Vous êtes sur la page 1sur 9

The

Oncologist ®

Lymphoma
EBV-Associated Lymphoproliferative Disorders: Classification
and Treatment

ANTONINO CARBONE,a ANNUNZIATA GLOGHINI,a GIAMPIETRO DOTTIb


a
Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, Milano, Italy; bCenter for Cell and

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


Gene Therapy, Baylor College of Medicine, The Methodist Hospital, Houston, Texas, USA

Key Words. EBV • EBV-associated lymphomas • Post-transplant–associated lymphoproliferative disorders •


HIV-associated lymphoproliferative disorders • EBV cell– based immunotherapy

Disclosure: No potential conflicts of interest were reported by the authors, planners, reviewers, or staff managers of this article.

LEARNING OBJECTIVES
After completing this course, the reader will be able to:
1. Assess patients with EBV-associated lymphoproliferative disorders.
2. Describe the pathogenesis of the lymphoproliferative disorders linked to EBV infection.
3. Evaluate EBV cell– based immunotherapy for use in patients with EBV-associated lymphoproliferative disorders.
CME This article is available for continuing medical education credit at CME.TheOncologist.com.

ABSTRACT
Since its discovery as the first human tumor virus, encodes a series of products interacting with or exhib-
Epstein-Barr virus (EBV) has been implicated in the iting homology to a wide variety of antiapoptotic mol-
development of a wide range of B-cell lymphoprolif- ecules, cytokines, and signal transducers, hence
erative disorders, including Burkitt’s lymphoma, promoting EBV infection, immortalization, and
classic Hodgkin’s lymphoma, and lymphomas arising transformation. However, the exact mechanism by
in immunocompromised individuals (post-transplant which EBV promotes oncogenesis is an area of active
and HIV-associated lymphoproliferative disorders). debate. The focus of this review is on the pathology,
T-cell lymphoproliferative disorders that have been diagnosis, classification, and pathogenesis of EBV-
reported to be EBV associated include a subset of pe- associated lymphomas. Recent advances in EBV cell–
ripheral T-cell lymphomas, angioimmunoblastic T- based immunotherapy, which is beginning to show
cell lymphoma, extranodal nasal type natural killer/ promise in the treatment of EBV-related disorders,
T-cell lymphoma, and other rare histotypes. EBV are discussed. The Oncologist 2008;13:577–585

Correspondence: A. Carbone, M.D., Department of Pathology, Fondazione IRCCS Istituto Nazionale Tumori, via Venezian 1, Milano
I-20133, Italy. Telephone: 39-02-2390-2876; Fax: 39-02-2390-2877; e-mail: antonino.carbone@istitutotumori.mi.it Received Febru-
ary 13, 2008; accepted for publication March 18, 2008. ©AlphaMed Press 1083-7159/2008/$30.00/0 doi: 10.1634/theoncologist.2008-
0036

The Oncologist 2008;13:577–585 www.TheOncologist.com


578 EBV-Associated Lymphomas

INTRODUCTION
Epstein-Barr virus (EBV) is a member of the herpesvirus Table 1. Latent EBV-encoded genes
family. As with other herpesviruses, EBV is an enveloped Latency
EBV-encoded genes Location type
virus that contains a DNA core surrounded by an icosahe-
dral nucleocapsid and a tegument. Family members include EBNA-1 Nucleus I, II, III
herpes simplex I and II and varicella zoster virus (␣-herpes- EBNA-2 Nucleus III
virus subfamily), cytomegalovirus and human herpesvirus EBNA-3 Nucleus III
(HHV)-6 and HHV-7 (␤-herpesvirus subfamily), and LMP-1 Membrane II, III
HHV-8 and EBV (␥-herpesvirus subfamily) [1]. LMP-2 Membrane II, III
Human tumors have been attributed to both HHV-8 EBER-1 and EBER-2 Nucleus I, II, III
(Kaposi’s sarcoma, primary effusion lymphoma [PEL], and Abbreviations: EBV, Epstein–Barr virus; EBNA,
Epstein–Barr nuclear antigen; LMP, latent membrane
multicentric Castleman’s disease) and EBV (Burkitt’s lym- protein; EBER, Epstein–Barr RNA.
phoma, nasopharyngeal carcinoma, and Hodgkin’s and

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


non-Hodgkin’s lymphomas) [2–5].
In this review we discuss only the EBV-associated lym- Table 2. EBV-associated lymphoproliferative disorders
phoproliferative disorders, dividing them into B-cell and EBV-associated B-cell lymphoproliferative disorders
T/natural killer (NK)-cell lymphomas. The focus is on the Burkitt’s lymphoma
pathology, diagnosis, and classification, pathogenesis, and Classic Hodgkin’s lymphoma
treatment of EBV-associated lymphomas. Post-transplant lymphoproliferative disorders
HIV-associated lymphoproliferative disorders
EBV PRODUCTS AND PATTERNS OF EBV Primary central nervous system lymphoma
GENE EXPRESSION Diffuse large B-cell lymphoma, immunoblastic
The EBV genome encodes a series of products interacting HHV-8–positive primary effusion lymphoma and its
solid variant
with or exhibiting homology to a wide variety of antiapo-
Plasmablastic lymphoma
ptotic molecules, cytokines, and signal transducers, hence
Other histotypes (rare)a
promoting EBV infection, immortalization, and transfor-
aOther histotypes include: lymphomatoid granulomatosis,
mation [6 – 8]. pyothorax-associated lymphoma, senile EBV-associated
Based on patterns of expression of the EBV genome, B-cell lymphoproliferative disorders.
three types of latent gene expression have been described: Abbreviations: EBV, Epstein–Barr virus; HHV-8, human
herpesvirus 8.
latency I, II, and III (Table 1). During latency I, Epstein–
Barr nuclear antigen 1 (EBNA-1) and the two small non-
coding Epstein–Barr RNAs (EBERs) are expressed [9].
EBV-ASSOCIATED B-CELL
EBV gene expression in latency II is usually limited to
LYMPHOPROLIFERATIVE DISORDERS
EBNA-1, the EBERs, latent membrane protein (LMP)-1,
Since its discovery as the first human tumor virus, EBV has
and LMP-2A and LMP-2B [10]. Latency III usually in-
been implicated in the development of a wide range of B-
volves the unrestricted expression of all EBNAs, EBERs,
cell lymphoproliferative disorders (Table 2).
and LMPs [10]. Latency I is generally associated with the
EBV-related Burkitt’s lymphoma [9], latency II has been Burkitt’s Lymphoma
associated with classic Hodgkin’s lymphoma (cHL) and T- Burkitt’s lymphoma is a highly proliferative B-cell tumor
cell non-Hodgkin’s lymphoma (NHL) [11], and latency III that includes three variants: endemic (affecting children in
occurs mainly in immunocompromised individuals suffer- equatorial Africa and New Guinea), sporadic (affecting
ing from post-transplant lymphoproliferative disorders children and young adults throughout the world) (Fig. 1),
(PTLDs) and HIV-associated lymphoproliferative disor- and immunodeficiency related (primarily in association
ders and in lymphoblastoid cell lines [12]. with HIV infection). EBV has been detected in virtually all
Furthermore, EBV encodes the following important cases of the endemic variant, 15%–20% of cases of the spo-
proteins that show sequence and functional homology to di- radic variant, and 30%– 40% of cases of the immunodefi-
verse human proteins: BCRF-1 and interleukin (IL)-10, ciency-related variant [1]. In all variants, irrespective of
BHRF-1 and BCL-2, BARF-1 and intracellular adhesion EBV status, constitutive activation of the c-myc oncogene
molecule 1 [13]. through its translocation into one of the immunoglobulin
Carbone, Gloghini, Dotti 579

transformed B cells. The detection of destructive somatic


mutations in the rearranged immunoglobulin genes of RS
cells of cHL indicates that they originate from preapoptotic
GC B cells [24]. In RS cells, constitutive nuclear factor
(NF)-␬B activation, which is essential for RS cell survival,
seems to result from various mechanisms. Several recep-
tors, such as CD30, CD40, receptor activator of nuclear fac-
tor ␬B (RANK), and RANK ligand, which activate the
classic NF-␬B pathway, are expressed by RS cells, and li-
gands for these receptors are frequently expressed on acti-
vated CD4⫹ T cells and other bystander cells surrounding
RS cells, including eosinophils, neutrophils, and B-cell
subsets. In EBV-positive cHL cases, the LMP-1 gene also
contributes to NF-␬B activation because it mimics acti-

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


Figure 1. Burkitt’s lymphoma. EBV is detected in almost all vated CD40 receptors [25–27].
tumor cells. (Inset): Tumor consists of a homogeneous prolif-
eration of medium-sized cells displaying a cohesive pattern.
In HIV/AIDS persons with severe immunosuppression,
Tumor cells have round nuclei, multiple centrally located nu- the acquisition of an antiapoptotic phenotype by RS cells is
cleoli, and limited cytoplasm. (EBER in situ hybridization, he- not a result of CD40 –CD40L interactions, which are lack-
matoxylin counterstain, original magnification ⫻200; inset, ing, but rather is induced by EBV-encoded LMP-1, which is
hematoxylin-eosin, original magnification ⫻400.)
functionally homologous to activated CD40. Thus, EBV is
Abbreviations: EBER, Epstein–Barr RNA; EBV, Epstein–
Barr virus. thought to play a pivotal role in the pathogenesis of lym-
phoma in patients with immunosuppression [28].

loci is clearly the key factor in the oncogenesis of Burkitt’s EBV-Associated Lymphomas in
lymphoma [1]. The detection of somatic hypermutations in Immunocompromised Individuals
the V region of the immunoglobulin genes and the pheno- There exist several distinct classes of EBV-associated lym-
type of the Burkitt’s lymphoma cells indicate a germinal phoproliferative disorders in immunocompromised indi-
center (GC) cell origin of the lymphoma [14]. The tumors viduals. First, there is a disorder resulting from an inherited
isolated from nonendemic Burkitt’s lymphoma patients are immunodeficiency known as X-linked lymphoproliferative
usually from different stages of B-cell development than disorder. Second, there are lymphomas associated with
those isolated from patients with endemic Burkitt’s lym- immunosuppressive drugs given to transplant recipients.
phoma [15, 16]. Finally, there are AIDS-related lymphoproliferative dis-
Most EBV-positive cases exhibit a highly restrictive orders. The most common gene-expression pattern in these
pattern of expression of latent encoded proteins, only ex- disorders is latency III.
pressing EBNA-1 and the EBERs (latency I) [1]. However,
it was recently reported that some cases, in addition to PTLDs
EBNA-1 and the EBERs, express EBNA-3A, EBNA-3B, Since the original report in 1969 [29, 30], it has been well
EBNA-3C, and EBNA leader protein but still lack EBNA-2 established that there is a higher incidence of lymphopro-
and the latent membrane proteins [17]. EBNA-1 plays a liferative disorders in transplant recipients of both a solid
crucial role in the maintenance and replication of the viral organ and bone marrow. According to the World Health Or-
genome, but its oncogenic potential is highly controversial ganization (WHO) classification [31], PTLDs may be clas-
[1, 18]. Conversely, as the EBERs are believed to possess sified into: (a) early lesions, generally represented by EBV-
antiapoptotic activity, it has been postulated that they may driven polyclonal lymphoproliferations and (b) true
play an essential role in the oncogenesis of Burkitt’s lym- monoclonal diseases, including polymorphic PTLD and
phoma [19]. monomorphic PTLD; the latter is further distinguished into
Burkitt’s lymphoma/Burkitt’s-like lymphoma, diffuse
cHL large B-cell lymphoma (DLBCL), and cHL.
Not all subtypes of cHL harbor EBV to the same degree Early-onset PTLDs are mainly regarded as EBV-driven
[20 –22]. There are also data that suggest that the incidence lymphoproliferations that are frequently, though not al-
of EBV-positive cHL is age related [23]. ways, polyclonal or oligoclonal, whereas most late-onset
Hodgkin’s Reed-Sternberg (RS) cells of cHL represent PTLDs are true monoclonal lymphoid malignancies that are

www.TheOncologist.com
580 EBV-Associated Lymphomas

not necessarily associated with EBV infection. Correlative


studies of the morphologic and molecular features of
PTLDs have contributed to the recognition of specific dis-
ease categories and have provided prognostic indicators for
these disorders [32]. Among monoclonal B-cell PTLD, der-
ivation from GC-related B cells occurs independently of the
type of organ transplanted, the interval between transplant
and lymphoma, the histology, and the site of origin of the
lymphoma.
Oncogenic viruses known to be involved in PTLD
pathogenesis include EBV and HHV-8. Both EBV and
HHV-8 act predominantly through direct mechanisms, that
is, the virus is able to directly infect the tumor clone and
exerts a transforming effect upon B cells. Viral infection in

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


PTLD exploits several strategies to ensure persistent infec- Figure 2. HIV-associated diffuse large B-cell lymphoma
with immunoblastic features. Most large tumor cells express
tion, namely, prevention of death of infected cells, enhance- EBV infection, the signal is nuclear. (Inset): Tumor cells con-
ment of their proliferation to maintain the infected tain abundant cytoplasm and round, oval or ovoid nuclei com-
reservoir, and evasion of the immune system [33–35]. Sev- monly containing prominent nucleoli. (EBER in situ
eral lines of evidence suggest that EBV infection has a ma- hybridization, hematoxylin counterstain, original magnifica-
tion ⫻400; inset, hematoxylin-eosin, original magnification
jor pathogenetic role in PTLDs: (a) EBV infects ⬃60%–
⫻400.)
80% PTLD patients, including 100% of early-onset PTLD Abbreviations: EBER, Epstein–Barr RNA; EBV, Epstein–
patients [6]; (b) in many cases of monomorphic PTLD, Barr virus.
EBV infection is monoclonal, consistent with the hypothe-
sis that the virus has been present in the tumor progenitor
cell since the early phases of clonal expansion; (c) a de- clude two rare entities, namely, PEL and plasmablastic
crease in EBV-specific cytotoxic T lymphocytes (CTLs) lymphoma of the oral cavity [38, 39]. (c) Lymphomas also
and an increase in the EBV viral load are strongly associ- occurring in other immunodeficiency states.
ated with PTLD development [36]; and (d) treatment of EBV-associated HIV-NHL includes primary central
PTLDs with autologous EBV-specific CTLs results in viral nervous system lymphoma, systemic lymphomas having a
load control and tumor size reduction. DLBCL immunoblastic morphology (Fig. 2), HHV-8 –
positive PEL and its solid variant with and without serous
HIV-Associated Lymphoproliferative Disorders effusions (Fig. 3), and plasmablastic lymphoma [7, 40 – 46]
HIV-associated lymphoproliferative disorders are a het- (Table 2).
erogeneous group of diseases that arise in the presence of
HIV-associated immunosuppression, a state that permits
the unchecked proliferation of EBV-infected lympho-
EBV-ASSOCIATED T/NK-CELL
cytes. Traditionally, these aggressive disorders include LYMPHOPROLIFERATIVE DISORDERS
both central nervous system and systemic lymphomas. EBV is known primarily for its ability to infect B cells, but
PEL also occurs and often involves EBV in addition to it can also infect other cells. Several types of non-B-cell
HHV-8. NHL are associated with EBV (Table 3). T-cell lymphopro-
The categories of HIV-associated NHL (HIV-NHL) in- liferative disorders that have been reported to be EBV as-
cluded in the latest WHO proposal [37] are grouped as fol- sociated include a subset of peripheral T-cell lymphomas,
lows. (a) Lymphomas also occurring in immunocompetent angioimmunoblastic T-cell lymphoma (AILT), extranodal
patients. The vast majority of these HIV-NHLs belong to nasal type NK/T-cell lymphoma, enteropathy-type T-cell
three high-grade B-cell lymphomas: Burkitt’s lymphoma, lymphoma, ␥␦ T-cell lymphomas (hepatosplenic and non-
DLBCL with centroblastic features, and DLBCL with im- hepatosplenic), T-cell lymphoproliferative disorders after
munoblastic features. According to the site of involvement, chronic EBV infection, EBV-associated cutaneous T-cell
the present spectrum of HIV-NHL includes extranodal/ lymphoproliferative disorders (especially in Asia), and ag-
nodal lymphomas and primary central nervous system gressive NK-cell leukemia/lymphoma [8]. This section fo-
lymphomas. (b) Unusual lymphomas occurring more cuses on the two types in which EBV has been most directly
specifically in HIV-positive patients. These lymphomas in- implicated: AILT and nasal T/NK-cell lymphoma.
Carbone, Gloghini, Dotti 581

and anti–smooth muscle antibodies. The clinical behavior


is very aggressive, the response to therapy is scarce, and the
long-term outcome is dismal. The differential diagnosis
with other PTCLs, and in particular with unspecified PTCL,
is puzzling and mainly based on different features of the re-
active components (i.e., prominent vascular structures and
abundant follicular dendritic cells) and phenotype (CD10
and BCL-6 expression) [50]. Notably, no unique markers
can reliably discriminate between these two entities. Re-
cently, chemokine (C-X-C motif) ligand 13 was proposed
as a possible candidate to distinguish the two diseases [51].
The molecular pathogenesis of AILT, as in general for
all peripheral T-cell neoplasms, is poorly understood. The
karyotype is often characterized by complex abnormalities,

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


Figure 3. HHV-8-associated extracavitary solid lymphoma but specific alterations have not been identified, and only
without serous effusion. Large tumor cells express EBV infec-
tion as detected by EBER in situ hybridization. Tumor cells are
few reports focused on the gene-expression profile of nodal
pleomorphic and of heterogeneous size. (EBER in situ hybrid- PTCLs [51, 52]. In one study, an AILT molecular signature
ization, hematoxylin counterstain, original magnification was identified together with a molecular link between AILT
⫻400.) and T follicular helper lymphocytes [52].
Abbreviations: EBER, Epstein–Barr RNA; EBV, Epstein–
AILT is a lymphoma in which expanding B-cell clones
Barr virus; HHV-8, human herpesvirus 8.
are often present beside the T-cell clones. EBV infection is
seen mainly in the B lymphocytes and B immunoblasts, al-
Table 3. EBV-associated lymphoproliferative disorders though the virus also occurs in rare neoplastic and non-
EBV-associated T/NK-cell lymphoproliferative neoplastic T cells. The presence of EBV in only a
disorders subpopulation of cells suggests that EBV infection is sec-
Peripheral T-cell lymphoma, unspecified ondary to malignancy or that the viral genome has been lost
Angioimmunoblastic T-cell lymphoma from the malignant cell [6].
Extranodal nasal T/NK-cell lymphoma
Other histotypes (rare)a Nasal T/NK-Cell Lymphoma
aOther histotypes include hepatosplenic T-cell lymphoma, Nasal T/NK-cell lymphoma cells exhibit several unique ge-
nonhepatosplenic ␥␦ T-cell lymphomas, enteropathy-type notypic and phenotypic features. These features include an
T-cell lymphoma. absence of T-cell antigens, the expression of the NK cell
Abbreviations: EBV, Epstein–Barr virus; NK, natural
killer. marker CD56, and the absence of T-cell receptor gene re-
arrangement [15]. Clinically, these tumors occur in the na-
sal and upper aerodigestive area. EBV is consistently
Peripheral T-Cell Lymphomas associated with these lymphomas, regardless of geograph-
Peripheral T-cell lymphomas (PTCLs) represent 10%–15% ical location [53, 54] (Fig. 4).
of all lymphoid neoplasms [47]. They are a heterogeneous
group of tumors that in the Revised European-American TREATMENT
Lymphoma/WHO classification are subdivided into speci- This section focuses on the treatment of patients with EBV-
fied and unspecified forms [47, 48]. associated lymphomas in which EBV cell– based immuno-
therapy has been most directly applied.
AILT
AILT is a PTCL characterized by systemic disease, a poly- EBV Cell–Based Immunotherapy
morphous infiltrate primarily involving lymph nodes, and
prominent proliferation of high endothelial venules and fol- Treatment of EBV-Related PTLDs
licular dendritic cells [49]. AILT is the second most com- Because the great majority of PTLDs occurring in patients
mon PTCL subtype, accounting for 15%–20% of cases receiving a solid organ transplant (SOT) or allogeneic stem
[49]. Laboratory findings include polyclonal hyper-␥- cell transplant are a result of EBV infection/reactivation
globulinemia, circulating immunocomplexes, cold aggluti- that follows the impairment of the immune system, modu-
nins with hemolytic anemia, positive rheumatoid factor, lation of the immunosuppressive treatment is recom-

www.TheOncologist.com
582 EBV-Associated Lymphomas

by a new increase in the EBV viral load and eventually re-


lapse of the PTLD [61].
Chemotherapy still remains the only option for many
patients with aggressive PTLDs. The response rate and
overall survival time were widely variable on retrospective
analysis, and few prospective studies have been reported so
far [62– 64]. The best time and the best chemotherapy
schedule for these patients remain controversial. Low-dose
chemotherapy has been prospectively used in order to re-
duce the morbidity and mortality associated with standard
protocols. Although this approach significantly reduces
toxicities, it is also associated with a high relapse rate, sug-
gesting that more conventional chemotherapy regimens are
indicated in patients with aggressive PTLD [65]. CHOP-

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


Figure 4. Nasal T/NK-cell lymphoma. EBV-infected tumor like regimens are currently recommended by the European
cells display an angiocentric pattern of growth. The figure
shows several vascular structures of heterogeneous size.
guidelines at least for renal transplantation [66].
(EBER in situ hybridization, hematoxylin counterstain, origi- Is there a role for cellular immunotherapy in patients
nal magnification ⫻400.) with EBV-related PTLD? Adoptive transfer of donor-de-
Abbreviations: EBER, Epstein–Barr RNA; EBV, Epstein– rived EBV-specific CTLs (EBV-CTLs) has been successful
Barr virus; NK, natural killer.
when used as prophylaxis and treatment of EBV-related
PTLDs in the context of allogeneic stem cell transplantation
[67]. Analogously, adoptive transfer of EBV-CTLs may
mended as a first-line approach in patients with PTLDs
help to restore the balance of the immune compartment in
[55]. This approach induces tumor regression in 25%–50%
patients receiving a SOT, while avoiding the risk for graft
of patients [55]. Importantly, modulation of immunosup-
rejection that is frequently associated with the withdrawal
pression is currently applied by many transplant groups in
of immunosuppression.
patients with a high EBV-DNA viral load in order to pre-
Several groups recently reported their preliminary ex-
vent the occurrence of a PTLD [56].
periences with adoptive EBV-CTL transfer in SOT recipi-
The management of patients with PTLDs who fail the
ents as pre-emptive treatment in cases of high EBV DNA
reduction in immunosuppression is complex and remains
viral load or as treatment in the presence of clinical evi-
controversial. Chemotherapy and monoclonal antibodies
dence of a PTLD [68, 69]. Overall, these studies indicate
are the therapeutic options most frequently offered to these that CTL infusions are well tolerated. No graft toxicity was
patients. The use of rituximab has significantly changed the reported. Although EBV DNA did not decrease uniformly
therapeutic approach to PTLDs. The administration of the in all patients, in several studies EBV-CTLs prevented the
antibody is generally well tolerated and rapidly induces de- development of PTLDs, and in small series tumor regres-
pletion of mature B lymphocytes in the peripheral blood, sion after CTL therapy was also reported.
thus reducing the compartment of EBV-infected cells, with The main limitations to the extensive application of
an associated normalization of the viral load [57]. In the CTL therapies in these patients remain the cost and the time
case of PTLDs occurring in SOT patients, very enthusiastic required for EBV-CTL line generation. PTLDs can be very
results were reported in small uncontrolled studies, with a aggressive and rapidly fatal, while the generation of autol-
complete remission rate in the range of 30%–70% [58, 59]. ogous EBV-CTL lines requires several weeks. The use of
However, more recent phase II studies including larger co- partly HLA-matched EBV-CTLs seems a promising alter-
horts of patients have shown an overall median survival native to the generation of autologous EBV-CTLs [70]. In
time of 15 months, suggesting that rituximab should be in- our first experience, we generated EBV-CTL lines for pa-
tegrated with chemotherapy in patients with poor prognos- tients with a high EBV DNA viral load in the peripheral
tic factors, such as a high tumor burden, an EBV-negative blood (⬎4,000 DNA copies). However, because the EBV
PTLD, and PTLD occurring late after transplantation [60]. DNA viral load is not always predictive of PTLD occur-
In our own experience, the duration of remission in SOT rence in SOT recipients, we are currently generating EBV-
patients with EBV-related PTLD treated with rituximab can CTL lines only for patients who have received previous
be relatively short, because the recovery of B cells in the treatment for a PTLD. Because the relapse rate remains
presence of impaired T-cell immunity is often accompanied high in these patients, we are proposing the generation and
Carbone, Gloghini, Dotti 583

infusion of EBV-CTLs as a salvage treatment, thus reduc- function is still partially impaired by the immunosuppres-
ing the cost of manufacturing and decreasing the risk– sive drugs [73]. We and others are currently evaluating
benefit ratio for this procedure. strategies to genetically modify EBV-CTLs to make them
resistant to the effects of immunosuppressive drugs.
Treatment of EBV-Associated cHL A more robust in vivo expansion of adoptively trans-
Modern radiotherapy and/or chemotherapy regimens have ferred CTLs may help to increase the antitumor effects.
dramatically improved the cure rate of patients with cHL This concept was proven in previous clinical trials in mel-
[71]. However, despite the identification of clinical prog- anoma patients in which adoptive T-cell transfer followed
nostic factors and the optimal use of primary and secondary the administration of nonmyeloablative chemotherapy reg-
treatments, cHL remains fatal for ⬎15% of patients [71]. imens to create a lymphodepleted environment that would
Therefore, new therapeutic agents are required for primary favor the expansion of CTLs [74]. The tumor microenvi-
refractory patients and biological strategies are also desir- ronment may be particularly hostile for antitumor immu-
able to maintain remission in high-risk patients after con- nity. Indeed, many tumor cells, such as HL tumor cells,
ventional treatment. In addition, biological treatments may produce factors, including transforming growth factor ␤,

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


reduce many of the serious long-term side effects correlated thymus and activation-regulated chemokine, IL-10 and Fas
with radiation and chemotherapy [71]. ligand, that significantly impair the function or survival of
Therapy with monoclonal antibodies, and in particular antitumor CTLs. Genetic modification of CTLs to over-
with anti-CD30 monoclonal antibodies, has been used in come tumor escape mechanisms and increase their survival
patients with cHL. Although the great majority of patients in the tumor microenvironment may help in arming CTLs
enrolled in these studies were heavily resistant to previous and increasing their antitumor effects [75]. Finally, the ca-
therapies, clinical responses were minimal and not compa- pacity of tumor cells to downregulate tumor-associated an-
rable with the response rate obtained with rituximab in B- tigens needs to be carefully evaluated. Modern technology
cell– derived lymphomas [72]. allows the genetic modification of CTLs to expand their an-
The association between EBV and a subset of cHL and tigen specificity. In particular, gene transfer of chimeric an-
the expression of the latent proteins LMP-1 and LMP-2 by tigen receptors targeting crucial molecules expressed on the
tumor cells constitute the rationale to assess the feasibility cell surface of cHL cells, such as CD30, may improve their
of EBV-CTL adoptive transfer for patients with EBV-re- antitumor effects. Phase I clinical studies will help in clar-
lated cHL. We have conducted several phase I clinical trials ifying the role of these mechanisms to improve the treat-
in patients with cHL using both polyclonal EBV-CTLs and ment of these malignancies.
CTLs enriched in precursors targeting LMP-2 [73]. CTL in-
fusions were well tolerated, with objective and sustained ACKNOWLEDGMENTS
clinical responses in several patients. This work was supported in part by a Grant from the Istituto
Superiore Sanità, VI Programma Nazionale di Ricerche
sull’AIDS, Rome, Italy.
EBV-CTL THERAPIES AND FUTURE DIRECTIONS
The authors thank Maria Morelli for help in the prepa-
These studies demonstrate both the feasibility and efficacy
ration of the review and Gianni Roncato for photographic
of CTL therapies in patients with PTLDs after SOT and in
assistance.
patients with EBV-associated malignancies like cHL.
However, other important factors need to be considered to AUTHOR CONTRIBUTIONS
improve the clinical benefit of these approaches. Conception/design: Antonino Carbone
Financial support: Antonino Carbone
SOT patients require continuous immunosuppression to Manuscript writing: Antonino Carbone, Annunziata Gloghini, Giampietro
prevent graft rejection. Although adoptive transfer of EBV- Dotti
Final approval of manuscript: Antonino Carbone, Annunziata Gloghini,
CTLs may help in restoring immunocompetence, CTL Giampietro Dotti

REFERENCES tumours: An update for the attention of the working pathologist. J Clin
Pathol 2007;60:1358 –1364.
1 Young LS, Rickinson AB. Epstein-Barr virus: 40 years on. Nat Rev Cancer
2004;4:757–768. 4 Du MQ, Bacon CM, Isaacson PG. Kaposi sarcoma-associated herpesvirus/
2 Elgui de Oliveira D. DNA viruses in human cancer: An integrated overview human herpesvirus 8 and lymphoproliferative disorders. J Clin Pathol
on fundamental mechanisms of viral carcinogenesis. Cancer Lett 2007;247: 2007;60:1350 –1357.
182–196. 5 Chen YB, Rahemtullah A, Hochberg E. Primary effusion lymphoma. The
3 Delecluse HJ, Feederle R, O’Sullivan B et al. Epstein Barr virus-associated Oncologist 2007;12:569 –576.

www.TheOncologist.com
584 EBV-Associated Lymphomas

6 Thompson MP, Kurzrock R. Epstein-Barr virus and cancer. Clin Cancer tigen on Reed-Sternberg cells and Hodgkin’s disease cell lines. Blood
Res 2004;10:803– 821. 1995;85:780 –789.
7 Dolcetti R, Guidoboni M, Gloghini A et al. EBV-associated tumors: Patho- 28 Carbone A, Gloghini A, Larocca LM et al. Human immunodeficiency vi-
genetic insights for improved disease monitoring and treatment. Curr Can- rus-associated Hodgkin’s disease derives from post-germinal center B
cer Ther Rev 2005;1:27– 44. cells. Blood 1999;93:2319 –2326.
8 Rezk SA, Weiss LM. Epstein-Barr virus-associated lymphoproliferative 29 McKhann CF. Primary malignancy in patients undergoing immunosup-
disorders. Hum Pathol 2007;38:1293–1304. pression for renal transplantation. Transplantation 1969;8:209 –212.
9 Sbih-Lammali F, Djennaoui D, Belaoui H et al. Transcriptional expression 30 Penn I, Hammond W, Brettschneider L et al. Malignant lymphomas in
of Epstein-Barr virus genes and proto-oncogenes in north African nasopha- transplantation patients. Transplant Proc 1969;1:106 –112.
ryngeal carcinoma. J Med Virol 1996;49:7–14.
31 Borish B, Raphael M, Swerdlow SH et al. Lymphoproliferative diseases
10 Liebowitz D, Kieff E. Epstein-Barr virus. In: Roizman B, Whitley RJ, associated with primary immune disorders. In Jaffe ES, Harris NL, Stein H
Lopez C, eds. The Human Herpesvirus. Raven Press: New York, 1993: et al., eds. World Health Organization Classification of Tumours, Pathol-
107–172. ogy and Genetics of Tumours of Haematopoietic and Lymphoid Tissues.
11 Cesarman E, Mesri EA. Virus associated lymphomas. Curr Opin Oncol Lyon, France: IARC Press, 2001:257–259.
1999;11:322–332. 32 Chadburn A, Chen JM, Hsu DT et al. The morphologic and molecular ge-

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


12 Niedobitek G, Young LS, Herbst H. Epstein-Barr virus infection and the netic categories of posttransplantation lymphoproliferative disorders are
pathogenesis of malignant lymphomas. Cancer Surv 1997;30:143–162. clinically relevant. Cancer 1998;82:1978 –1987.
13 Hayes DP, Brink AA, Vervoot MB et al. Expression of Epstein-Barr virus 33 Cohen JI. Benign and malignant Epstein-Barr virus-associated B-cell lym-
(EBV) transcripts encoding homologues to important human proteins in di- phoproliferative diseases. Semin Hematol 2003;40:116 –123.
verse EBV associated diseases. Mol Pathol 1999;52:97–103. 34 Hsieh WS, Lemas MV, Ambinder RF. The biology of Epstein- Barr virus in
14 Küppers R. B cells under influence: Transformation of B cells by Epstein- post-transplant lymphoproliferative disease. Transplant Infect Dis 1999;1:
Barr virus. Nat Rev Immunol 2003;3:801– 812. 204 –212.
15 Wensing B, Farrell PJ. Regulation of cell growth and death by Epstein-Barr 35 Tanner JE, Alfieri C. The Epstein-Barr virus and post-transplant lympho-
virus. Microbes Infect 2000;2:77– 84. proliferative disease: Interplay of immunosuppression, EBV, and the im-
16 Okano M. Epstein-Barr virus infection and its role in the expanding spec- mune system in disease pathogenesis. Transplant Infect Dis 2001;3:60 – 69.
trum of human diseases. Acta Paediatr 1998;87:11–18. 36 Davis JE, Sherritt MA, Bharadwaj M et al. Determining virological, sero-
17 Kelly GL, Milner AE, Baldwin GS et al. Three restricted forms of Epstein- logical and immunological parameters of EBV infection in the develop-
Barr virus latency counteracting apoptosis in c-myc– expressing Burkitt ment of PTLD. Int Immunol 2004;16:983–989.
lymphoma cells. Proc Natl Acad Sci U S A 2006;103:14935–14940. 37 Raphael M, Borisch B, Jaffe ES. Lymphomas associated with infection by
18 Wilson JB, Bell JL, Levine AJ. Expression of Epstein-Barr virus nuclear the human immune deficiency virus (HIV). In: Jaffe ES, Harris NL, Stein H
antigen-1 induces B cell neoplasia in transgenic mice. EMBO J 1996;15: et al., eds. World Health Organization Classification of Tumours, Pathol-
3117–3126. ogy and Genetics of Tumours of Haematopoietic and Lymphoid Tissues.
Lyon, France: IARC Press, 2001:260 –263.
19 Kitagawa N, Goto M, Kurozumi K et al. Epstein-Barr virus– encoded
poly(A)(-) RNA supports Burkitt’s lymphoma growth through interleu- 38 Delecluse HJ, Anagnostopoulos I, Dallenbach F et al. Plasmablastic lym-
kin-10 induction. EMBO J 2000;19:6742– 6750. phomas of the oral cavity: A new entity associated with the human immu-
nodeficiency virus infection. Blood 1997;89:1413–1420.
20 Chapman A, Rickinson A. Epstein-Barr virus in Hodgkin’s disease. Ann
Oncol 1998;9(suppl 5):S5–S16. 39 Carbone A, Gaidano G, Gloghini A et al. AIDS-related plasmablastic lym-
phomas of the oral cavity and jaws: A diagnostic dilemma. Ann Otol Rhinol
21 Gledhill S, Gallagher A, Jones DB et al. Viral involvement in Hodgkin’s
Laryngol 1999;108:95–99.
disease: Detection of clonal type A Epstein-Barr virus genomes in tumor
samples. Br J Cancer 1991;64:227–232. 40 Carbone A, Gloghini A. AIDS-related lymphomas: From pathogenesis to
pathology. Br J Haematol 2005;130:662– 670.
22 Boyle M, Vasak E, Tschuchnigg M et al. Subtypes of Epstein-Barr virus
(EBV) in Hodgkin’s disease: Association between B-type EBV and immu- 41 Carbone A, Gloghini A. HHV-8-associated lymphoma: State-of-the-art re-
nocompromise. Blood 1993;81:468 – 474. view. Acta Haematol 2007;117:129 –131.

23 Armstrong AA, Alexander FE, Cartwright R et al. Epstein-Barr virus and 42 Cesarman E, Chang Y, Moore PS et al. Kaposi’s sarcoma-associated herpes-
Hodgkin’s disease: Further evidence for the three disease hypothesis. Leu- virus-like DNA sequences in AIDS-related body-cavity-based lymphomas.
kemia 1998;12:1272–1276. N Engl J Med 1995;332:1186–1191.
24 Kanzler H, Küppers R, Hansmann ML et al. Hodgkin and Reed-Sternberg 43 Carbone A, Gloghini A, Vaccher E et al. Kaposi’s sarcoma-associated her-
cells in Hodgkin’s disease represent the outgrowth of a dominant tumor pesvirus DNA sequences in AIDS-related and AIDS-unrelated lymphoma-
clone derived from (crippled) germinal center B cells. J Exp Med 1996;184: tous effusions. Br J Haematol 1996;94:533–543.
1495–1505. 44 Nador RG, Cesarman E, Chadburn A et al. Primary effusion lymphoma: A
25 Carbone A, Gloghini A, Gruss HJ et al. CD40 ligand is constitutively ex- distinct clinicopathologic entity associated with the Kaposi’s sarcoma-
pressed in a subset of T cell lymphomas and on the microenvironmental associated herpes virus. Blood 1996;88:645– 656.
reactive T cells of follicular lymphomas and Hodgkin’s disease. Am J 45 Gaidano G, Capello D, Cilia AM et al. Genetic characterization of HHV-
Pathol 1995;147:912–922. 8/KSHV-positive primary effusion lymphoma reveals frequent mutations
26 Re D, Küppers R, Diehl V. Molecular pathogenesis of Hodgkin’s lym- of BCL6: Implications for disease pathogenesis and histogenesis. Genes
phoma. J Clin Oncol 2005;23:6379 – 6386. Chromosomes Cancer 1999;24:16 –23.
27 Carbone A, Gloghini A, Gattei V et al. Expression of functional CD40 an- 46 Fan W, Bubman D, Chadburn A et al. Distinct subsets of primary effusion
Carbone, Gloghini, Dotti 585

lymphoma can be identified based on their cellular gene expression profile in B-cell post-transplantation lymphoproliferative disorders: Results of a
and viral association. J Virol 2005;79:1244 –1251. prospective multicenter phase 2 study. Blood 2006;107:3053–3057.
47 Jaffe ES, Ralfkiaer E. Mature T-cell and NK-cell neoplasms. In: Jaffe ES, 61 Savoldo B, Rooney CM, Quiros-Tejeira RE et al. Cellular immunity to Ep-
Harris NL, Stein H et al., eds. World Health Organization Classification of stein-Barr virus in liver transplant recipients treated with rituximab for
Tumours, Pathology and Genetics of Tumours of Haematopoietic and post-transplant lymphoproliferative disease. Am J Transplant 2005;5:566 –
Lymphoid Tissue. Lyon, France: IARC Press, 2001:189 –235. 572.
48 Harris NL, Jaffe ES, Stein H et al. A revised European-American classifi- 62 Dotti G, Fiocchi R, Motta T et al. Lymphomas occurring late after solid-
cation of lymphoid neoplasms: A proposal from the International Lym- organ transplantation: Influence of treatment on the clinical outcome.
phoma Study Group. Blood 1994;84:1361–1392. Transplantation 2002;74:1095–1102.

49 Went P, Agostinelli C, Gallamini A et al. Marker expression in peripheral 63 Gross TG, Bucuvalas JC, Park JR et al. Low-dose chemotherapy for Ep-
T-cell lymphoma: A proposed clinical-pathologic prognostic score. J Clin stein-Barr virus-positive post-transplantation lymphoproliferative disease
Oncol 2006;24:2472–2479. in children after solid organ transplantation. J Clin Oncol 2005;23:6481–
6488.
50 Ballester B, Ramuz O, Gisselbrecht C et al. Gene expression profiling iden-
tifies molecular subgroups among nodal peripheral T-cell lymphomas. On- 64 Choquet S, Trappe R, Leblond V et al. CHOP-21 for the treatment of post-
cogene 2006;25:1560 –1570. transplant lymphoproliferative disorders (PTLD) following solid organ
transplantation. Haematologica 2007;92:273–274.

Downloaded from http://theoncologist.alphamedpress.org/ by guest on April 29, 2017


51 Dupuis J, Boye K, Martin N et al. Expression of CXCL13 by neoplastic
65 EBPG Expert Group on Renal Transplantation. European best practice
cells in angioimmunoblastic T-cell lymphoma (AITL): A new diagnostic
guidelines for renal transplantation. Section IV: Long-term management of
marker providing evidence that AITL derives from follicular helper T cells.
the transplant recipient. Nephrol Dial Transplant 2002;17(suppl 4):1– 67.
Am J Surg Pathol 2006;30:490 – 494.
66 Rooney CM, Smith CA, Ng CY et al. Infusion of cytotoxic T cells for the
52 de Leval L, Rickman DS, Thielen C et al. The gene expression profile of
prevention and treatment of Epstein-Barr virus-induced lymphoma in allo-
nodal peripheral T-cell lymphoma demonstrates a molecular link between
geneic transplant recipients. Blood 1998;92:1549 –1555.
angioimmunoblastic T-cell lymphoma (AITL) and follicular helper T
(TFH) cells. Blood 2007;109:4952– 4963. 67 Khanna R, Bell S, Sherritt M et al. Activation and adoptive transfer of Ep-
stein-Barr virus-specific cytotoxic T cells in solid organ transplant patients
53 Baumforth KR, Young LS, Flavell KJ et al. The Epstein-Barr virus and its
with posttransplant lymphoproliferative disease. Proc Natl Acad Sci U S A
association with human cancers. Mol Pathol 1999;52:307–322.
1999;96:10391–10396.
54 International Agency for Research on Cancer. Epstein-Barr virus and Ka- 68 Comoli P, Labirio M, Basso S et al. Infusion of autologous Epstein-Barr
posi’s sarcoma herpesvirus/human herpesvirus 8. IARC Monographs on virus (EBV)-specific cytotoxic T cells for prevention of EBV-related lym-
the Evaluation of Carcinogenic Risks to Humans, Volume 70. Lyon, phoproliferative disorder in solid organ transplant recipients with evidence
France: IARC Press, 1997:45–52. of active virus replication. Blood 2002;99:2592–2598.
55 Paya CV, Fung JJ, Nalesnik MA et al. Epstein-Barr virus-induced post- 69 Savoldo B, Goss JA, Hammer MM et al. Treatment of solid organ trans-
transplant lymphoproliferative disorders. ASTS/ASTP EBV-PTLD Task plant recipients with autologous Epstein Barr virus-specific cytotoxic T
Force and The Mayo Clinic Organized International Consensus Develop- lymphocytes (CTLs). Blood 2006;108:2942–2949.
ment Meeting. Transplantation 1999;68:1517–1525.
70 Haque T, Wilkie GM, Taylor C et al. Treatment of Epstein-Barr-virus-
56 Lee TC, Savoldo B, Rooney CM et al. Quantitative EBV viral loads and positive post-transplantation lymphoproliferative disease with partly HLA-
immunosuppression alterations can decrease PTLD incidence in pediatric matched allogeneic cytotoxic T cells. Lancet 2002;360:436 – 442.
liver transplant recipients. Am J Transplant 2005;5:2222–2228.
71 Yung L, Linch D. Hodgkin’s lymphoma. Lancet 2003;361:943–951.
57 Kuehnle I, Huls MH, Liu Z et al. CD20 monoclonal antibody (rituximab) 72 Ansell SM, Horwitz SM, Engert A et al. Phase I/II study of an anti-CD30
for therapy of Epstein-Barr virus lymphoma after hemopoietic stem-cell monoclonal antibody (MDX-060) in Hodgkin’s lymphoma and anaplastic
transplantation. Blood 2000;95:1502–1505. large-cell lymphoma. J Clin Oncol 2007;25:2764 –2769.
58 Milpied N, Vasseur B, Parquet N et al. Humanized anti-CD20 monoclonal 73 Roskrow MA, Suzuki N, Gan Y et al. Epstein-Barr virus (EBV)-specific
antibody (rituximab) in post transplant B-lymphoproliferative disorder: A cytotoxic T lymphocytes for the treatment of patients with EBV-positive
retrospective analysis on 32 patients. Ann Oncol 2000;11(suppl 1):113– relapsed Hodgkin’s disease. Blood 1998;91:2925–2934.
116.
74 Straathof KC, Bollard CM, Rooney CM et al. Immunotherapy for Epstein-
59 Dotti G, Rambaldi A, Fiocchi R et al. Anti-CD20 antibody (rituximab) ad- Barr virus-associated cancers in children. The Oncologist 2003;8:83–98.
ministration in patients with late occurring lymphomas after solid organ 75 Dotti G, Savoldo B, Pule M et al. Human cytotoxic T lymphocytes with
transplant. Haematologica 2001;86:618 – 623. reduced sensitivity to Fas-induced apoptosis. Blood 2005;105:4677-
60 Choquet S, Leblond V, Herbrecht R et al. Efficacy and safety of rituximab 4684.

www.TheOncologist.com

Vous aimerez peut-être aussi