrome biomarkers

A B

Figure 6. Specific miRNA signatures might


identify subgroups of antiphospholipid syn-
drome (APS) patients showing different
thrombotic risk profiles: cluster analysis. (A)
Clinical and laboratory parameters of the 3
clusters. (B) Comparison of the adjusted
global anti-phospholipid syndrome score
(aGAPSS) values among the different clus-
ters. (C) Evaluation of different microRNA
(miR) ratios expression among clusters.
HTA: arterial hypertension; LA: lupus antico-
agulant; aCL: anti-cardiolipin IgG/IgM; anti-
β2GPI: anti-β2 glycoprotein 1 IgG/IgM.

this autoimmune disorder. Moreover, this signature could of these ratios allows the identification of a combination
represent a useful tool to typify and stratify patients based of expression profiles closer to reality in vivo in patients,
on their thrombotic status and cardiovascular risk profile where the interactions between miRNAs and their specific
(Online Supplementary Figure S3). potential targets never occur in a unique or individualized
Circulating miRNAs were firstly described in peripheral way. In fact, it is likely that, in some cases, various
blood as promising specific biomarkers for a wide range of miRNAs, whose concentrations are shifted in opposite
diseases, such as cancer and other inflammatory patholo- directions in a particular pathology, contribute together
gies.25,26 Thereafter, several studies revealed the altered and specifically to certain clinical profiles.
expression of numerous miRNAs in plasma, blood cells, The signatures of circulating miRNAs identified in APS
and tissues of systemic autoimmune conditions, such as patients integrated miRNAs previously described to be
RA and SLE, which were directly associated to disease altered in other autoimmune and CVD. Thus, miR-19b
activity, making them potential useful biomarkers for clin- and miR-20a have been shown to be essential modulators
ical features and follow up.9,26-29 However, to date, the spe- of TF expression in APS and SLE patients,8 so that reduced
cific profile of circulating miRNAs in APS patients has not expression of such miRNAs contributes to the overexpres-
been evaluated. In the present study, the profiling of sion of TF in monocytes, which is directly associated with
miRNAs by PCR-array in plasma of APS patients has the occurrence of thrombotic events in APS.21 On the
helped to identify a set of miRNAs differentially expressed other hand, miR-124, found altered in APS, SLE and RA
and collectively associated to clinical features of the dis- patients at both cellular and plasma levels, modulates the
ease, such as inflammatory response, reproductive system overexpression of MCP-1, a key chemokine directly
disease, and CVD, among others. Using logistic regres- involved in CVD associated to these autoimmune condi-
sion, we further developed a model that identified 10 tions.30-33 Likewise, miR-133b and miR-145 have been
miRNA ratios, differentially expressed, that showed great identified as the most promising biomarkers of the patho-
potential as biomarkers of disease of APS patients. genesis of CVD. Both miRNAs participate in the differen-
Recent studies support the evidence that an miRNAs tiation of vascular smooth muscle cells. In addition, miR-
signature has a higher diagnostic value than individual 133b regulates angiogenesis and endothelial function,
miRNAs.14-19 The use of ratios is a feasible approach that while miR-145 participates in the stabilization of athero-
overcomes the controversial question of normalizing plas- matous plaque.34 The miR-34a is highly expressed in
ma levels of miRNAs, given the lack of a reliable normal- endothelial cells, and elevated circulating levels of this
izer for circulating miRNAs. In addition, the establishment miRNA have been associated to myocardial infarction.35

haematologica | 2018; 103(5) 915


C. Perez-Sánchez et al.

A B

C D

Figure 7. Antiphospholipid antibodies modulate the expression of both the circulating miRNAs that integrate the signature in antiphospholipid syndrome (APS)
and their putative protein targets. Human umbilical vein endothelial cells (HUVECs) were treated with antiphospholipid antibodies and secreted selected microRNAs
(miRNAs) (A) and putative target protein (B) levels were determined in the supernatant. Monocytes were also treated with antiphospholipid antibodies and secreted
selected miRNAs (C) and putative target proteins (D) levels were evaluated in the supernatant of culture. Differences were analyzed by Student t-test. Values are the
means and Standard Error of Mean of 4 independent experiments performed in triplicate. P<0.05 was considered statistically significant. TF: tissue factor; PAI-1:
plasminogen activator inhibitor-1; VEGF-A: vascular endothelial growth factor A; VEGF-R1: VEGF-Receptor-1; MCP-1: monocyte chemotactic protein.

Moreover, the main target of miR-34a is VEGF-A, a key study were mainly treated with anticoagulant and/or
inflammatory protein involved in numerous cardiovascu- antiplatelet agents. All of them have been shown to influ-
lar and autoimmune pathologies, including APS.23,36 In the ence miRNAs expression, an epigenetic process that might
same way, miR-374 has been described as regulator of help to delineate the mechanisms underlying their
maintenance of vascular integrity.37 The remaining effects.9,41,42 Thus, we evaluated the potential effect of
miRNAs members of the signature, including miR-296, these treatments on the circulating miRNA expression
miR-210, miR-206 and miRNA-15, have been found profile. No significant differences were observed in our
altered in severe pre-eclampsia, one of the leading causes cohort of APS patients between those who received
of maternal mortality and neonatal morbidity world- antiplatelet and those treated with anticoagulant agents,
wide.38-40 Thus, all the processes regulated by these suggesting that the prothrombotic status induced by
miRNAs seem to orchestrate distinct aspects of APS effects of aPLs, and the consequently deregulated
pathogenesis. miRNAs, were not differentially modulated among these
To assess the specificity of the circulating miRNA signa- drugs.
ture in APS we evaluated the miRNA profile in an addi- In order to understand the clinical relevance of the
tional cohort of patients characterized by the presence of altered circulating miRNA signature, association and cor-
previous thrombotic events in the absence of an associat- relation studies were perfomed. Altered expression of var-
ed autoimmune disease. The miRNAs analysis revealed a ious miRNA ratios was associated with the presence of
differential pattern of expression between these two previous fetal losses. In line with these findings, several
cohorts. Those results substantiate previous studies that studies have shown that the misregulation of circulating
evidenced the presence of a distinct miRNA profile in placental miRNAs in maternal blood might lead to preg-
monocytes and neutrophils of thrombotic non-autoim- nancy complications, thus acting as non-invasive diagnos-
mune patients compared to APS patients.9 This could tic and prognostic biomarkers for pregnancy monitoring.42-
44
reflect a differential mode of regulation and activity of Association studies further established a significantly
miRNAs in thrombotic patients compared to APS patients, increased expression of 2 miRNA ratios in APS patients
on which the role of autoantibodies might be crucial. that had suffered arterial thrombosis in comparison with
Moreover, the analysis of a parallel autoimmune popula- those who experienced venous thrombotic events.
tion (SLE patients) negative for aPL, also identified an Interestingly, both miRNA ratios were integrated by the
miRNA signature distinct from that of APS, thus underly- miR-20a, previously reported to be the main regulator of
ing the potential role of aPLs as regulators of thrombosis- TF, whose expression levels have been found to be related
related miRNAs in APS, and pointing to the presence of a to the development of arterial thrombosis in the setting of
specific miRNA profile relative to the pathogenesis of APS.8,45 Finally, we identified 2 miRNA ratios as clinical rel-
each disease. evant biomarkers related to early atherosclerosis develop-
Antiphospholipid syndrome patients recruited in this ment in APS patients, which were integrated by the miR-

916 haematologica | 2018; 103(5)


MicroRNAs as antiphospholipid syndrome biomarkers

19b and miR-124, both of them critical players in the defined. In addition, since we did not perform a complete
expression of proteins related to inflammation and throm- plasma human microarray analysis, we cannot exclude the
bosis in APS and SLE.8,9 complementary role of other circulating miRNAs in the
Correlation studies revealed that the altered circulating physiopathology of APS.
miRNA signature in APS is linked to parameters related to Interestingly, our analysis supports a clinical role for the
increased risk of peripheral artery disease such as ABI. use of miRNA ratios when stratifying patients for their
Moreover, correlations between circulating miRNA levels thrombotic risk. While studying the miRNA expression
and numerous altered parameters related to inflammation profile has widened the understanding of APS pathogene-
and thrombosis were also identified. These correlations sis,9 its clinical utility is still a question of debate. Our data
support the relationship observed in the in silico study support the view that specific miRNA signatures could
between the selected miRNAs and potential target pro- identify subgroups of APS patients showing different clin-
teins involved in various clinical features of APS. The ical profiles (in terms of site of thrombosis and risk of
influence of the autoimmunity in the circulating profile of recurrences), potentially paving the way for their use as
miRNAs in APS was also revealed by the significant corre- useful biomarkers that will increase the specificity and
lation between high titers of aPL-IgG and the altered sensitivity of thrombotic risk assessment.
expression of several miRNAs integrating the signature. Taken together, our data suggest that differentially
These relationships further sustain those previously iden- expressed miRNAs in the plasma of APS patients, modu-
tified among the altered profile of miRNAs in APS and SLE lated at least partially by aPL-IgG antibodies, might have
at cellular level and the autoimmune and inflammatory the potential to serve as novel biomarkers of disease fea-
profile of both autoimmune conditions.9 Therefore, our tures and could help typify the atherothrombotic status of
data suggest that the altered plasma profile of miRNAs is patients, thus constituting a useful tool in the manage-
an important mechanisin that might contribute to the reg- ment of this disease.
ulation of the pro-atherothrombotic status of APS
patients, on which aPL seem to play a key role. Acknowledgments
Our in vitro studies further confirmed this hypothesis, We thank all patients for their participation in this study.
demonstrating that aPL-IgG antibodies promoted a signif-
icant deregulation in the expression levels of both the Funding
selected miRNAs and their potential protein targets in the This study was supported by grants from the Junta de
supernatant of cultured monocytes and HUVECs, the Andalucia (CTS-7940), the Instituto de Salud Carlos III (ref. n.
main drivers of the CVD in the setting of APS. These PI15/01333), Cofinanciado por el Fondo Europeo de Desarrollo
results also confirm and complement previous studies Regional de la Unión Europea 'Una manera de hacer Europa',
which showed the in vitro effects of aPL-IgG in the induc- Spain, and the Spanish Inflammatory and Rheumatic Diseases
tion of prothrombotic/inflammatory mediators3,31 and the Network (RIER), Instituto de Salud Carlos III
modulation of specific cellular miRNAs involved in their (RD16/0012/0015). CL-P was supported by a contract from the
modulation.8,9 Nevertheless, although our data show spe- Spanish Junta de Andalucía. YJ-G was supported by a contract
cific effects of aPL-IgG on the secretion of several circulat- from the University of Cordoba (Co-financing of the Research
ing microRNAs related to CVD, the contribution of other Plan of the University of Cordoba and the Operating Program of
components of the vascular and immune system to the the European Regional Development Funds -ERDF- for
altered profile of circulating miRNAs still has to be Andalusia).

immune regulatory functions. Clin Dev and extracellular communicators in cardio-


References Immunol. 2012;2012:9746-9748. vascular disease? Circ Res. 2012;110(3):
5. Huang Y, Shen XJ, Zou Q, Wang SP, Tang 483-495.
1. Young SP, Kapoor SR, Viant MR, et al. The SM, Zhang GZ. Biological functions of 11. Heegaard NHH, Carlsen AL, Skovgaard K,
impact of inflammation on metabolomic microRNAs: a review. J Physiol Biochem. Heegaard PMH. Circulating Extracellular
profiles in patients with arthritis. Arthritis 2011;67(1):129-139. microRNA in Systemic Autoimmunity.
Rheum. 2013;65(8):2015-2023. 6. Xiao C, Rajewsky K. MicroRNA control in EXS. 2015;106:171-195.
2. Palisi A, Grimaldi M, Sabatini P, et al. A the immune system: basic principles. Cell. 12. Sciascia S, Sanna G, Murru V, Roccatello D,
serum nuclear magnetic resonance-based 2009;136(1):26-36. Khamashta MA, Bertolaccini ML. GAPSS:
metabolomic signature of antiphospholipid 7. Alevizos I, Illei GG. MicroRNAs as bio- The global anti-phospholipid syndrome
syndrome. J Pharm Biomed Anal. markers in rheumatic diseases. Nat Rev score. Rheumatology (Oxford).
2017;133:90-95. Rheumatol. 2010;6(7):391-398. 2015;54(1):134-138.
3. Perez-Sanchez C, Barbarroja N, Messineo 8. Teruel R, Pérez-Sánchez C, Corral J, et al. 13. Castro-Villegas C, Pérez-Sánchez C,
S, et al. Gene profiling reveals specific Identification of miRNAs as potential mod- Escudero A, et al. Circulating miRNAs as
molecular pathways in the pathogenesis of ulators of tissue factor expression in potential biomarkers of therapy effective-
atherosclerosis and cardiovascular disease patients with systemic lupus erythemato- ness in rheumatoid arthritis patients treat-
in antiphospholipid syndrome, systemic sus and antiphospholipid syndrome. J ed with anti-TNFa. Arthritis Res Ther.
lupus erythematosus and antiphospholipid Thromb Haemost. 2011;9(10):1985-1992. 2015;17:49.
syndrome with lupus. Ann Rheum Dis. 9. Pérez-Sánchez C, Aguirre MA, Ruiz-Limón 14. Boeri M, Verri C, Conte D, et al. MicroRNA
2015;74(7):1441-1449. P, et al. Atherothrombosis-associated signatures in tissues and plasma predict
4. López-Pedrera C, Pérez-Sánchez C, microRNAs in Antiphospholipid syndrome develoment and prognosis of computed
Ramos-Casals M, Santos-Gonzalez M, and Systemic Lupus Erythematosus tomography detected lung cancer. Proc
Rodriguez-Ariza A, Cuadrado MJ. patients. Sci Rep. 2016;6:31375. Natl Acad Sci USA. 2011;108(9):3713-3718.
Cardiovascular risk in systemic autoim- 10. Creemers EE, Tijsen AJ, Pinto YM. 15. Hennessey PT, Sanford T, Choudhary A, et
mune diseases: epigenetic mechanisms of Circulating microRNAs: novel biomarkers al. Serum microRNA biomarkers for detec-

haematologica | 2018; 103(5) 917


C. Perez-Sánchez et al.

tion of non-samll cell lung cancer. PLoS Natl Acad Sci USA. 2008;105(30):10513- MicroRNA-34a regulates cardiac fibrosis
One. 2012;7(2):e32307. 10518. after myocardial infarction by targeting
16. Sheinerman KS, Tsivinsky VG, Abdullah L, 26. Chen X, Ba Y, Ma L, et al. Characterization Smad4. Expert Opin Ther Targets. 2014;
Crawford F, Umansky SR. Plasma of microRNAs in serum: a novel class of bio- 18(12):1355-1365.
microRNA biomarkers for detection of mild markers for diagnosis of cancer and other 36. Li N, Wang K, Li PF. MicroRNA-34 Family
cognitive impairment: biomarker validation diseases. Cell Res. 2008;18(10):997-1006. and Its Role in Cardiovascular Disease. Crit
study. Aging (Albany NY). 2013;5(12):925- 27. Dai Y, Huang YS, Tang M, et al. Microarray Rev Eukaryot Gene Expr. 2015;25(4):293-
938. analysis of microRNA expression in 297.
17. Matthaei H, Wylie D, Lloyd MB, et al. peripheral blood cells of systemic lupus 37. Licholai S, Blaż M, Kapelak B, Sanak M.
miRNA biomarkers in cyst fluid augment erythematosus patients. Lupus. 2007;16: Unbiased Profile of MicroRNA Expression
the diagnosis and management of pancreatic 939-946. in Ascending Aortic Aneurysm Tissue
cysts. Clin Cancer Res. 2012;18(17):4713- 28. Rodriguez A, Vigorito E, Clare S, et al. Appoints Molecular Pathways Contributing
4724. Requirement of bic/microRNA-155 for nor- to the Pathology. Ann Thorac Surg.
18. Fortunato O, Boeri M, Verri C, et al. mal immune function. Science. 2016;102(4):1245-1252
Assessment of circulating microRNAs in 2007;316(5824):608-611. 38. Choi SY, Yun J, Lee OJ, et al. MicroRNA
plasma of lung cancer patients. Molecules. 29. Wang J, Chen J, Sen S. MicroRNA as expression profiles in placenta with severe
2014;19(3):3038-3054. Biomarkers and Diagnostics. J Cell Physiol. preeclampsia using a PNA-based microarray.
19. Sharova E, Grassi A, Marcer A, et al. A circu- 2016;231(1):25-30. Placenta. 2013;34(9):799-804.
lating miRNA assay as a first-line test for 30. Nakamachi Y, Kawano S, Takenokuchi M. 39. Sheikh AM, Small HY, Currie G, Delles C.
prostate cancer screening. Br J Cancer. MicroRNA-124a is a key regulator of prolif- Systematic Review of Micro-RNA
2016;114(12):1362-1366. eration and monocyte chemoattractant pro- Expression in Pre-Eclampsia Identifies a
20. Mayburd AL, Martlínez A, Sackett D, et al. tein 1 secretion in fibroblast-like synovio- Number of Common Pathways Associated
Ingenuity network-assisted transcription cytes from patients with rheumatoid arthri- with the Disease. PLoS One. 2016;11(8):
profiling: Identification of a new pharmaco- tis. Arthritis Rheum. 2009;60(5):1294-1304. e0160808.
logic mechanism for MK886. Clin Cancer 31. Pérez-Sánchez C, Ruiz-Limón P, Aguirre 40. Akehurst C, Small HY, Sharafetdinova L, et
Res. 2006;12(6):1820-1827. MA, et al. Mitochondrial dysfunction in al. Differential expression of microRNA-206
21. Cuadrado MJ, López-Pedrera C, Khamashta antiphospholipid síndrome: implications in and its target genes in preeclampsia. J
MA, et al. Thrombosis in primary antiphos- the pathogenesis of the disease and effects Hypertens. 2015;33(10):2068-2074.
pholipid syndrome: a pivotal role for mono- of coenzyme Q10 treatment. Blood. 2012; 41. Chan ES, Cronstein BN. Methotrexate--how
cyte tissue factor expression. Arthritis 119(24):5859-5870. does it really work? Nat Rev Rheumatol.
Rheum. 1997;40(5):834-841. 32. Ruiz-Limón P, Barbarroja N, Pérez-Sánchez 2010;6(3):175-178.
22. López-Pedrera C, Aguirre MA, Buendía P, et C, et al. Atherosclerosis and cardiovascular 42. Tsochandaridis M, Nasca L, Toga C, Levy-
al. Differential expression of protease acti- disease in systemic lupus erythematosus: Mozziconacci A. Circulating microRNAs as
vated receptors in monocytes from patients effects of in vivo statin treatment. Ann clinical biomarkers in the predictions of
with primary Antiphospholipid syndrome. Rheum Dis. 2015;74(7):1450-1458. pregnancy complications. Biomed Res Int.
Arthritis Rheum. 2010; 62(3):869-877. 33. Barbarroja N, Pérez-Sanchez C, Ruiz-Limon 2015;2015:294954.
23. Cuadrado MJ, Buendía P, Velasco F, et al. P, et al. Anticyclic citrullinated protein anti- 43. Zhao Z, Moley KH, and Gronowski AM.
Vascular endothelial growth factor expres- bodies are implicated in the development of Diagnostic potential for miRNAs as bio-
sion in monocytes from patients with pri- cardiovascular disease in rheumatoid arthri- markers for pregnancy-specific diseases.
mary antiphospholipid syndrome. J Thromb tis. Arterioscler Thromb Vasc Biol. Clinical Biochemistry. 2013;46(10-11):953-
Haemost. 2006;4(11):2461-2469. 2014;34(12):2706-2716. 960.
24. Xiao B, Wang Y, Li W, et al. Plasma 34. Navickas R, Gal D, Laucevičius A, 44. Wu L, Zhou H, Lin H, et al. Circulating
microRNA signature as a noninvasive bio- Taparauskaitė A, Zdanytė M, Holvoet P. microRNAs are elevated in plasma from
marker for acute graft-versus-host disease. Identifying circulating microRNAs as bio- severe preeclamptic pregnancies.
Blood. 2013;122(19):3365-3375. markers of cardiovascular disease: a system- Reproduction. 2012;143(3):389-397.
25. Mitchell PS, Parkin RK, Kroh EM, et al. atic review. Cardiovasc Res. 45. Tatsumi K, Mackman N. Tissue Factor and
Circulating microRNAs as stable blood- 2016;111(4):322-337. Atherothrombosis. J Atheroscler Thromb.
based markers for cancer detection. Proc 35. Huang Y, Qi Y, Du JQ, Zhang DF. 2015;22(6):543-549

918 haematologica | 2018; 103(5)

Vous aimerez peut-être aussi