Vous êtes sur la page 1sur 4

correspondence

Detection of circulating tumour DNA in patients with


aggressive B-cell non-Hodgkin lymphoma

Current methods for detecting the presence of disease in Seventeen patients were enrolled on this study (Table I).
patients with diffuse large B cell lymphoma (DLBCL) or None of 13 patients biopsied had bone marrow involvement.
mediastinal large B-cell lymphoma (MLBCL) rely primarily At least one dominant tumour clonotype could be estab-
on imaging methods, which are associated with significant lished from the diagnostic biopsy for all 16 patients with suf-
cost and radiation exposure. Very few patients with DLBCL ficient amounts of amplifiable DNA. The diagnostic biopsy
have evidence of circulating disease using flow cytometric sample for one patient had <0.3 ng of DNA, and a dominant
assays (Mancuso et al, 2010). This has so far precluded the tumour clonotype was not identified. Among the 16 patients
development of minimal residual disease (MRD) assessment with an established tumour clonotype, circulating tumour
tools in those diseases, in contrast to tumours with a circu- DNA could be detected pre-treatment in plasma in 11 of the
lating component, where MRD assays are emerging as 16 (69%, 90% confidence interval [CI] 45–87%), in PBMCs
important methods (Ferrero et al, 2011). The availability of in eight of the 16 (50%, 90%CI 28–72%), and in either in 13
high-throughput sequencing techniques now provides an of the 16 (81%, 90%CI 58–95%) (Table I, Fig 1A, B). The
opportunity to probe for the presence of very small amounts three patients in whom no DNA could be detected at diag-
of circulating tumour genetic material in peripheral blood nosis all had stage I disease and had no evidence of disease
(PB). If sequencing-based methods can reliably detect circu- by positron emission tomography-computerized tomography
lating disease, they could eventually find a role in the treat- (PET-CT) after their diagnostic procedure at the time that
ment and monitoring of patients with those tumours. We the study sample was drawn. Therefore, tumour DNA was
present here a pilot study of a sequencing method designed detected in all 13 (100%) of patients with evidence of fluoro-
to examine whether tumour DNA is detectable in patients deoxyglucose-avid disease at the time of sample collection.
with newly diagnosed DLBCL/MLBCL, and whether it There was no apparent correlation between the level of circu-
becomes undetectable after therapy. lating tumour DNA and clinical characteristics including his-
Tumour samples obtained from PB samples and forma- tology, stage, burden of disease, or International Prognostic
lin-fixed paraffin-embedded (FFPE) or frozen tissue were Index (The International Non-Hodgkin’s Lymphoma Prog-
analysed using the Sequenta LymphoSIGHTTM method nostic Factors Project, 1993). There was also no association
(Sequenta Inc., South San Francisco, CA, USA), as previ- between the presence or level of circulating DNA and lactate
ously described (Faham et al, 2012a, 2012b). Briefly, geno- dehydrogenase elevation pre-treatment.
mic DNA was extracted from tumour cells, peripheral blood All patients were treated with chemo-immunotherapy; one
mononuclear cells (PBMCs) or plasma, amplified using patient with MLBCL also received radiotherapy. Seven
locus-specific primer sets for IGH and IGK rearrangements, patients from the DFCI cohort returned for collection of
and sequenced. A frequency >5% in the diagnostic tumour post-therapy samples; the remaining received their care out-
sample was considered to represent one of the tumour clo- side of DFCI and did not return for follow-up. At a median
notypes; the frequency of the tumour clonotype(s) in PB follow-up of 269 (range, 220–281) days from the beginning
was calculated relative to the total number of reads in the of therapy, none of the patients have relapsed. Among the
sample. seven evaluable patients, six (86%, 90%CI 48–99%) had no
We approached consecutive patients with newly diagnosed detectable circulating tumour DNA at the conclusion of
DLBCL or MLBCL who presented for care at Dana-Farber chemoimmunotherapy (Table I, Fig 1C).
Cancer Institute (DFCI). Independently, five patients with Using a novel high-throughput sequencing technique, we
DLBCL who had banked tumour and blood samples at the could determine the dominant tumour clonotype in 94% of
M.D. Anderson Cancer Center (MDACC) were enrolled on a 17 patients with DLBCL/MLBCL from routine diagnostic
similar study. Written informed consent was obtained from tumour samples, and in 100% of those with adequate
all patients. All patients had a sample of tumour from their amount of tumour material for analysis. Among those
diagnostic biopsy sent to Sequenta Inc. for analysis. We col- patients, 81% had detectable circulating tumour DNA in
lected a sample of whole blood before the first cycle of ther- plasma or PBMCs; the proportion was 100% among the
apy; when possible, we also collected a sample after the patients with evidence of disease by PET-CT scan after their
conclusion of therapy (only in the DFCI cohort). diagnostic surgical biopsy. Furthermore, the tumour DNA

ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 163, 123–144
124
Table I. Patient characteristics and tumour DNA results.

Clinical characteristics Tumour DNA measurements*


Correspondence

Dominant
tumour Post-
Center Bone Ki67 Treatment sequence treatment
patient Histology stage Age marrow IPI LDH (%) Treatment outcome identified Pre-treatment PB assay PB assay

DFCI 1 DLBCL IE (bowel) 43 Negative 0 Nl 50 RCHOP 9 6 CR Yes Negative† Negative


DFCI 2 DLBCL IIE (thyroid) 46 Negative 0 Nl 90 DA-REPOCH 9 6 IT chemo CR‡ Yes Positive in plasma and PBMCs Positive in
PBMCs only
DFCI 3 DLBCL IV (bowel) 36 Negative 2 Nl 30–40 RCHOP 9 6 CR Yes Positive in plasma only Negative
DFCI 4 MLBCL I 29 N/A 1 Hi 80–90 RCHOP 9 6 + IFRT PR by PET after Yes Positive in plasma only Negative
RCHOP§
DFCI 5 DLBCL IV (testis) 75 N/A 2 Nl 90 RCHOP 9 6 IT chemo CR (after three Yes Positive in plasma and PBMCs Negative
cycles)
DFCI 6 DLBCL I 66 Negative 1 Nl 80–90 RCHOP 9 6 CR (after three Yes Negative† N/A
cycles)
DFCI 7 DLBCL IE (bone) 31 N/A 0 Nl 80 RCHOP 9 6 + IFRT CR (after three Yes Positive in plasma and PBMCs N/A
cycles)
DFCI 8 DLBCL III 84 N/A 2 Nl 60–70 RCHOP 9 6 CR No N/A N/A
DFCI 9 DLBCL IV 72 Negative 2 Nl 80–90 RCHOP 9 6 No follow-up Yes Positive in PBMCs only N/A
DFCI 10 DLBCL II 53 Negative 0 Nl 60 RCHOP 9 6 CR Yes Positive in plasma and PBMCs Negative
DFCI 11 MLBCL II 61 Negative 2 Nl 40 RCHOP 9 6 CR Yes Positive in plasma only Negative
DFCI 12 DLBCL IE (spleen) 64 Negative 2 Nl >90 RCHOP 9 6 CR Yes Negative† N/A
MDACC 13 DLBCL III 52 Negative 1 Nl 10 RCHOP 9 6 CR Yes Positive in plasma and PBMCs N/A
MDACC 14 DLBCL IV (ovary) 31 Negative 2 Hi 90 RhCVAD 9 6 CR Yes Positive in plasma only N/A
MDACC 15 DLBCL IV (lung) 77 Negative 3 Hi 50 RCHOP 9 6 CR Yes Positive in PBMCs only N/A
MDACC 16 DLBCL IV (bone) 79 Negative 3 Hi 80 RCHOP 9 4 CR Yes Positive in plasma only N/A
MDACC 17 DLBCL III 68 Negative 3 Hi 80 RCHOP 9 6 CR Yes Positive in plasma and PBMCs N/A

IPI, International Prognostic Index (The International Non-Hodgkin’s Lymphoma Prognostic Factors Project, 1993); Nl, normal; Hi, High; DFCI, Dana-Farber Cancer Institute; MDACC, M.D.
Anderson Cancer Center; DLBCL, diffuse large B cell lymphoma; MLBCL, primary mediastinal B-cell lymphoma; N/A, not available; CR, complete remission; PR, partial remission; R, rituximab;
CHOP, cyclophosphamide, adriamycin, vincristine and prednisone; DA-REPOCH, dose-adjusted rituximab, etoposide, prednisone, vincristine, cyclophosphamide, adriamycin; hCVAD, hyperfraction-
ated cyclophosphamide, vincristine, adriamycin, dexamethasone; IFRT, involved field radiotherapy; IT, intrathecal; PBMCs, peripheral blood mononuclear cells.
*Measurements for all available samples consisted of separate plasma and PBMC assays. ‘Negative’ means that tumour sequences were detected in neither plasma nor PBMCs.
†No evidence of disease by positron emission tomography -PET/computerized tomography (PET-CT) after diagnostic biopsy/surgery at the time the pre-treatment sample was drawn.
‡Low level fluorodeoxyglucose-positive residual in thyroid with negative biopsy.
§This patient subsequently received involved field radiotherapy and achieved CR.

ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 163, 123–144
Correspondence

(A)

(B)

Fig 1. DLBCL/MLBCL tumour clone sequences


in peripheral blood of patients. (A). Tumour
sequences pre-treatment in peripheral blood (C)
mononuclear cells (PBMCs) for each patient.
The patient number corresponds to that in
Table I; Patient 8 had no established tumour
clonotype. The number of sequences is
reported per million PBMCs. Values shown are
the mean of the maximum calibrating receptor
clones for IGH DJ (circles), IGH VDJ
(squares), or IGK (triangles). (B). Tumour
sequences pre-treatment in plasma for each
patient. The number of sequences is reported
per millilitre of plasma. (C). Changes in
peripheral blood mononuclear cells (left panel)
and plasma (right panel) tumour clone
sequences pre- and post-treatment. Values are
shown for each of the seven patients with both
pre- and post-treatment samples.

cleared in all but one patient after curative-intent chemo-


Acknowledgments
immunotherapy. This sequencing method may therefore rep-
resent a reliable dynamic tumour assessment. Our study is P.A. is supported by an ASCO/Conquer Cancer Foundation
based on a small number of cases, and leaves many questions Career Development Award. The blood and tissue samples
still unanswered, which await the maturation of ongoing lar- from the University of Texas M. D. Anderson Cancer Center
ger studies with longer follow-up. Nonetheless, our prelimin- were obtained from the Lymphoma Tissue Bank supported
ary findings recall the results obtained with minimal residual by the Lymphoma SPORE (CA136411) to A.Y. and S.S.N.,
disease (MRD) assays in diseases where MRD detection has the Cancer Center Support Grant CA16672, and the Fredrick
important clinical implications, and in which this sequencing B. Hagemeister Research Fund.
method has already shown promising results (Logan et al,
2011, 2012; Faham et al, 2012a,b; Harris et al, 2012). They
Author contributions
raise the possibility that MRD may become an evaluable and
actionable result in DLBCL and MLBCL, which are not tradi- P.A.and Y.O. designed and performed the research, analysed
tionally thought to have a circulating component. the data, and wrote the paper. D.S.N. designed the research,

ª 2013 John Wiley & Sons Ltd 125


British Journal of Haematology, 2013, 163, 123–144
Correspondence

analysed the data, and edited the paper. M.F. collected data, Eric D. Jacobsen1
analysed the data, and edited the paper. C.C. analysed the data Matthew S. Davids1
and edited the paper. M.K. analysed the data and edited the Caron Jacobson1
paper. L.W. collected data, analysed the data, and edited the David C. Fisher1
paper. S.B. collected data and edited the paper. A.S.L. collected Jennifer R. Brown1
data and edited the paper. E.D.J. collected data and edited the Nathan H. Fowler2
paper. M.D. collected data, and edited the paper. C.J. collected M. Alma Rodriguez2
data, analysed the data, and edited the paper. D.C.F. collected Michael J. Wallace5
data and edited the paper. J.R.B. collected data, and edited the Sattva S. Neelapu2
paper. N.H.F. collected data, and edited the paper. M.A.R. col- Scott Rodig6
lected data, and edited the paper. S.N. collected data, and edited Anas Younes2
the paper. S.R. collected data, and edited the paper. A.Y. Arnold S. Freedman1
1
designed the research, collected data and edited the paper. A.S.F. Department of Medical Oncology, Dana-Farber Cancer Institute,
designed the research, collected data and edited the paper. Boston, MA , 2Department of Lymphoma and Myeloma, The University
of Texas, M.D. Anderson Cancer Center, Houston, TX , 3Department
of Biostatistics and Computational Biology, Dana-Farber Cancer
Conflict of interest
Institute, Boston, MA , 4Sequenta Inc, South San Francisco, CA ,
5
M.F., C.C., M.K. and L.W. are employees of Sequenta Inc. The Department of Diagnostic Radiology, The University of Texas, M.D.
authors have no other relevant conflicts of interest to disclose. Anderson Cancer Center, Houston, TX, and 6Department of Pathology,
Brigham and Women’s Hospital, Boston, MA, USA
Philippe Armand1*
E-mail: parmand@partners.org*
Yasuhiro Oki2*
Donna S. Neuberg3
*These two authors contributed equally to this work.
Malek Faham4
Craig Cummings4
Keywords: lymphomas, minimal residual disease, sequences
Mark Klinger4
Li Weng4
First published online 25 June 2013
Sangeetha Bhattar1
doi: 10.1111/bjh.12439
Ann S. LaCasce1

References Harris, M., Faham, M., Fang, L., Moorhead, M., M., Willis, T., Zhang, B., Waqar, A., Zehnder, J.L.
Faham, M., Brody, J., Kohrt, H.E., Logan, A.C., Zheng, J., Neuberg, D.S., Sallan, S.E., Silverman, & Miklos, D.B. (2012) Minimal Residual Disease
Advani, R.H., Czerwinski, D.K., Weng, W., L.B. & Stevenson, K.E. (2012) Comparison of Quantification in Acute Lymphoblastic Leukemia
Negrin, R., Carlton, B. & Levy, R. (2012a) Deep Minimal Residual Disease Detection Using Using High-Throughput Sequencing of Multiple
B and T Cell Repertoire Sequencing to Evaluate High-Throughput Sequencing and Allele-Specific Immunoreceptor Genes Predicts Relapse and Sur-
Minimal Residual Disease and T Cell Responses Oligonucleotide PCR Methods in Pediatric vival After Allogeneic Hematopoietic Cell Trans-
in a Therapeutic Vaccine Trial for Mantle Cell B-Lineage Acute Lymphoblastic Leukemia. Blood plantation. Blood (ASH Annual Meeting
Lymphoma. Blood (ASH Annual Meeting (ASH Annual Meeting Abstracts), 120, 2532. Abstracts), 120, 2512.
Abstracts), 120, 582. Logan, A.C., Faham, M., Carlton, V., Zheng, J., Mancuso, P., Calleri, A., Antoniotti, P., Quarna, J.,
Faham, M., Zheng, J., Moorhead, M., Carlton, V.E., Moorhead, M., Willis, T., Jones, C.D., Zhang, Pruneri, G. & Bertolini, F. (2010) If it is in the
Stow, P., Coustan-Smith, E., Pui, C.H. & M.B., Waqar, A.N., Zehnder, J.L. & Miklos, D.B. marrow, is it also in the blood? An analysis of
Campana, D. (2012b) Deep-sequencing approach (2011) High-Throughput Immunoglobulin Gene 1,000 paired samples from patients with B-cell
for minimal residual disease detection in acute Sequencing Quantifies Minimal Residual Disease non-Hodgkin lymphoma. BMC Cancer, 10, 644.
lymphoblastic leukemia. Blood, 120, 5173–5180. in CLL with 10e-6 Sensitivity and Strongly Pre- The International Non-Hodgkin’s Lymphoma Prog-
Ferrero, S., Drandi, D., Mantoan, B., Ghione, P., dicts Relapse After Allogeneic Hematopoietic nostic Factors Project. (1993) A predictive model
Omede, P. & Ladetto, M. (2011) Minimal resid- Cell Transplantation. Blood (ASH Annual Meet- for aggressive non-Hodgkin’s lymphoma. The
ual disease detection in lymphoma and multiple ing Abstracts), 118, 2542. International Non-Hodgkin’s Lymphoma Prog-
myeloma: impact on therapeutic paradigms. Logan, A.C., Vashi, N., Faham, M., Carlton, nostic Factors Project. The New England Journal
Hematological Oncology, 29, 167–176. V., Buno, I., Zheng, J., Moorhead, M., Klinger, of Medicine, 329, 987–994.

126 ª 2013 John Wiley & Sons Ltd


British Journal of Haematology, 2013, 163, 123–144

Vous aimerez peut-être aussi