Among the most common HIV-associated lymphomas are Burkitt lymphoma (BL) and diffuse large B-cell lymphoma (DLBCL) with immunoblastic-plasmacytoid differentiation (also involving the central nervous system). Lymphomas occurring specifically in HIV-positive patients include primary effusion lymphoma (PEL) and its solid variants, plasmablastic lymphoma of the oral cavity type and large B-cell lymphoma arising in Kaposi sarcoma herpesvirus (KSHV)–associated multicentric Castleman disease. These lymphomas together with BL and DLBCL with immunoblastic-plasmacytoid differentiation frequently carry EBV infection and display a phenotype related to plasma cells. EBV infection occurs at different rates in different lymphoma types, whereas KSHV is specifically associated with PEL, which usually occurs in the setting of profound immunosuppression. The current knowledge about HIV-associated lymphomas can be summarized in the following key points: (1) lymphomas specifically occurring in patients with HIV infection are closely linked to other viral diseases; (2) AIDS lymphomas fall in a spectrum of B-cell differentiation where those associated with EBV or KSHV commonly exhibit plasmablastic differentiation; and (3) prognosis for patients with lymphomas and concomitant HIV infection could be improved using better combined chemotherapy protocols in-corporating anticancer treatments and antiretroviral drugs.

Infectious agents, mainly viruses, are among the few known causes of cancer and contribute to a variety of malignancies worldwide. The agents considered here, termed Epstein-Barr virus (EBV) and Kaposi sarcoma herpesvirus (KSHV–human herpesvirus 8 [HHV8]), are members of the gamma-herpesvirus subfamily.1 

Since its discovery as the first human tumor virus, EBV has been implicated in the development of a wide range of B-cell lymphoproliferative disorders, including Burkitt lymphoma (BL), nasopharyngeal carcinoma, and Hodgkin and non-Hodgkin lymphomas (NHLs). KSHV, one of the most recently discovered human tumor viruses and the cause of Kaposi sarcoma (KS),2  also plays a role in the pathogenesis of primary effusion lymphoma [PEL], and multicentric Castleman disease [MCD]).3-6  Intriguingly, EBV and KSHV have been shown to associate with distinct lymphoproliferative diseases occurring most often in persons with HIV infection/AIDS7,8  or in association with other immunodeficiency conditions, such as iatrogenic immunodeficiency following solid organ transplantation.9,10 

HIV-associated lymphoproliferative disorders are a heterogeneous group of diseases that arise in the presence of HIV-associated immunosuppression, a state that permits the unchecked proliferation of EBV- and KSHV-infected lymphocytes. Traditionally, these aggressive disorders mainly include both central nervous system and systemic lymphomas,11  whereas lymphomas specifically occurring in the setting of HIV infection include PEL and its solid variant, plasmablastic lymphoma (PBL) of the oral cavity type and large B-cell lymphoma arising in KSHV-associated MCD.12-18  Thus, HIV-related lymphomas are closely linked to EBV infection of the tumor clone or are associated with KSHV. PEL and its variants often involve EBV in addition to KSHV.19 

We review here the current knowledge on these gamma-herpesvirus–associated lymphomas in the setting of HIV infection. The focus will be on pathology, diagnosis and classification, pathogenesis, and treatment of these lymphomas specifically occurring in HIV-induced immunodeficiency.

EBV-associated lymphomas

EBV has been implicated in the development of a wide range of B-cell lymphoproliferative disorders, including BL, classic Hodgkin lymphoma (HL), and lymphomas arising in immunocompromised individuals (posttransplantation and HIV-associated lymphoproliferative disorders; Figure 1).1,2  It is also associated with B-cell lymphomas in association with congenital immunodeficiencies, such as X-linked lymphoproliferative syndrome (XLP). T-cell lymphoproliferative disorders that have been reported to be EBV associated include a subset of peripheral T-cell lymphomas, angioimmunoblastic T-cell lymphoma, extranodal nasal type natural killer/T-cell lymphoma, and other rare histotypes.1,2 

Figure 1

Relationship of HIV-associated lymphomas with EBV and KSHV/HHV8-associated lymphoproliferative disorders. *Other histotypes include lymphomatoid granulomatosis, DLBCL associated with chronic inflammation, EBV-positive DLBCL of the elderly. BL indicates Burkitt lymphoma; DLBCL, diffuse large B-cell lymphoma; EBV, Epstein-Barr virus; HHV8, human herpesvirus 8; KSHV, Kaposi sarcoma herpesvirus; MCD, multicentric Castleman disease; and PEL, primary effusion lymphoma.

Figure 1

Relationship of HIV-associated lymphomas with EBV and KSHV/HHV8-associated lymphoproliferative disorders. *Other histotypes include lymphomatoid granulomatosis, DLBCL associated with chronic inflammation, EBV-positive DLBCL of the elderly. BL indicates Burkitt lymphoma; DLBCL, diffuse large B-cell lymphoma; EBV, Epstein-Barr virus; HHV8, human herpesvirus 8; KSHV, Kaposi sarcoma herpesvirus; MCD, multicentric Castleman disease; and PEL, primary effusion lymphoma.

Close modal

EBV-associated lymphomas in AIDS include BL, diffuse large B-cell lymphoma (DLBCL) with immunoblastic (IB) morphology, primary central nervous system lymphoma (PCNSL), KSHV+ PEL and its solid variant, and PBL of the oral cavity type.11-16,18  However, the percentage of cases within each histotypes with EBV viral infection is variable, ranging from 60% to 100%.

PEL, KSHV-associated solid lymphomas, and other KSHV-associated lymphomas

PEL and its extracavitary variant are by definition KSHV-associated lymphomas.11-14  KSHV-associated extracavitary lymphomas have been reported preceding the development of an effusion lymphoma or following resolution of PEL. Recently, the spectrum of KSHV-associated lymphoproliferative diseases in the HIV setting has been expanded by the identification of cases of KSHV-associated extracavitary lymphomas without serous effusions (Figure 1).13,14 

In addition to PEL, KSHV is associated with another rare neoplastic lymphoproliferative disorder, namely large B-cell lymphoma arising in KSHV-associated MCD. KSHV-infected B cells in MCD have a pre–plasma cell phenotype and plasmacytic/plasmablastic morphology.17  A new KSHV-associated lymphoproliferative disorder has recently been described in HIV-seronegative persons.5  This disease, called germinotropic lymphoproliferative disorder, is characterized by plasmablasts that are coinfected by KSHV and EBV and preferentially involve the germinal centers of lymph nodes.

Relationship of HIV-associated lymphomas with EBV and KSHV infection

Figure 2 summarizes the relationship of HIV-associated lymphomas with EBV and KSHV infection. BL with plasmacytoid differentiation is often HIV associated and closely linked to EBV infection. When carrying EBV infection, the HIV-DLBCL-IB frequently displays a plasmacytoid differentiation. HIV-DLBCL-IB displays a phenotype mostly related to plasma cells while retaining features of the immunoblastic stage of B-cell development, suggesting that the normal cellular counterpart of AIDS-DLBCL-IB is a cell that might be defined as plasmablastic.20,21  PEL and its solid variant are universally linked to KSHV. Most HIV-associated PBLs of the oral cavity type are linked to EBV infection. Therefore, it follows that the spectrum of lymphomas occurring in HIV-infected patients is more clearly characterized thanks to the frequent plasma cell phenotype and the intriguing link to infection by gamma-herpesviruses. Although these lymphomas can occur in the general population, their presence in HIV-seropositive persons is predominant or almost exclusive.

Figure 2

The spectrum of HIV-associated lymphomas. Relationship with EBV and KSHV infection. The spectrum of lymphomas occurring in HIV-infected patients includes pathologic subtypes displaying specific association with distinct viruses. BL and DLBCL-IB with plasmacytoid differentiation are often HIV associated and closely linked to EBV infection. The HIV-associated DLBCL-IB is distinct from other large cell lymphomas occurring in both HIV-seropositive and -seronegative patients because HIV-associated DLBCL-IB lymphomas display a plasma cell–related phenotype; interestingly, the gene expression profile of PEL is plasmablastic. Therefore, most HIV-associated lymphoproliferative disorders, including primary central nervous system lymphoma, systemic DLBCL IB-plasmacytoid, KSHV + PEL and its solid variant, and PBLs of the oral cavity type, display a phenotype related to plasma cells and are linked to EBV infection. Red circle indicates positive infection (inside are the percentages); empty circle indicates no infection. DLBCL-IB indicates diffuse large B-cell lymphoma-immunoblastic; EBV, Epstein-Barr virus; KSHV, Kaposi sarcoma herpesvirus; MCD, multicentric Castleman disease; PBL, plasmablastic lymphoma; and PEL, primary effusion lymphoma.

Figure 2

The spectrum of HIV-associated lymphomas. Relationship with EBV and KSHV infection. The spectrum of lymphomas occurring in HIV-infected patients includes pathologic subtypes displaying specific association with distinct viruses. BL and DLBCL-IB with plasmacytoid differentiation are often HIV associated and closely linked to EBV infection. The HIV-associated DLBCL-IB is distinct from other large cell lymphomas occurring in both HIV-seropositive and -seronegative patients because HIV-associated DLBCL-IB lymphomas display a plasma cell–related phenotype; interestingly, the gene expression profile of PEL is plasmablastic. Therefore, most HIV-associated lymphoproliferative disorders, including primary central nervous system lymphoma, systemic DLBCL IB-plasmacytoid, KSHV + PEL and its solid variant, and PBLs of the oral cavity type, display a phenotype related to plasma cells and are linked to EBV infection. Red circle indicates positive infection (inside are the percentages); empty circle indicates no infection. DLBCL-IB indicates diffuse large B-cell lymphoma-immunoblastic; EBV, Epstein-Barr virus; KSHV, Kaposi sarcoma herpesvirus; MCD, multicentric Castleman disease; PBL, plasmablastic lymphoma; and PEL, primary effusion lymphoma.

Close modal

In contrast to other lymphomas, a high frequency of EBV association has been shown in HL (80%–100%) tissues from HIV-infected people and the EBV-transforming protein, EBV-encoded latent membrane protein-1 (LMP-1), is expressed in virtually all HIV-HL cases.22-28  On this basis, HL in HIV-infected persons appears to be an EBV-driven lymphoma (Figure 3).25 

Figure 3

Expression of EBV LMP1 in the Reed-Sternberg cells of Hodgkin lymphoma. The immunostaining with LMP1 antibody is cytoplasmic (arrows). Original magnifications: ×400 (left), ×1000 (right). Images kindly provided by Dr L. Young, Birmingham. Images were assembled using Adobe Photoshop 6 (Adobe Systems, San Jose, CA).

Figure 3

Expression of EBV LMP1 in the Reed-Sternberg cells of Hodgkin lymphoma. The immunostaining with LMP1 antibody is cytoplasmic (arrows). Original magnifications: ×400 (left), ×1000 (right). Images kindly provided by Dr L. Young, Birmingham. Images were assembled using Adobe Photoshop 6 (Adobe Systems, San Jose, CA).

Close modal

PEL and its solid variant

PEL is a distinct type of B-cell non-Hodgkin lymphoma (NHL) that presents most frequently in body cavities as lymphomatous effusions without an associated tumor mass. In addition, some lymphomas occurring in HIV-infected individuals have very similar molecular and immunophenotypic characteristics, and yet do not involve body cavities. Thus, an extracavitary variant of PEL has been recognized. A defining property of PEL is its consistent association with KSHV infection. Most cases are also coinfected by EBV. It is believed that KSHV, rather than EBV, is a driving force in these tumors, as in PEL, at least 5 KSHV viral genes are expressed, which provide proliferative and antiapoptotic signals. In contrast, EBV has a restricted latency pattern of gene expression in PEL, where only EBNA1 and EBERs are expressed.

PELs have a distinctive set of morphologic and immunophenotypic properties, which prompted their description in the literature29,30  even before the discovery of KSHV. In cytospin preparations, the cells can have a range of appearances, from cells with anaplastic morphology to large immunoblastic or plasmablastic cells. Binucleated or multinucleated cells resembling Reed-Sternberg (RS) cells can be found. Nuclei are large with prominent nucleoli. The cytoplasm is usually abundant and is deeply basophilic. Some cases have cytoplasmic vacuoles and frequently a perinuclear hof consistent with plasmacytoid differentiation is seen. There is a high proliferation rate, as appreciated by numerous mitotic figures. The cells often appear more uniform in histologic sections than in cytospin preparations.31,32  Extracavitary PELs usually are immunoblastic in appearance, and have a high mitotic rate and variable amounts of apoptotic debris.13  However, they exhibit a spectrum of morphologic features, ranging from cells with moderate amounts of amphophilic to acidophilic cytoplasm with occasional perinuclear hofs and large nuclei containing a single prominent, centrally placed nucleolus, to cases with more variable and pleomorphic cells, some containing binucleated or multinucleated cells reminiscent of RS cells and variants. Some cases have a prominent “starry-sky” appearance.

PEL cells commonly express CD45, but lack pan-B-cell markers, including CD19, CD20, and CD79a as well as surface and cytoplasmic immunoglobulins.29,32  However, cases of extracavitary PEL express immunoglobulins somewhat more often than the classical effusion PEL.13  Expression of BCL6 is generally absent. Activation and plasma cell markers and miscellaneous non–lineage-associated antigens such as HLA-DR, CD30, CD38, Vs38c, CD138, and EMA are often expressed.11,12,18,20,21  PELs usually lack T/natural killer (NK)–cell antigens, although aberrant expression of T-cell markers may occur.33-35 

Many cases are sent to cytopathology laboratories, where a cell block or smear shows the presence of neoplastic cells, but lymphoma may not be suspected. In conjunction with the aberrant phenotype these cases may be difficult to classify. A helpful procedure is immunohistochemistry for KSHV, which is best achieved with antibodies to the viral latency-associated nuclear antigen (LANA; ORF73). Positive staining shows characteristic nuclear dots. Extracavitary PELs are frequently classified as diffuse large cell, immunoblastic, or anaplastic large cell lymphomas in HIV+ individuals, and the diagnosis is made by immunohistochemistry for LANA, which allows demonstration of the presence of KSHV in practically all the lymphoma cells.13,36  Other KSHV proteins are also present in both cavitary and extracavitary PEL, in particular viral interleukin-6 (vIL-6) which is expressed by a variable proportion of neoplastic cells, so detection of this protein by immunohistochemistry can provide a confirmatory assay. In situ hybridization for EBV EBERs is also useful, as many cases contain both viral genomes. Further confirmation can be provided by molecular techniques, such as polymerase chain reaction documenting the presence of the viral genome.

PELs are of B-cell origin, which can be demonstrated by the presence of clonal immunoglobulin gene rearrangements. Evidence points toward a post–germinal center B-cell derivation, as most PELs contain somatic hypermutation of Ig genes as well as frequent somatic hypermutation of the noncoding region of the BCL6 gene.37,38  Consistent with this notion is the expression of plasma cell markers such as CD138/Syndecan-1. Recently, gene expression analysis of PEL showed features most similar to AIDS immunoblastic lymphoma and multiple myeloma, again indicating a pre–plasma cell or “plasmablastic” profile.21,39 

PEL and KSHV unrelated effusion lymphomas

PEL needs to be differentiated from those lymphomas occurring in patients in whom effusions complicate a tissue-based lymphoma, the so-called secondary lymphomatous effusion. However, secondary lymphomatous effusions closely mimic phenotypic and genotypic features of the corresponding tissue-based lymphoma and are consistently devoid of KSHV infection40-42  (Figure 4).

Figure 4

Classification and differential diagnosis of non-Hodgkin lymphomas involving the serous body cavities and presenting as effusion lymphomas. Lymphomas primarily involving the serous body cavities include a certain number of BLs, mainly occurring in the context of AIDS, which present as primary lymphomatous effusions without mass formation. The most specific biologic markers discriminating PEL from BL presenting as a primary lymphomatous effusion are represented by KSHV infection (assessed by ORF73/LNA-1 immunoreactivity), which clusters with PEL, and by translocation of the c-MYC proto-oncogene, which segregates with BL. KSHV-unrelated large B-cell lymphomas, also termed as KSHV-unrelated PEL-like lymphomas, can be differentiated from PEL because the neoplastic cells do not display evidence of KSHV infection, but display features related to large B-cell lymphoma. KSHV/HHV8 indicates Kaposi sarcoma herpesvirus/human herpesvirus 8; EBV, Epstein-Barr virus; IBL, immunoblastic lymphoma; ALCL, anaplastic large cell lymphoma; DLBCL, diffuse large B-cell lymphoma; BL, Burkitt lymphoma; and PAL, pyothorax-associated lymphoma, now called DLBCL with chronic inflammation.

Figure 4

Classification and differential diagnosis of non-Hodgkin lymphomas involving the serous body cavities and presenting as effusion lymphomas. Lymphomas primarily involving the serous body cavities include a certain number of BLs, mainly occurring in the context of AIDS, which present as primary lymphomatous effusions without mass formation. The most specific biologic markers discriminating PEL from BL presenting as a primary lymphomatous effusion are represented by KSHV infection (assessed by ORF73/LNA-1 immunoreactivity), which clusters with PEL, and by translocation of the c-MYC proto-oncogene, which segregates with BL. KSHV-unrelated large B-cell lymphomas, also termed as KSHV-unrelated PEL-like lymphomas, can be differentiated from PEL because the neoplastic cells do not display evidence of KSHV infection, but display features related to large B-cell lymphoma. KSHV/HHV8 indicates Kaposi sarcoma herpesvirus/human herpesvirus 8; EBV, Epstein-Barr virus; IBL, immunoblastic lymphoma; ALCL, anaplastic large cell lymphoma; DLBCL, diffuse large B-cell lymphoma; BL, Burkitt lymphoma; and PAL, pyothorax-associated lymphoma, now called DLBCL with chronic inflammation.

Close modal

A more subtle diagnosis consists in differentiating PEL from other types of lymphomas primarily involving the serous body cavities that can present with a primary neoplastic effusion (Figure 4).41,43-47  Many of these cases are KSHV-unrelated large B-cell lymphomas, also termed KSHV-unrelated PEL-like lymphomas.45  KSHV-unrelated PEL-like lymphoma cases are associated with hepatitis C virus (HCV) (30%–40%). The most involved sites are peritoneum and pleura; lymphoma cells most commonly show large cell type morphology (80%) and B-cell immunophenotype (90%). In contrast, PEL cases are universally associated with KSHV and mostly occur in immunodeficiency states. They demonstrate distinctive morphology, and lack c-MYC gene rearrangement and B cell–associated antigen expression. It seems that PEL and KSHV-unrelated PEL-like lymphomas are different in terms of pathogenesis, morphologic-immunophenotypic features, clinical behavior, and prognosis.

Large B-cell lymphoma arising in KSHV-associated MCD

Our current knowledge indicates that Castleman disease48  (CD) actually represents several different clinicopathologic entities. Prior to the discovery of KSHV, 2 histopathologic types of CD had been described: (1) the hyaline vascular (HV) variant that is the most common form, affecting 90% of patients and usually involving a single lymph node in the mediastinum, and (2) the plasma cell (PC) variant, which is characterized by hyperplastic germinal centers, abundant plasma cells in the interfollicular areas, persistence of sinuses, and associated clinical and laboratory abnormalities.49  Two clinical entities were also described: (1) the localized form, which usually presents as lymph node hyperplasia in a single lymph node–bearing region (in most cases, the mediastinum) and that resolves with resection, and (2) MCD, which manifests as generalized lymphadenopathy with systemic symptoms and is characterized by a more aggressive clinical course and the potential for malignant transformation. MCD resembles the PC variant histopathologically, and it is frequently described as such in the literature. The PC variant can also be localized and the histologic appearance of MCD is somewhat different, so MCD should be classified separately. Besides primary MCD, cases associated with other diseases (secondary MCD) are common, and MCD represents one of the most ubiquitous associations in the literature. Secondary MCD is a large and heterogeneous group of clinical entities and is often referred to as interleukin-6 (IL-6) syndrome because of evidence that an overproduction of IL-6, probably in association with other cytokines, occurs in MCD-associated diseases as well as in MCD itself, suggesting a common underlying pathogenetic mechanism.

Understanding of the pathogenesis of MCD has greatly increased since the discovery of MCD's association with KSHV, which has been found in approximately half of the cases of MCD occurring in immunocompetent patients and in almost all those infected with HIV, suggesting a pathogenetic role in this disease.50  A plasmablastic variant of MCD characterized by the presence of medium-sized to large plasmablastic cells scattered in the mantle zones of the follicles has been described, most frequently in HIV-infected individuals. Whereas immunoglobulin M (IgM)–positive immunoblasts have been usually described in the interfollicular region in MCD,51  a unique population of cells with a similar morphology was found in the mantle zone of a subset of cases of MCD in association with KSHV infection.17,52-54  The cells harboring KSHV in MCD have been called plasmablasts, but they have been described as having classic immunoblastic features, including a moderate amount of amphophilic cytoplasm and a large vesicular nucleus containing 1 or 2 prominent nucleoli.17,52-54  These KSHV-positive immunoblasts are immature cells that express cytoplasmic IgMλ, have a blastic morphology, and are seen predominantly in the mantle zones. The consistent restricted expression of lambda light chain in the KSHV-positive plasmablasts is intriguing and could be involved in the mechanism of KSHV entry in the cells or selection for those cells. These cells may be scattered or found in small confluent clusters, sometimes coalesced to form foci of “microlymphomas” or in large sheets of cells thought to represent frank lymphomas.17,55  One study reported analysis of clonality and showed that despite monotypic expression of IgMλ, the scattered plasmablasts in MCD are polyclonal; 6 of the 8 cases with small clusters called microlymphomas were also polyclonal, whereas the investigated cases of large B-cell lymphomas arising in KSHV-associated MCD were monoclonal.56 

Secondary MCD can be found in association with a variety of pathologic conditions, including HIV infection, plasma-cell dyscrasias (ie, POEMS syndrome), KS, B-cell lymphoma, and HL. In KSHV-positive cases, a common association is KS17,50  and a specific variant of NHL referred to as “plasmablastic lymphoma,”17  the so-called large B-cell lymphoma arising in KSHV-associated MCD. This lymphoma is specifically associated with KSHV and is considered KSHV-linked disease entity. The lymphoma cells show exactly the same phenotypic features as the plasmablasts described in MCD, including cytoplasmic IgMλ expression and lack of EBV infection,17,57  suggesting that the plasmablastic variant of MCD could precede the development of frank KSHV-positive lymphoma. PEL is different from large B-cell lymphoma arising in KSHV-associated MCD in that the tumor cells in PEL frequently lack expression of B-cell antigens and are frequently coinfected with EBV. In addition, in contrast to PEL, large B-cell lymphomas arising in KSHV-associated MCD lack somatic hypermutation of immunoglobulin genes and are therefore thought to derive from pre–plasma cells that bypassed the germinal center.55 

PBLs of the oral cavity type

PBLs of the oral cavity type were first described as lymphomas occurring mostly in HIV+ individuals having an unusual immunophenotype (low or no CD45 and CD20), and frequent presence of EBV.15  This rare entity typically involves the jaw and oral cavity of HIV patients even if it has been documented in other sites than the oral cavity such as the anorectum, nasal and paranasal regions, skin, testes, bones, and lymph nodes. PBLs of the oral cavity type are composed of large neoplastic cells with a very high proliferation rate displaying a marked degree of plasma cell differentiation.15,16,58  Since lymphomas associated with MCD, which contain KSHV, have also been called plasmablastic lymphomas, these 2 diseases have been confused. However, they represent distinct entities, and most studies have found that PBLs of the oral cavity type do not contain KSHV.58  In PBLs of the oral cavity type, features of CD are absent. The neoplastic cells have round nuclei, moderately clumped chromatin, a single prominent nucleolus, and moderate to abundant basophilic cytoplasm with an excentric nucleus.58  The mitotic rate is very high, and there are frequent apoptotic cells and single-cell necrosis. The cytoplasm is usually deep basophilic with a paranuclear hof, and binucleation and multinucleation are common. Cells with features of maturing plasma cells can be seen and there is usually a spectrum of differentiation than can be appreciated morphologically. Phenotypically, PBLs of the oral cavity type display an unusual profile characterized by weak or absent expression of B-cell antigens (eg, CD20 and PAX5) coupled to strong immunostaining with the plasma cell markers CD138/syndecan-1, MUM1/IRF4, and VS38c (Figures 5,6). CD45 is expressed in most cases, but can be weak or negative. A recent study reported that only 5 of 11 cases express cytoplasmic Ig, which were IgGκ or IgGλ.58  EBV can be detected by EBER ISH, but LMP-1 and LMP2 are not expressed, consistent with a restricted latency, which is in contrast to AIDS-related IB lymphomas that usually express LMP-1.2 

Figure 5

Strong immunostaining with the plasma cell markers CD138/syndecan1 and MUM1/IRF4 in plasmablastic lymphoma. Images were taken using a Nikon Eclipse 80i microscope (Nikon, Tokyo, Japan) with a pan fluor 40×/0.75 objective and Nikon digital sight DS-Fi1 camera equipped with control unit-DS-L2 (Nikon). Images were assembled using Adobe Photoshop 6 (Adobe Systems).

Figure 5

Strong immunostaining with the plasma cell markers CD138/syndecan1 and MUM1/IRF4 in plasmablastic lymphoma. Images were taken using a Nikon Eclipse 80i microscope (Nikon, Tokyo, Japan) with a pan fluor 40×/0.75 objective and Nikon digital sight DS-Fi1 camera equipped with control unit-DS-L2 (Nikon). Images were assembled using Adobe Photoshop 6 (Adobe Systems).

Close modal
Figure 6

Lymphomas specifically arising in HIV-induced immunosuppression. Stage of differentiation and the putative cell of origin. Lymphoma histotypes occurring specifically in HIV-infected patients exhibit a common normal cellular counterpart that might be defined as plasmablast. The figure defines the following main subgroups of lymphoma with plasmablastic differentiation: (1) HIV-associated lymphoma showing IB morphology with plasmacytoid differentiation; these lymphomas can be either systemic or primary central nervous system lymphomas; (2) tumors classified as PELs and their extracavitary variant exhibiting infection by both KSHV and EBV; (3) tumors classified as PBL of the oral cavity type, showing a monomorphic population of immunoblasts with no or minimal plasmacytic differentiation; most patients are HIV infected and tumor cells are EBV positive but KSHV negative; most cases present in the oral mucosa, whereas a significant number of cases present in other extranodal or nodal site; and (4) large B-cell lymphoma arising in KSHV-associated multicentric Castleman disease (MCD) consisting of KSHV-infected plasmablasts, which show evidence of light chain restriction and may represent a monotypic cell population, found in small clusters surrounding or replacing follicles, in MCD. Therefore, important features to subclassify these neoplasms include the stage of differentiation of the putative cell of origin and association with viruses. Lymphomas with plasmablastic differentiation are a heterogeneous group of neoplasms with different clinicopathological characteristics and different associations with specific viruses. DLBCL-IB indicates diffuse large B-cell lymphoma-immunoblastic; EBV, Epstein-Barr virus; KSHV, Kaposi sarcomaherpesvirus; MCD, multicentric Castleman disease; PBL, plasmablastic lymphoma; PC, plasma cell; and PEL, primary effusion lymphoma.

Figure 6

Lymphomas specifically arising in HIV-induced immunosuppression. Stage of differentiation and the putative cell of origin. Lymphoma histotypes occurring specifically in HIV-infected patients exhibit a common normal cellular counterpart that might be defined as plasmablast. The figure defines the following main subgroups of lymphoma with plasmablastic differentiation: (1) HIV-associated lymphoma showing IB morphology with plasmacytoid differentiation; these lymphomas can be either systemic or primary central nervous system lymphomas; (2) tumors classified as PELs and their extracavitary variant exhibiting infection by both KSHV and EBV; (3) tumors classified as PBL of the oral cavity type, showing a monomorphic population of immunoblasts with no or minimal plasmacytic differentiation; most patients are HIV infected and tumor cells are EBV positive but KSHV negative; most cases present in the oral mucosa, whereas a significant number of cases present in other extranodal or nodal site; and (4) large B-cell lymphoma arising in KSHV-associated multicentric Castleman disease (MCD) consisting of KSHV-infected plasmablasts, which show evidence of light chain restriction and may represent a monotypic cell population, found in small clusters surrounding or replacing follicles, in MCD. Therefore, important features to subclassify these neoplasms include the stage of differentiation of the putative cell of origin and association with viruses. Lymphomas with plasmablastic differentiation are a heterogeneous group of neoplasms with different clinicopathological characteristics and different associations with specific viruses. DLBCL-IB indicates diffuse large B-cell lymphoma-immunoblastic; EBV, Epstein-Barr virus; KSHV, Kaposi sarcomaherpesvirus; MCD, multicentric Castleman disease; PBL, plasmablastic lymphoma; PC, plasma cell; and PEL, primary effusion lymphoma.

Close modal

EBV and lymphoma in AIDS patients

Diffuse large cell B-cell lymphoma.

The transforming EBV protein LMP-1 is frequently expressed in DLBCL.59,60  LMP-1 plays a crucial role in the transformation of B-lymphocytes by EBV (reviewed in Young and Rickinson61 ). Thus, LMP-1 transforms rodent fibroblasts,62  transgenic mice that express LMP-1 in B cells show increased development of B-cell lymphomas,63  and LMP-1 deletion mutants of EBV are compromised in their ability to immortalize human primary B cells.64  LMP-1 activates the NFkB as well as the JNK and p38 pathways,65-67  by recruiting cellular TRAF 1–3 and TRADD molecules to 2 short sequence motifs, CTAR-1 and CTAR-2, respectively, in the cytoplasmic domain of the LMP-1 molecule.68-70  In B cells, LMP-1 increases the expression of the antiapoptotic proteins A20 and bcl-2, the adherence molecule CD54/ICAM-1, the cell-cycle regulator p27Kip,71  and many others (reviewed in Brinkmann and Schulz72 ). In DLBCL, expression of LMP-1 correlates inversely with the expression of BCL6, a marker for germinal center B cells, suggesting that, among DLBCLs, the impact of EBV LMP-1 is likely to be strongest in tumors representing a post–germinal center plasmacytic differentiation profile.73  In addition, knockdown of LMP-1 in cell lines derived from AIDS-DLBCL results in apoptosis, indicating that this viral oncoprotein plays a role in lymphoma pathogenesis.74 

EBV-associated DLBCLs have therefore been considered as EBV-driven lymphoproliferations occurring in the context of a defective T-cell immunity against EBV.75  However, unlike EBV-driven lymphoproliferative disease in transplant recipients, which includes monoclonal, oligoclonal, as well as polyclonal B-cell proliferations, DLBCL is always monoclonal. This suggests that, in addition to the effects contributed by EBV LMP-1, additional factors such as genetic damage are likely to contribute to the pathogenesis of AIDS-DLBCL.

Burkitt lymphoma.

About 30% to 60% of AIDS-BLs are EBV positive and the transforming EBV protein LMP-1 is not expressed in BL.61  Although not essential in the pathogenesis of BL, EBV supports tumor development. EBNA-1, a viral protein required for the replication and maintenance of the latent viral episomal DNA, is found consistently in BL cells.61  EBNA-1 transgenic mice develop B-cell lymphoma with a very long latency,76  and EBNA-1 and c-myc may cooperate.77  The presence of latent EBV in BL cells has been shown to promote genetic instability,78  suggesting a mechanism by which latent EBV could contribute to genetic alterations required for the development of BL. In addition, some latent EBV transcription patterns found in BL produce viral proteins that are likely to protect BL cells from apoptosis induced by deregulated c-myc expression.79  The importance of apoptosis protection during B-cell immortalization has recently been highlighted by the failure of EBV deletion mutants lacking both viral bcl-2 homologues (BALF1, BHRF1) to efficiently immortalize human B cells.80  Given the strong apoptotic effects of overexpressed c-myc, the role of EBV in some cases of BL could therefore consist of protecting BL cells against this side effect of c-MYC translocation.

Hodgkin lymphoma.

The latent EBV proteins EBNA-1, LMP-1, and LMP2A are expressed in the RS cells, the malignant cell population of this tumor.81  RS cells are derived from B cells that have passed through the germinal center, as shown by the presence of somatic mutations in the rearranged Ig variable region of their immunoglobulin genes.82  Notably, many of these hypermutations are incompatible with the expression of a functional B-cell receptor (BCR), suggesting that RS cells may have developed from germinal center B cells that should have been eliminated by apoptosis, but managed to survive.83,84 

LMP2A interferes with normal B-cell development, allows BCR-negative B cells to leave the bone marrow/colonize peripheral lymphoid organs,85  and induces a transcriptome pattern in B cells, which resembles that of HL RS cells.86  Following EBV infection, LMP2A is essential for the survival and continued proliferation of germinal center B cells lacking a functional B-cell receptor.87,88  LMP2A may therefore promote the survival of “crippled” germinal center B cells and could thus aid their development into RS cells.

LMP-1 may also induce an “HL-like” transcriptional program in germinal center B cells.89  Among the cellular genes up-regulated by LMP-1 in HL cells is bmi-1, a polycomb family member known to cause lymphoma in transgenic mice and to down-regulate the ATM tumor suppressor.90  EBNA-1 was shown to induce CCL-20 secretion in RS cell lines and to thereby promote the migration of regulatory T cells, which could be envisaged to downmodulate EBV-specific T-cell responses.91  Protein tyrosine phosphatase receptor kappa (PTPRK) suppresses the growth of HL cell lines and is downmodulated by EBV.92  These results provide suggestions of how EBV LMP-1, LMP2A, and EBNA-1 may contribute to the development of RS cells.

Kaposi sarcoma herpesvirus and AIDS lymphoma

PEL.

PEL cells contain multiple copies (in the order of 50–150 copies/cell) of episomal KSHV genomes.93  In most cells, a latent viral gene expression pattern involves the expression of the LANA, a viral D-type cyclin homologue (vcyc), a viral homologue of FLICE inhibitory protein (vFLIP), a pre-miRNA transcript encoding 11 viral miRNAs, as well as vIRF3/K10.5/LANA-2.94-101  In addition, a homologue of IL-6 (vIL-6) is also expressed in some PEL cells.54,102,103 

A detailed description of the functional properties of individual KSHV proteins can be found in recent review articles.101,104,105  The evidence for an involvement of the above-cited 5 to 6 KSHV genes in the pathogenesis of PEL is as follows: gene silencing experiments using shRNA or siRNA106,107  indicate that silencing of vFLIP and/or vcyc leads to increased apoptosis of PEL cell lines. Since vcyc and vFLIP are translated from the same bicistronic latent transcript, silencing of vcyc also lead to the silencing of vFLIP, and vice versa.106,107  However, apoptosis was found to be due to vFLIP suppression as reconstitution with transfected vcyc does not rescue the cells from apoptosis.106  vFLIP is a potent activator of the NFkB pathway,108-111  and NFkB inhibition also induces apoptosis in PEL cells,112  providing additional evidence that vFLIP is essential for the survival of PEL cells. Its downstream effects include the induction of IL-6, an important growth factor for B cells, as well as the cellular antiapoptotic factors cFLIPL, cIAP-1, and cIAP-2.106,111 

The viral D-type cyclin homologue vcyc associates with cdk2, cdk4, and cdk6 but appears to promote phosphorylation of its targets mainly in concert with cdk6.94-101,103-113  Its targets include not only RB, but also other cellular targets including histone H1, Id2, CDC6, cdc25A, Orc-1, the antiapoptotic protein bcl-2, and the cdk inhibitors p27Kip and p21CIP.113,114  Phosphorylation of p27Kip by the vcyc/cdk6 complex on Ser10 during latency leads to sequestration of p27Kip in the cytoplasm, thereby allowing PEL cells to proliferate in the presence of high p27Kip levels.113  Likewise, phosphorylation of p21CIP1 on serine 130 by vcyc allows vcyc to bypass the p21CIP1-mediated G1 arrest.114  Vcyc has been shown to promote S phase entry and also to induce apoptosis in cells with high cdk6 expression, which can be counteracted by the action of the viral bcl-2 homologue, vbcl-2115 ; it can induce a DNA damage response in endothelial cells.116  It is likely that some of these biochemical features of vcyc will play a role in PEL pathogenesis.

KSHV LANA also interacts with several cellular components that have been linked to cancer development. It binds to and antagonizes p53 and Rb, sequestrates GSK-3β and thereby stabilizes β-catenin as well as c-myc, and additionally activates c-myc–mediated transcription by promoting its phosphorylation.117-120  A DNA damage response pathway appears to be active in PEL cells, which is normally balanced by the ability of LANA to interact with p53 and its E3 ligase, mdm2; disruption of the LANA/p53/mdm2 complex by nutlin 3a, an inhibitor of the p53-mdm2 interaction, induces apoptosis in KSHV-infected PEL cell lines, but not in EBV-transformed LCLs.121  This observation underlines the importance of the LANA/p53/mdm2 complex in PEL cells for their survival. However, in contrast to vFLIP, silencing of LANA only reduced the KSHV genome copy number without affecting cell survival.

KSHV vIRF3 interferes with p53-induced transcription as well as p53- and protein kinase R (PKR)–mediated apo-ptosis97-101,103-122 ; its silencing by siRNA/shRNA in PEL cell lines results in an increased apoptosis and caspase 3/7 activity, suggesting that this protein also contributes to the survival of PEL cells.123 

A fourth latent transcript in PEL cells serves as a precursor RNA for 12 microRNAs (miRNAs), small 19- to 23-nt RNAs that regulate cellular mRNA turnover or stability.98-100  One of these, miR-K12-11, has been found to target the same cellular mRNAs as miR-155/BIC, a cellular miRNA regulating the germinal center reaction during B-cell maturation.124-126  Both miR-K12-11 and miR-155 down-regulate several proapoptotic cellular genes, such as LDOC1, Bim, BCLAF1 (Bcl2-associated transcription factor 1), and the NFkB regulator BAZF.125,126  MiR-K12-11 may therefore be involved in the late stages of B-cell differentiation, could contribute to the plasmablastic phenotype of PEL cells, or could play a role in the protection of PEL cells against apoptosis.

The viral IL-6 homologue, vIL-6, is expressed in a subpopulation of PEL cells in vivo and in many KSHV-infected B cells in MCD lymphoid follicles.54,103,127  It induces proliferation, angiogenesis, and hematopoiesis in IL-6–dependent cell lineages127-129  and serves as an autocrine factor in PEL cell lines130 ; it also induces vascular endothelial growth factor (VEGF), which has been implicated in the pathogenesis of PEL and KS.131  A single chain antibody to vIL-6 blocking its interaction with the IL-6 receptor complex was found to inhibit the proliferation of a PEL cell line and to inhibit vIL-6–induced STAT 3 phosphorylation in vIL-6–transfected cells.132  Therefore vIL-6 may contribute to PEL cell proliferation and to the angiogenesis noted in patients with this lymphoma.

MCD.

Among KSHV-associated pathologies, MCD appears to be the one with the highest number of productively infected cells.53,103  A sizeable fraction of MCD B cells expresses vIL-6 and it is thought that its downstream effects on B-cell proliferation and VEGF secretion play a role in their proliferation and in the strong angiogenic component characteristic for MCD lesions. In patients with MCD, exacerbations of the disease were reported to correlate with increased viral load and increased IL-6 and IL-10 levels, underlining the importance of productive viral replication and cellular cytokines in the pathogenesis of this disorder.133 

Conclusion

From the direct transforming role of EBV in many transplant-associated and some AIDS lymphomas, via the impact of EBV and KSHV on the DNA damage response in infected B cells, the contribution of individual EBV and KSHV proteins to protection against apoptosis and promotion of cell survival to possible direct effects on B-cell differentiation and maturation, these viruses have developed multiple strategies that allow them to act as cofactors in lymphoma development acting in concert with immune suppression and presumably other oncogenic factors (Figure 7).

Figure 7

A diagram of the genomes of EBV and KSHV/HHV8 and their expression pattern in EBV- and KSHV-associated AIDS lymphoma. (A) The EBV genome is shown in the same orientation as the KSHV genome. The terminal repeats of the 2 viral genomes are indicated by a pair of taller boxes at the end of the long unique coding region. EBV adopts its most restricted pattern of gene expression (latency pattern I) in BL and PEL cells; this involves expression of EBNA-1 from the Qp promoter, of the untranslated EBERs, and probably of a group of transcripts from the Bam A region. In some BL tumors, the use of an alternative latent EBV promoter, Wp, leads to the expression of EBNA-3A,B,C in the absence of EBNA-2 and LMP-1, resulting a significant protection against apoptosis of c-myc–expressing cells.79  In HL, EBV adopts latency pattern II, which involves expression of EBNA-1, the EBERs, and the 2 latent membrane proteins, LMP-1 and LMP-2A. LMP-2A is translated from a transcript that spans the terminal repeats in the circular viral episome found during latency. In DLBCL, EBV latency pattern III includes the expression of EBNA-2, EBNA-LP, EBNA-3A,B,C from the latent Wp promoter, as shown. Details on the function of EBNA-2, EBNA-LP, and EBNA-3A,B,C can be found in a recent review.61  (B) KSHV/HHV8 genome and viral genes expressed in PEL or MCD. The latent KSHV genes LANA, v-cyc, vFLIP, K12/kaposin, and vIRF-3/K10.5/LANA-2 are shown as black stippled boxes; the position of the KSHV miRNAs is indicated by vertical lines. These genes are expressed in the majority of tumor cells in vivo and in PEL cell lines. Viral genes expressed in only a subpopulation of PEL or MCD cells (eg, vIL6) are cross-hatched, and those expressed only during the later stages of the productive (lytic) viral replication cycle are stippled. The viral gene expression pattern in PEL is more restricted than in MCD. Whereas LANA, vcyc/vFLIP, the miRNAs, and vIRF-3/K10.5/LANA-2 are expressed in most, vIL6 is expressed only in a small proportion of lymphoma cells. Other lytic genes are only rarely expressed and are therefore not shown in this diagram for PEL. In MCD, several lytic KSHV genes are expressed in a few cells, suggesting noticeable productive viral replication in this condition.

Figure 7

A diagram of the genomes of EBV and KSHV/HHV8 and their expression pattern in EBV- and KSHV-associated AIDS lymphoma. (A) The EBV genome is shown in the same orientation as the KSHV genome. The terminal repeats of the 2 viral genomes are indicated by a pair of taller boxes at the end of the long unique coding region. EBV adopts its most restricted pattern of gene expression (latency pattern I) in BL and PEL cells; this involves expression of EBNA-1 from the Qp promoter, of the untranslated EBERs, and probably of a group of transcripts from the Bam A region. In some BL tumors, the use of an alternative latent EBV promoter, Wp, leads to the expression of EBNA-3A,B,C in the absence of EBNA-2 and LMP-1, resulting a significant protection against apoptosis of c-myc–expressing cells.79  In HL, EBV adopts latency pattern II, which involves expression of EBNA-1, the EBERs, and the 2 latent membrane proteins, LMP-1 and LMP-2A. LMP-2A is translated from a transcript that spans the terminal repeats in the circular viral episome found during latency. In DLBCL, EBV latency pattern III includes the expression of EBNA-2, EBNA-LP, EBNA-3A,B,C from the latent Wp promoter, as shown. Details on the function of EBNA-2, EBNA-LP, and EBNA-3A,B,C can be found in a recent review.61  (B) KSHV/HHV8 genome and viral genes expressed in PEL or MCD. The latent KSHV genes LANA, v-cyc, vFLIP, K12/kaposin, and vIRF-3/K10.5/LANA-2 are shown as black stippled boxes; the position of the KSHV miRNAs is indicated by vertical lines. These genes are expressed in the majority of tumor cells in vivo and in PEL cell lines. Viral genes expressed in only a subpopulation of PEL or MCD cells (eg, vIL6) are cross-hatched, and those expressed only during the later stages of the productive (lytic) viral replication cycle are stippled. The viral gene expression pattern in PEL is more restricted than in MCD. Whereas LANA, vcyc/vFLIP, the miRNAs, and vIRF-3/K10.5/LANA-2 are expressed in most, vIL6 is expressed only in a small proportion of lymphoma cells. Other lytic genes are only rarely expressed and are therefore not shown in this diagram for PEL. In MCD, several lytic KSHV genes are expressed in a few cells, suggesting noticeable productive viral replication in this condition.

Close modal

Combined chemotherapy and rituximab

Before the introduction of highly active antiretroviral therapy (HAART), several studies demonstrated that the use of aggressive chemotherapy regimens leads to high mortality rate because of the incidence of opportunistic infections (OIs). Therefore low-dose M-BACOD (methotrexate, bleomycin, doxorubicin, cyclophosphamide, vincristine, and dexamethasone)134  or CHOP (cyclophosphamide, vincristine, doxorubicin, prednisone)135  were considered the gold standard in the treatment of these patients even if other reports suggested a superiority of continuous infusional chemotherapy regimens (CDE [cyclophosphamide, doxorubicin, etoposide] or EPOCH [cyclophosphamide, doxorubicin, etoposide, vincristine, prednisone]).136,137 

The introduction of rituximab has significantly improved survival from NHL in the general population,138-141  and based on these data, several authors have explored the feasibility and effectiveness of rituximab plus chemotherapy in patients with HIV-NHL. All published data suggest the high activity of rituximab plus chemotherapy in comparison with historical control with chemotherapy alone (see Table 1 for the comparison of R-CDE vs CDE) even if a slight increase in the rate of OIs has been reported.141-146 Table 2 summarizes the results of these studies.

Table 1

Comparison between CDE and R-CDE

CDE (95% CI)R-CDE (95% CI)
No. of patients 55 74 
Median age, y 40 38 
Median CD4 count/μL 227 161 
Histology, % 
    Burkitt or Burkitt-like 22 28 
    Diffuse large cell or variants 78 72 
Age-adjusted IPI, % 
    Low or low intermediate 42 43 
    High or high intermediate 58 57 
Complete remission rate, % 45 (30–58) 70 (59–81) 
Disease-free survival at 2 y, % 38 (25–51) 59 (47–71) 
Overall survival at 2 y, % 45 (20–58) 64 (52–76) 
CDE (95% CI)R-CDE (95% CI)
No. of patients 55 74 
Median age, y 40 38 
Median CD4 count/μL 227 161 
Histology, % 
    Burkitt or Burkitt-like 22 28 
    Diffuse large cell or variants 78 72 
Age-adjusted IPI, % 
    Low or low intermediate 42 43 
    High or high intermediate 58 57 
Complete remission rate, % 45 (30–58) 70 (59–81) 
Disease-free survival at 2 y, % 38 (25–51) 59 (47–71) 
Overall survival at 2 y, % 45 (20–58) 64 (52–76) 

Data are from Spina et al.141 

CDE indicates cyclophosphamide, doxorubicin, etoposide; and R, rituximab.

Table 2

Rituximab and chemotherapy in HIV-related non-Hodgkin lymphomas: review of the literature

R-CDE141 R-CHOP143 R-CHOP144 R-CHOP145 R-EPOCH146 
No. of patients 74 61 95 60 51 
Stage III-IV (%) 70 69 80 63 70 
Histology, % 
    Diffuse large cell or variants 72 72 81 100 74 
    Burkitt or Burkitt-like 28 26 26 
    PBL of the oral cavity type 
IPI at least 2, % 57 48 58 64 69 
Median CD4/dL 161 172 128 152 181 
Complete remission rate, % 70 77 57 66 69 
Febrile neutropenia, % 31 25 32 NA 16 
Deaths from infections, % 11 10 
R-CDE141 R-CHOP143 R-CHOP144 R-CHOP145 R-EPOCH146 
No. of patients 74 61 95 60 51 
Stage III-IV (%) 70 69 80 63 70 
Histology, % 
    Diffuse large cell or variants 72 72 81 100 74 
    Burkitt or Burkitt-like 28 26 26 
    PBL of the oral cavity type 
IPI at least 2, % 57 48 58 64 69 
Median CD4/dL 161 172 128 152 181 
Complete remission rate, % 70 77 57 66 69 
Febrile neutropenia, % 31 25 32 NA 16 
Deaths from infections, % 11 10 

PBL indicates plasmablastic lymphoma; R-CDE, rituximab–cyclophosphamide, doxorubicin, etoposide; R-CHOP, rituximab–cyclophosphamide, vincristine, doxorubicin, prednisone; and R-EPOCH, rituximab–cyclophosphamide, doxorubicin, etoposide, vincristine, prednisone.

Current treatment of BL and DLBCL in the HAART era

In the HAART era the treatment of BL remains a big challenge. In fact, HAART has improved the NHL course, with the only exception being BL, which has turned out to be clinically more aggressive than DLBCL. This is related to the strong positive effect HAART has on the outcome of DLBCL, whereas the outcome for BL is unchanged.147,148  Since this lymphoma subtype affects survival, a question has arisen whether it should be treated more aggressively. A retrospective analysis has been conducted on the feasibility of intensive aggressive chemotherapy regimens, which are usually used in the treatment of BL in the general population, and also in HIV patients. American and Spanish investigators report a 63% to 68% CR rate, a 46% to 60% failure-free survival rate at 2 years, and the same toxicity as in the general population, which confirms the feasibility of aggressive regimens also in the HIV setting.149-151 

Treatment of unusual entities

Few data have been reported on the treatment of other rare entities, that is, PEL and PBL of the oral cavity type. The classic PEL presents in advanced course of HIV infection with a typical abundant effusion. All published data showed that despite the use of standard chemotherapy regimens (CHOP or CHOP-like) the prognosis is poor in comparison with that of other HIV-NHL subtypes.6,46,152  Considering the aggressiveness of PEL and its pathogenesis, several nonchemotherapy approaches have been tested, including HAART alone or antiviral treatment (ie, cidofovir, AZT, IFN) with promising results.153-155 

Similarly to PEL, the prognosis of PBLs of the oral cavity type remains poor despite the use of HAART and chemotherapy.156,157 

Lymphoma progression is the leading cause of death in 35% to 55% of the patients with HIV-NHL receiving chemotherapy, of whom approximately half need second-line chemotherapy following progression or relapse of the disease. To date, the results achieved by salvage therapies that do not include a standard high-dose chemotherapy regimen with peripheral blood stem cell (PBSC) transplantation have been very frustrating (median survival, 2–4 months).158  With the introduction of HAART into clinical practice, more aggressive treatment protocols can be taken into consideration, whose effectiveness has already been documented in HIV-negative patients. Preliminary studies support the feasibility of high-dose chemotherapy in combination with PBSC transplantation in HIV-NHL, proving that PBSC collections are adequate, anchoring rates are similar to those recorded in HIV-negative patients, and high-dose chemotherapy is well tolerated with no increase in the incidence of OIs.159,160  Within the GICAT, a study has been performed on a group of patients with NHL or refractory or recurred HL: the results support the feasibility of an adequate peripheral blood stem cell collection, with no transplantation-related mortality and a very good outcome—60% of the patients being alive and disease-free.161  The long-term follow-up of patients who underwent high-dose chemotherapy and PBSC support confirms that the outcome of HIV patients with chemosensitive disease is superimposable to that of HIV-negative patients.162 

It can be drawn from the above data that prognosis for patients with lymphomas and concomitant HIV infection could be improved using better combined chemotherapy protocols incorporating anticancer treatments and antiretroviral drugs. We envision that in the future, therapies will be developed that target specific viral oncogenes to which the lymphoma cells are addicted (ie, EBV LMP1, KSHV vFLIP), and that these will provide therapeutic benefit. For the time being, the administration of HAART during chemotherapy can improve control of the underlying HIV infection. The inclusion of hematopoietic growth factors in the treatment of this patient group makes it possible to increase chemotherapy doses and prolong the administration of antiretroviral drugs with the intent to improve survival. At the present time, we strongly recommend that patients with lymphoma and HIV infection should be treated as patients with lymphoma of the general population.

This work was supported in part by a grant from the Ministero della Salute (Rome, Italy) within the framework of the Progetto Integrato Oncologia-Advanced Molecular Diagnostics project (RFPS-2006-2-342010.7; A.C.), and by the European Union Integrated project INCA (LSHC-CT-2005-18704; T.F.S.).

Contribution: A.C. designed the review; and all authors contributed to writing and proofreading of the paper.

Conflict-of-interest disclosure: The authors declare no competing financial interests.

Correspondence: Antonino Carbone, Chairman of the Department of Pathology, National Cancer Institute, via Venezian, 1, 20133 Milan, Italy; e-mail: antonino.carbone@istitutotumori.mi.it.

1
Young
 
LS
Rickinson
 
AB
Epstein-Barr virus: 40 years on.
Nat Rev Cancer
2004
, vol. 
4
 (pg. 
757
-
768
)
2
IARC Monographs on the Evaluation of Carcinogenic Risks to Humans. Human immunodeficiency viruses and human T-cell lymphotrophic viruses.
1996
, vol. 
Vol 67
 
Lyon, France
International Agency for Research on Cancer/World Health Organization
3
Elgui de Oliveira
 
D
DNA viruses in human cancer: an integrated overview on fundamental mechanisms of viral carcinogenesis.
Cancer Lett
2007
, vol. 
247
 (pg. 
182
-
196
)
4
Delecluse
 
HJ
Feederle
 
R
O'Sullivan
 
B
Taniere
 
P
Epstein Barr virus-associated tumours: an update for the attention of the working pathologist.
J Clin Pathol
2007
, vol. 
60
 (pg. 
1358
-
1364
)
5
Du
 
MQ
Bacon
 
CM
Isaacson
 
PG
Kaposi sarcoma-associated herpesvirus/human herpesvirus 8 and lymphoproliferative disorders.
J Clin Pathol
2007
, vol. 
60
 (pg. 
1350
-
1357
)
6
Chen
 
YB
Rahemtullah
 
A
Hochberg
 
E
Primary effusion lymphoma.
Oncologist
2007
, vol. 
12
 (pg. 
569
-
576
)
7
Schulz
 
TF
Epidemiology of Kaposi's sarcoma-associated herpesvirus/human herpesvirus 8.
Adv Cancer Res
1999
, vol. 
76
 (pg. 
121
-
160
)
8
Chang
 
Y
Cesarman
 
E
Pessin
 
MS
, et al. 
Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi's sarcoma.
Science
1994
, vol. 
226
 (pg. 
1865
-
1869
)
9
Dotti
 
G
Fiocchi
 
R
Motta
 
T
, et al. 
Primary effusion lymphoma after heart transplantation: a new entity associated with human herpesvirus-8.
Leukemia
1999
, vol. 
13
 (pg. 
664
-
670
)
10
Jones
 
D
Ballestas
 
M
Kaye
 
KM
, et al. 
Primary-effusion lymphoma an Kaposi's sarcoma in a cardiac-transplant recipient.
N Eng J Med
1998
, vol. 
339
 (pg. 
444
-
449
)
11
Raphael
 
M
Borisch
 
B
Jaffe
 
ES
Jaffe
 
ES
Harris
 
NL
Stein
 
H
Vardiman
 
JW
Lymphomas associated with infection by the human immune deficiency virus (HIV).
World Health Organization Classification of Tumours, Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues
2001
Lyon, France
IARC Press
(pg. 
260
-
263
)
12
Cesarman
 
E
Chang
 
Y
Moore
 
PS
Said
 
JW
Knowles
 
DM
Kaposi's sarcoma-associated herpesvirus-like DNA sequences are present in AIDS-related body cavity-based lymphoma.
New Engl J Med
1995
, vol. 
332
 (pg. 
1186
-
1191
)
13
Chadburn
 
A
Hyjek
 
E
Mathew
 
S
Cesarman
 
E
Said
 
J
Knowles
 
DM
KSHV-positive solid lymphomas represent an extra-cavitary variant of primary effusion lymphoma.
Am J Surg Pathol
2004
, vol. 
28
 (pg. 
1401
-
1416
)
14
Carbone
 
A
Gloghini
 
A
Vaccher
 
E
, et al. 
Kaposi's sarcoma-associated herpesvirus/human herpesvirus type 8-positive solid lymphomas: a tissue-based variant of primary effusion lymphoma.
J Mol Diagn
2005
, vol. 
7
 (pg. 
17
-
27
)
15
Delecluse
 
HJ
Anagnostopoulos
 
I
Dallenbach
 
F
, et al. 
Plasmablastic lymphomas of the oral cavity: a new entity associated with the human immunodeficiency virus infection.
Blood
1997
, vol. 
89
 (pg. 
1413
-
1420
)
16
Carbone
 
A
Gaidano
 
G
Gloghini
 
A
Ferlito
 
A
Rinaldo
 
A
Stein
 
H
AIDS-related plasmablastic lymphomas of the oral cavity and jaws: a diagnostic dilemma.
Ann Otol Rhinol Laryngol
1999
, vol. 
108
 (pg. 
95
-
99
)
17
Dupin
 
N
Diss
 
TL
Kellam
 
P
, et al. 
HHV-8 is associated with a plasmablastic variant of Castleman disease that is linked to HHV-8-positive plasmablastic lymphoma.
Blood
2000
, vol. 
95
 (pg. 
1406
-
1412
)
18
Swerdlow
 
SH
Campo
 
E
Harris
 
NL
, et al. 
World Health Organization Classification of Tumours, Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues.
2008
Lyon, France
IARC Press
19
Cesarman
 
E
Knowles
 
DM
The role of Kaposi's sarcoma-associated herpesvirus (KSHV/HHV-8) in lymphoproliferative diseases.
Semin Cancer Biol
1999
, vol. 
9
 (pg. 
165
-
174
)
20
Carbone
 
A
Gloghini
 
A
Larocca
 
LM
, et al. 
Expression profile of MUM1/IRF4, BCL-6, and CD138/syndecan-1 defines novel histogenetic subsets of human immunodeficiency virus-related lymphomas.
Blood
2001
, vol. 
97
 (pg. 
744
-
751
)
21
Klein
 
U
Gloghini
 
A
Gaidano
 
G
, et al. 
Gene expression profile analysis of AIDS-related primary effusion lymphoma (PEL) suggests a plasmablastic derivation and identifies PEL-specific transcripts.
Blood
2003
, vol. 
101
 (pg. 
4115
-
4121
)
22
Biggar
 
RJ
Jaffe
 
ES
Goedert
 
JJ
Chaturvedi
 
A
Pfeiffer
 
R
Engels
 
EA
Hodgkin lymphoma and immunodeficiency in persons with HIV/AIDS.
Blood
2006
, vol. 
108
 (pg. 
3786
-
3791
)
23
Grogg
 
KL
Miller
 
RF
Dogan
 
A
HIV infection and lymphoma.
J Clin Pathol
2007
, vol. 
60
 (pg. 
1365
-
1372
)
24
Tirelli
 
U
Errante
 
D
Dolcetti
 
R
, et al. 
Hodgkin's disease and human immunodeficiency virus infection: clinicopathologic and virologic features of 114 patients from the Italian Cooperative Group on AIDS and Tumors.
J Clin Oncol
1995
, vol. 
13
 (pg. 
1758
-
1767
)
25
Carbone
 
A
Gloghini
 
A
Larocca
 
LM
, et al. 
Human immunodeficiency virus-associated Hodgkin's disease derives from post-germinal center B cells.
Blood
1999
, vol. 
93
 (pg. 
2319
-
2326
)
26
Rezk
 
SA
Weiss
 
LM
Epstein-Barr virus-associated lymphoproliferative disorders.
Hum Pathol
2007
, vol. 
38
 (pg. 
1293
-
1304
)
27
Said
 
JW
Immunodeficiency-related Hodgkin lymphoma and its mimics.
Adv Anat Pathol
2007
, vol. 
14
 (pg. 
189
-
194
)
28
Carbone
 
A
Gloghini
 
A
Dotti
 
G
EBV-associated lymphoproliferative disorders: classification and treatment.
Oncologist
2008
, vol. 
13
 (pg. 
577
-
585
)
29
Knowles
 
DM
Inghirami
 
G
Ubriaco
 
A
Dalla-Favera
 
R
Molecular genetic analysis of three AIDS-associated neoplasms of uncertain lineage demonstrates their B-cell derivation and the possible pathogenetic role of the Epstein-Barr virus.
Blood
1989
, vol. 
73
 (pg. 
792
-
799
)
30
Walts
 
AE
Shintaku
 
IP
Said
 
JW
Diagnosis of malignant lymphoma in effusions from patients with AIDS by gene rearrangement.
Am J Clin Path
1990
, vol. 
94
 (pg. 
170
-
175
)
31
Ansari
 
MQ
Dawson
 
DB
Nador
 
R
, et al. 
Primary body-cavity based AIDS-related lymphomas.
Am J Clin Path
1996
, vol. 
105
 (pg. 
221
-
229
)
32
Nador
 
RG
Cesarman
 
E
Chadburn
 
A
, et al. 
Primary effusion lymphoma: a distinct clinicopathologic entity associated with the Kaposi's sarcoma-associated herpesvirus.
Blood
1996
, vol. 
88
 (pg. 
645
-
656
)
33
Beaty
 
MW
Kumar
 
S
Sorbara
 
L
Miller
 
K
Raffeld
 
M
Jaffe
 
ES
A biophenotypic human herpesvirus 8–associated primary bowel lymphoma.
Am J Surg Pathol
1999
, vol. 
23
 (pg. 
992
-
994
)
34
Boulanger
 
E
Hermine
 
O
Fermand
 
JP
, et al. 
Human herpesvirus 8 (HHV-8)-associated peritoneal primary effusion lymphoma (PEL) in two HIV-negative elderly patients.
Am J Hematol
2004
, vol. 
76
 (pg. 
88
-
91
)
35
Said
 
JW
Shintaku
 
IP
Asou
 
H
, et al. 
Herpesvirus 8 inclusions in primary effusion lymphoma: report of a unique case with T-cell phenotype.
Arch Pathol Lab Med
1999
, vol. 
123
 (pg. 
257
-
260
)
36
Engels
 
EA
Pittaluga
 
S
Whitby
 
D
, et al. 
Immunoblastic lymphoma in persons with AIDS-associated Kaposi's sarcoma: a role for Kaposi's sarcoma-associated herpesvirus.
Mod Pathol
2003
, vol. 
16
 (pg. 
424
-
429
)
37
Gaidano
 
G
Capello
 
D
Cilia
 
AM
, et al. 
Genetic characterization of HHV-8/KSHV-positive primary effusion lymphoma reveals frequent mutations of BCL6: implications for disease pathogenesis and histogenesis.
Genes Chromosomes Cancer
1999
, vol. 
24
 (pg. 
16
-
23
)
38
Matolcsy
 
A
Nador
 
RG
Cesarman
 
E
Knowles
 
DM
Immunoglobulin VH gene mutational analysis suggests that primary effusion lymphomas derive from different stages of B cell maturation.
Am J Pathol
1998
, vol. 
153
 (pg. 
1609
-
1614
)
39
Jenner
 
RG
Maillard
 
K
Cattini
 
N
Kaposi's sarcoma-associated herpesvirus-infected primary effusion lymphoma has a plasma cell gene expression profile.
Proc Natl Acad Sci U S A
2003
, vol. 
100
 (pg. 
10399
-
10404
)
40
Das
 
DK
Serous effusion in malignant lymphomas: a review.
Diagn Cytopathol
2006
, vol. 
34
 (pg. 
335
-
347
)
41
Carbone
 
A
Gloghini
 
A
PEL and HHV8-unrelated effusion lymphomas: classification and diagnosis.
Cancer
2008
, vol. 
114
 (pg. 
225
-
227
)
42
Cesarman
 
E
Nador
 
RG
Aozasa
 
K
Delsol
 
G
Said
 
JW
Knowles
 
DM
Kaposi's sarcoma-associated herpesvirus in non-AIDS related lymphomas occurring in body cavities.
Am J Pathol
1996
, vol. 
149
 (pg. 
53
-
57
)
43
Carbone
 
A
Gloghini
 
A
Vaccher
 
E
, et al. 
Kaposi's sarcoma-associated herpes virus DNA sequenze in AIDS-related and AIDS-unrelated lymphomatous effusions.
Br J Haematol
1996
, vol. 
94
 (pg. 
533
-
543
)
44
Gaidano
 
G
Carbone
 
A
Primary effusion lymphoma: a liquid phase lymphoma of fluid-filled body cavities.
Adv Cancer Res
2001
, vol. 
80
 (pg. 
115
-
146
)
45
Kobayashi
 
Y
Kamitsuji
 
Y
Kuroda
 
J
, et al. 
Comparison of human herpes virus 8 related primary effusion lymphoma with human herpes virus 8 unrelated primary effusion lymphoma-like lymphoma on the basis of HIV: report of 2 cases and review of 212 cases in the literature.
Acta Haematol
2007
, vol. 
117
 (pg. 
132
-
144
)
46
Matsumoto
 
Y
Nomura
 
K
Ueda
 
K
, et al. 
Human herpesvirus 8-negative malignant effusion lymphoma. A distinct clinical entity and successful treatment with rituximab.
Leuk Lymphoma
2005
, vol. 
46
 (pg. 
415
-
419
)
47
Simonelli
 
C
Spina
 
M
Cinelli
 
R
, et al. 
Clinical features and out come of primary effusion lymphoma in HIV-infected patients: a single-institution study.
J Clin Oncol
2003
, vol. 
21
 (pg. 
3948
-
3954
)
48
Castleman
 
B
Iverson
 
L
Menendez
 
VP
Localized mediastinal lymphnode hyperplasia resembling thymoma.
Cancer
1956
, vol. 
9
 (pg. 
822
-
830
)
49
Ballon
 
G
Cesarman
 
E
Volberding
 
PA
Palefsky
 
J
Castleman's Disease.
Viral and Immunolgogical Malignancies
2006
Hamilton, ON
BC Decker
(pg. 
108
-
121
)
50
Soulier
 
J
Grollet
 
L
Oksenhendler
 
E
, et al. 
Kaposi's sarcoma-associated herpesvirus-like DNA sequences in multicentric Castleman's disease.
Blood
1995
, vol. 
86
 (pg. 
1275
-
1280
)
51
Frizzera
 
G
Banks
 
PM
Massarelli
 
G
Rosai
 
J
A systemic lymphoproliferative disorder with morphologic features of Castleman's disease: pathological findings in 15 patients.
Am J Surg Pathol
1983
, vol. 
7
 (pg. 
211
-
231
)
52
Oksenhendler
 
E
Boulanger
 
E
Galicier
 
L
, et al. 
High incidence of Kaposi sarcoma-associated herpesvirus-related non-Hodgkin lymphoma in patients with HIV infection and multicentric Castleman disease.
Blood
2002
, vol. 
99
 (pg. 
2331
-
2336
)
53
Judde
 
JG
Lacoste
 
V
Briere
 
J
, et al. 
Monoclonality or oligoclonality of human herpesvirus 8 terminal repeat sequences in Kaposi's sarcoma and other diseases.
J Natl Cancer Inst
2000
, vol. 
92
 (pg. 
729
-
736
)
54
Parravicini
 
C
Chandran
 
B
Corbellino
 
M
, et al. 
Differential viral protein expression in Kaposi's sarcoma-associated herpesvirus-infected diseases: Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease.
Am J Pathol
2000
, vol. 
156
 (pg. 
743
-
749
)
55
Amin
 
HM
Medeiros
 
LJ
Manning
 
JT
Jones
 
D
Dissolution of the lymphoid follicle is a feature of the HHV8+ variant of plasma cell Castleman's disease.
Am J Surg Pathol
2003
, vol. 
27
 (pg. 
91
-
100
)
56
Du
 
MQ
Liu
 
H
Diss
 
TC
, et al. 
Kaposi sarcoma-associated herpesvirus infects monotypic (IgM lambda) but polyclonal naive B cells in Castleman disease and associated lymphoproliferative disorders.
Blood
2001
, vol. 
97
 (pg. 
2130
-
2136
)
57
Gould
 
SJ
Diss
 
T
Isaacson
 
PG
Multicentric Castleman's disease in association with a solitary plasmacytoma: a case report.
Histopathol
1990
, vol. 
17
 (pg. 
135
-
140
)
58
Dong
 
HY
Scadden
 
DT
de Leval
 
L
Tang
 
Z
Isaacson
 
PG
Harris
 
NL
Plasmablastic lymphoma in HIV-positive patients: an aggressive Epstein-Barr virus-associated extramedullary plasmacytic neoplasm.
Am J Surg Pathol
2005
, vol. 
29
 (pg. 
1633
-
1641
)
59
Hamilton-Dutoit
 
SJ
Rea
 
D
Raphael
 
M
, et al. 
Epstein-Barr virus-latent gene expression and tumor cell phenotype in acquired immunodeficiency syndrome-related non-Hodgkin's lymphoma: correlation of lymphoma phenotype with three distinct patterns of viral latency.
Am J Pathol
1993
, vol. 
143
 (pg. 
1072
-
1085
)
60
Carbone
 
A
Tirelli
 
U
Gloghini
 
A
Volpe
 
R
Boiocchi
 
M
Human immunodeficiency virus-associated systemic lymphomas may be subdivided into two main groups according to Epstein-Barr viral latent gene expression.
J Clin Oncol
1993
, vol. 
11
 (pg. 
1674
-
1681
)
61
Young
 
LS
Rickinson
 
AB
Epstein-Barr virus: 40 years on.
Nat Rev Cancer
2004
, vol. 
4
 (pg. 
757
-
768
)
62
Wang
 
D
Liebowitz
 
D
Kieff
 
E
An EBV membrane protein expressed in immortalized lymphocytes transforms established rodent cells.
Cell
1985
, vol. 
43
 (pg. 
831
-
840
)
63
Kulwichit
 
W
Edwards
 
RH
Davenport
 
EM
Baskar
 
JF
Godfrey
 
V
Raab-Traub
 
N
Expression of the Epstein-Barr virus latent membrane protein 1 induces B cell lymphoma in transgenic mice.
Proc Natl Acad Sci U S A
1998
, vol. 
95
 (pg. 
11963
-
11968
)
64
Dirmeier
 
U
Neuhierl
 
B
Kilger
 
E
Reisbach
 
G
Sandberg
 
ML
Hammerschmidt
 
W
Latent membrane protein 1 is critical for efficient growth transformation of human B cells by Epstein-Barr virus.
Cancer Res
2003
, vol. 
63
 (pg. 
2982
-
2989
)
65
Mosialos
 
G
Birkenbach
 
M
Yalamanchili
 
R
VanArsdale
 
T
Ware
 
C
Kieff
 
E
The Epstein-Barr virus transforming protein LMP1 engages signaling proteins for the tumor necrosis factor receptor family.
Cell
1995
, vol. 
80
 (pg. 
389
-
399
)
66
Eliopoulos
 
AG
Young
 
LS
Activation of the cJun N-terminal kinase (JNK) pathway by the Epstein-Barr virus-encoded latent membrane protein 1 (LMP1).
Oncogene
1998
, vol. 
16
 (pg. 
1731
-
1742
)
67
Eliopoulos
 
AG
Gallagher
 
NJ
Blake
 
SM
Dawson
 
CW
Young
 
LS
Activation of the p38 mitogen-activated protein kinase pathway by Epstein-Barr virus-encoded latent membrane protein 1 coregulates interleukin-6 and interleukin-8 production.
J Biol Chem
1999
, vol. 
274
 (pg. 
16085
-
16096
)
68
Huen
 
DS
Henderson
 
SA
Croom-Carter
 
D
Rowe
 
M
The Epstein-Barr virus latent membrane protein-1 (LMP1) mediates activation of NF-kappa B and cell surface phenotype via two effector regions in its carboxy-terminal cytoplasmic domain.
Oncogene
1995
, vol. 
10
 (pg. 
549
-
560
)
69
Devergne
 
O
Hummel
 
M
Koeppen
 
H
, et al. 
A novel interleukin-12 p40-related protein induced by latent Epstein-Barr virus infection in B lymphocytes.
J Virol
1996
, vol. 
70
 (pg. 
1143
-
1153
)
70
Izumi
 
KM
Kaye
 
KM
Kieff
 
ED
The Epstein-Barr virus LMP1 amino acid sequence that engages tumor necrosis factor receptor associated factors is critical for primary B lymphocyte growth transformation.
Proc Natl Acad Sci U S A
1997
, vol. 
94
 (pg. 
1447
-
1452
)
71
Gloghini
 
A
Gaidano
 
G
Larocca
 
LM
, et al. 
Expression of cyclin-dependent kinase inhibitor p27(Kip1) in AIDS-related diffuse large-cell lymphomas is associated with Epstein-Barr virus-encoded latent membrane protein 1.
Am J Pathol
2002
, vol. 
161
 (pg. 
163
-
171
)
72
Brinkmann
 
MM
Schulz
 
TF
Regulation of intracellular signalling by the terminal membrane proteins of members of the Gammaherpesvirinae.
J Gen Virol
2006
, vol. 
87
 (pg. 
1047
-
1074
)
73
Gaidano
 
G
Carbone
 
A
Dalla-Favera
 
R
Pathogenesis of AIDS-related lymphomas: molecular and histogenetic heterogeneity.
Am J Pathol
1998
, vol. 
152
 (pg. 
623
-
630
)
74
Guasparri
 
I
Bubman
 
D
Cesarman
 
E
EBV LMP2A affects LMP1-mediated NF-kappaB signaling and survival of lymphoma cells by regulating TRAF2 expression.
Blood
2008
, vol. 
111
 (pg. 
3813
-
3820
)
75
Rowe
 
M
Young
 
LS
Crocker
 
J
Stokes
 
H
Henderson
 
S
Rickinson
 
AB
Epstein-Barr virus (EBV)-associated lymphoproliferative disease in the SCID mouse model: implications for the pathogenesis of EBV-positive lymphomas in man.
J Exp Med
1991
, vol. 
173
 (pg. 
147
-
158
)
76
Wilson
 
JB
Bell
 
JL
Levine
 
AJ
Expression of Epstein-Barr virus nuclear antigen-1 induces B cell neoplasia in transgenic mice.
EMBO J
1996
, vol. 
15
 (pg. 
3117
-
3126
)
77
Drotar
 
ME
Silva
 
S
Barone
 
E
, et al. 
Epstein-Barr virus nuclear antigen-1 and Myc cooperate in lymphomagenesis.
Int J Cancer
2003
, vol. 
106
 (pg. 
388
-
395
)
78
Kamranvar
 
SA
Gruhne
 
B
Szeles
 
A
Masucci
 
MG
Epstein-Barr virus promotes genomic instability in Burkitt's lymphoma.
Oncogene
2007
, vol. 
26
 (pg. 
5115
-
5123
)
79
Kelly
 
GL
Milner
 
AE
Baldwin
 
GS
Bell
 
AI
Rickinson
 
AB
Three restricted forms of Epstein-Barr virus latency counteracting apoptosis in c-myc-expressing Burkitt lymphoma cells.
Proc Natl Acad Sci U S A
2006
, vol. 
103
 (pg. 
14935
-
14940
)
80
Altmann
 
M
Hammerschmidt
 
W
Epstein-Barr virus provides a new paradigm: a requirement for the immediate inhibition of apoptosis.
PLoS Biol
2005
, vol. 
3
 pg. 
e404
 
81
Young
 
LS
Murray
 
PG
Epstein-Barr virus and oncogenesis: from latent genes to tumours.
Oncogene
2003
, vol. 
22
 (pg. 
5108
-
5121
)
82
Küppers
 
R
Rajewsky
 
K
Zhao
 
M
, et al. 
Hodgkin disease: Hodgkin and Reed-Sternberg cells picked from histological sections show clonal immunoglobulin gene rearrangements and appear to be derived from B cells at various stages of development.
Proc Natl Acad Sci U S A
1994
, vol. 
91
 (pg. 
10962
-
10966
)
83
Kanzler
 
H
Küppers
 
R
Hansmann
 
ML
Rajewsky
 
K
Hodgkin and Reed-Sternberg cells in Hodgkin's disease represent the outgrowth of a dominant tumor clone derived from (crippled) germinal center B cells.
J Exp Med
1996
, vol. 
184
 (pg. 
1495
-
1505
)
84
Bräuninger
 
A
Schmitz
 
R
Bechtel
 
D
Renné
 
C
Hansmann
 
ML
Küppers
 
R
Molecular biology of Hodgkin's and Reed/Sternberg cells in Hodgkin's lymphoma.
Int J Cancer
2006
, vol. 
118
 (pg. 
1853
-
1861
)
85
Caldwell
 
RG
Wilson
 
JB
Anderson
 
SJ
Longnecker
 
R
Epstein-Barr virus LMP2A drives B cell development and survival in the absence of normal B cell receptor signals.
Immunity
1998
, vol. 
9
 (pg. 
405
-
411
)
86
Portis
 
T
Dyck
 
P
Longnecker
 
R
Epstein-Barr Virus (EBV) LMP2A induces alterations in gene transcription similar to those observed in Reed-Sternberg cells of Hodgkin lymphoma.
Blood
2003
, vol. 
102
 (pg. 
4166
-
4178
)
87
Mancao
 
C
Altmann
 
M
Jungnickel
 
B
Hammerschmidt
 
W
Rescue of “crippled” germinal center B cells from apoptosis by Epstein-Barr virus.
Blood
2005
, vol. 
106
 (pg. 
4339
-
4344
)
88
Mancao
 
C
Hammerschmidt
 
W
Epstein-Barr virus latent membrane protein 2A is a B-cell receptor mimic and essential for B-cell survival.
Blood
2007
, vol. 
110
 (pg. 
3715
-
3721
)
89
Vockerodt
 
M
Morgan
 
S
Kuo
 
M
, et al. 
The Epstein-Barr virus oncoprotein, latent membrane protein-1, reprograms germinal centre B cells towards a Hodgkin's Reed-Sternberg-like phenotype.
J Pathol
2008
, vol. 
216
 (pg. 
83
-
92
)
90
Dutton
 
A
Woodman
 
CB
Chukwuma
 
MB
, et al. 
Bmi-1 is induced by the Epstein-Barr virus oncogene LMP1 and regulates the expression of viral target genes in Hodgkin lymphoma cells.
Blood
2007
, vol. 
109
 (pg. 
2597
-
2603
)
91
Baumforth
 
KR
Birgersdotter
 
A
Reynolds
 
GM
, et al. 
Expression of the Epstein-Barr virus-encoded Epstein-Barr virus nuclear antigen 1 in Hodgkin's lymphoma cells mediates up-regulation of CCL20 and the migration of regulatory T cells.
Am J Pathol
2008
, vol. 
173
 (pg. 
195
-
204
)
92
Flavell
 
JR
Baumforth
 
KR
Wood
 
VH
, et al. 
Down-regulation of the TGF-beta target gene, PTPRK, by the Epstein-Barr virus encoded EBNA1 contributes to the growth and survival of Hodgkin lymphoma cells.
Blood
2008
, vol. 
111
 (pg. 
292
-
301
)
93
Lallemand
 
F
Desire
 
N
Rozenbaum
 
W
Nicolas
 
JC
Marechal
 
V
Quantitative analysis of human herpesvirus 8 viral load using a real-time PCR assay.
J Clin Microbiol
2000
, vol. 
38
 (pg. 
1404
-
1408
)
94
Chang
 
Y
Moore
 
PS
Talbot
 
SJ
, et al. 
Cyclin encoded by KS herpesvirus.
Nature
1996
, vol. 
382
 pg. 
410
 
95
Russo
 
JJ
Bohenzky
 
RA
Chien
 
MC
, et al. 
Nucleotide sequence of the Kaposi sarcoma-associated herpesvirus (HHV8).
Proc Natl Acad Sci U S A
1996
, vol. 
93
 (pg. 
14862
-
14867
)
96
Rainbow
 
L
Platt
 
GM
Simpson
 
GR
, et al. 
The 222- to 234-kilodalton latent nuclear protein (LNA) of Kaposi's sarcoma-associated herpesvirus (human herpesvirus 8) is encoded by orf73 and is a component of the latency-associated nuclear antigen.
J Virol
1997
, vol. 
71
 (pg. 
5915
-
5921
)
97
Rivas
 
C
Thlick
 
AE
Parravicini
 
C
Moore
 
PS
Chang
 
Y
Kaposi's sarcoma-associated herpesvirus LANA2 is a B-cell-specific latent viral protein that inhibits p53.
J Virol
2001
, vol. 
75
 (pg. 
429
-
438
)
98
Cai
 
X
Lu
 
S
Zhang
 
Z
Gonzalez
 
CM
Damania
 
B
Cullen
 
BR
Kaposi's sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells.
Proc Natl Acad Sci U S A
2005
, vol. 
102
 (pg. 
5570
-
5575
)
99
Pfeffer
 
S
Sewer
 
A
Lagos-Quintana
 
M
, et al. 
Identification of microRNAs of the herpesvirus family.
Nat Methods
2005
, vol. 
2
 (pg. 
269
-
276
)
100
Samols
 
MA
Hu
 
J
Skalsky
 
RL
Renne
 
R
Cloning and identification of a microRNA cluster within the latency-associated region of Kaposi's sarcoma-associated herpesvirus.
J Virol
2005
, vol. 
79
 (pg. 
9301
-
9305
)
101
Schulz
 
TF
The pleiotropic effects of Kaposi's sarcoma herpesvirus.
J Pathol
2006
, vol. 
208
 (pg. 
187
-
198
)
102
Schulz
 
TF
KSHV/HHV8-associated lymphoproliferations in the AIDS setting.
Eur J Cancer
2001
, vol. 
37
 (pg. 
1217
-
1226
)
103
Katano
 
H
Sato
 
Y
Kurata
 
T
Mori
 
S
Sata
 
T
Expression and localization of human herpesvirus 8-encoded proteins in primary effusion lymphoma, Kaposi's sarcoma, and multicentric Castleman's disease.
Virology
2000
, vol. 
269
 (pg. 
335
-
344
)
104
Moore
 
PS
Chang
 
Y
Molecular virology of Kaposi's sarcoma-associated herpesvirus.
Philos Trans R Soc Lond B Biol Sci
2001
, vol. 
356
 (pg. 
499
-
516
)
105
Cesarman
 
E
Mesri
 
EA
Kaposi sarcoma-associated herpesvirus and other viruses in human lymphomagenesis.
Curr Top Microbiol Immunol
2007
, vol. 
312
 (pg. 
263
-
287
)
106
Guasparri
 
I
Keller
 
SA
Cesarman
 
E
KSHV vFLIP is essential for the survival of infected lymphoma cells.
J Exp Med
2004
, vol. 
199
 (pg. 
993
-
1003
)
107
Godfrey
 
A
Anderson
 
J
Papanastasiou
 
A
Takeuchi
 
Y
Boshoff
 
C
Inhibiting primary effusion lymphoma by lentiviral vectors encoding short hairpin RNA.
Blood
2005
, vol. 
105
 (pg. 
2510
-
2518
)
108
Chaudhary
 
PM
Jasmin
 
A
Eby
 
MT
Hood
 
L
Modulation of the NF-kappa B pathway by virally encoded death effector domains-containing proteins.
Oncogene
1999
, vol. 
18
 (pg. 
5738
-
5746
)
109
Liu
 
L
Eby
 
MT
Rathore
 
N
Sinha
 
SK
Kumar
 
A
Chaudhary
 
PM
The human herpes virus 8-encoded viral FLICE inhibitory protein physically associates with and persistently activates the Ikappa B kinase complex.
J Biol Chem
2002
, vol. 
277
 (pg. 
13745
-
13751
)
110
Field
 
N
Low
 
W
Daniels
 
M
, et al. 
KSHV vFLIP binds to IKK-gamma to activate IKK.
J Cell Sci
2003
, vol. 
116
 (pg. 
3721
-
3728
)
111
An
 
J
Sun
 
Y
Sun
 
R
Rettig
 
MB
Kaposi's sarcoma-associated herpesvirus encoded vFLIP induces cellular IL-6 expression: the role of the NF-kappaB and JNK/AP1 pathways.
Oncogene
2003
, vol. 
22
 (pg. 
3371
-
3385
)
112
Keller
 
SA
Schattner
 
EJ
Cesarman
 
E
Inhibition of NF-kappaB induces apoptosis of KSHV-infected primary effusion lymphoma cells.
Blood
2000
, vol. 
96
 (pg. 
2537
-
2542
)
113
Sarek
 
G
Järviluoma
 
A
Ojala
 
PM
KSHV viral cyclin inactivates p27KIP1 through Ser10 and Thr187 phosphorylation in proliferating primary effusion lymphomas.
Blood
2006
, vol. 
107
 (pg. 
725
-
732
)
114
Järviluoma
 
A
Child
 
ES
Sarek
 
G
, et al. 
Phosphorylation of the cyclin-dependent kinase inhibitor p21Cip1 on serine 130 is essential for viral cyclin-mediated bypass of a p21Cip1-imposed G1 arrest.
Mol Cell Biol
2006
, vol. 
26
 (pg. 
2430
-
2440
)
115
Ojala
 
PM
Tiainen
 
M
Salven
 
P
, et al. 
Kaposi's sarcoma-associated herpesvirus-encoded v-cyclin triggers apoptosis in cells with high levels of cyclin-dependent kinase 6.
Cancer Res
1999
, vol. 
59
 (pg. 
4984
-
4989
)
116
Koopal
 
S
Furuhjelm
 
JH
Järviluoma
 
A
, et al. 
Viral oncogene-induced DNA damage response is activated in Kaposi sarcoma tumorigenesis.
PLoS Pathog
2007
, vol. 
3
 (pg. 
1348
-
1360
)
117
Friborg
 
J
Kong
 
W
Hottiger
 
MO
Nabel
 
GJ
p53 inhibition by the LANA protein of KSHV protects against cell death.
Nature
1999
, vol. 
402
 (pg. 
889
-
894
)
118
Radkov
 
SA
Kellam
 
P
Boshoff
 
C
The latent nuclear antigen of Kaposi sarcoma-associated herpesvirus targets the retinoblastoma-E2F pathway and with the oncogene Hras transforms primary rat cells.
Nat Med
2000
, vol. 
6
 (pg. 
1121
-
1127
)
119
Liu
 
J
Martin
 
HJ
Liao
 
G
Hayward
 
SD
The Kaposi's sarcoma-associated herpesvirus LANA protein stabilizes and activates c-Myc.
J Virol
2007
, vol. 
81
 (pg. 
10451
-
10459
)
120
Bubman
 
D
Guasparri
 
I
Cesarman
 
E
Deregulation of c-Myc in primary effusion lymphoma by Kaposi's sarcoma herpesvirus latency-associated nuclear antigen.
Oncogene
2007
, vol. 
26
 (pg. 
4979
-
4986
)
121
Sarek
 
G
Kurki
 
S
Enbäck
 
J
, et al. 
Reactivation of the p53 pathway as a treatment modality for KSHV-induced lymphomas.
J Clin Invest
2007
, vol. 
117
 (pg. 
1019
-
1028
)
122
Esteban
 
M
García
 
MA
Domingo-Gil
 
E
Arroyo
 
J
Nombela
 
C
Rivas
 
C
The latency protein LANA2 from Kaposi's sarcoma-associated herpesvirus inhibits apoptosis induced by dsRNA-activated protein kinase but not RNase L activation.
J Gen Virol
2003
, vol. 
84
 (pg. 
1463
-
1470
)
123
Wies
 
E
Mori
 
Y
Hahn
 
A
, et al. 
The viral interferon-regulatory factor-3 is required for the survival of KSHV-infected primary effusion lymphoma cells.
Blood
2008
, vol. 
111
 (pg. 
320
-
327
)
124
Thai
 
TH
Calado
 
DP
Casola
 
S
, et al. 
Regulation of the germinal center response by microRNA-155.
Science
2007
, vol. 
316
 (pg. 
604
-
608
)
125
Skalsky
 
RL
Samols
 
MA
Plaisance
 
KB
, et al. 
Kaposi's sarcoma-associated herpesvirus encodes an ortholog of miR-155.
J Virol
2007
, vol. 
81
 (pg. 
12836
-
12845
)
126
Gottwein
 
E
Mukherjee
 
N
Sachse
 
C
, et al. 
A viral microRNA functions as an orthologue of cellular miR-155.
Nature
2007
, vol. 
450
 (pg. 
1096
-
1099
)
127
Moore
 
PS
Boshoff
 
C
Weiss
 
RA
Chang
 
Y
Molecular mimicry of human cytokine and cytokine response pathway genes by KSHV.
Science
1996
, vol. 
274
 (pg. 
1739
-
1344
)
128
Hoischen
 
SH
Vollmer
 
P
März
 
P
, et al. 
Human herpes virus 8 interleukin-6 homologue triggers gp130 on neuronal and hematopoietic cells.
Eur J Biochem
2000
, vol. 
267
 (pg. 
3604
-
36012
)
129
Burger
 
R
Neipel
 
F
Fleckenstein
 
B
, et al. 
Human herpesvirus type 8 interleukin-6 homologue is functionally active on human myeloma cells.
Blood
1998
, vol. 
91
 (pg. 
1858
-
1863
)
130
Jones
 
KD
Aoki
 
Y
Chang
 
Y
Moore
 
PS
Yarchoan
 
R
Tosato
 
G
Involvement of interleukin-10 (IL-10) and viral IL-6 in the spontaneous growth of Kaposi's sarcoma herpesvirus-associated infected primary effusion lymphoma cells.
Blood
1999
, vol. 
94
 (pg. 
2871
-
2879
)
131
Aoki
 
Y
Jaffe
 
ES
Chang
 
Y
, et al. 
Angiogenesis and hematopoiesis induced by Kaposi's sarcoma-associated herpesvirus-encoded interleukin-6.
Blood
1999
, vol. 
93
 (pg. 
4034
-
4043
)
132
Kovaleva
 
M
Bussmeyer
 
I
Rabe
 
B
, et al. 
Abrogation of viral interleukin-6 (vIL-6)-induced signaling by intracellular retention and neutralization of vIL-6 with an anti-vIL-6 single-chain antibody selected by phage display.
J Virol
2006
, vol. 
80
 (pg. 
8510
-
8520
)
133
Oksenhendler
 
E
Carcelain
 
G
Aoki
 
Y
, et al. 
High levels of human herpesvirus 8 viral load, human interleukin-6, interleukin-10, and C reactive protein correlate with exacerbation of multicentric castleman disease in HIV-infected patients.
Blood
2000
, vol. 
96
 (pg. 
2069
-
2073
)
134
Kaplan
 
LD
Straus
 
DJ
Testa
 
MA
Levine
 
AM
Kaplan
 
LD
Low-dose compared with standard-dose m-BACOD chemotherapy for non-Hodgkin's lymphoma associated with human immunodeficiency virus infection: National Institute of Allergy and Infectious Diseases AIDS Clinical Trials Group.
N Engl J Med
1997
, vol. 
336
 (pg. 
1641
-
1648
)
135
Mounier
 
N
Spina
 
M
Gabarre
 
J
, et al. 
AIDS-related non-Hodgkin lymphoma: final analysis of 485 patients treated with risk-adapted intensive chemotherapy.
Blood
2006
, vol. 
107
 (pg. 
3832
-
3840
)
136
Sparano
 
JA
Lee
 
S
Chen
 
MG
, et al. 
Phase II trial of infusional cyclophosphamide, doxorubicin, and etoposide in patients with HIV-associated non-Hodgkin's lymphoma: an Eastern Cooperative Oncology Group Trial (E1494).
J Clin Oncol
2004
, vol. 
22
 (pg. 
1491
-
1500
)
137
Little
 
RF
Pittaluga
 
S
Grant
 
N
, et al. 
Highly effective treatment of acquired immunodeficiency syndrome-related lymphoma with dose-adjusted EPOCH: impact of antiretroviral therapy suspension and tumor biology.
Blood
2003
, vol. 
101
 (pg. 
4653
-
4659
)
138
Coiffier
 
B
Lepage
 
E
Briere
 
J
, et al. 
CHOP chemotherapy plus rituximab compared with CHOP alone in elderly patients with diffuse large-B-cell lymphoma.
N Engl J Med
2002
, vol. 
346
 (pg. 
235
-
242
)
139
Habermann
 
TM
Weller
 
EA
Morrison
 
VA
, et al. 
Rituximab-CHOP versus CHOP alone or with maintenance rituximab in older patients with diffuse large B-cell lymphoma.
J Clin Oncol
2006
, vol. 
24
 (pg. 
3121
-
3127
)
140
Pfreundschuh
 
M
Trumper
 
L
Osterborg
 
A
, et al. 
CHOP-like chemotherapy plus rituximab versus CHOP-like chemotherapy alone in young patients with good-prognosis diffuse large-B-cell lymphoma: a randomised controlled trial by the MabThera International Trial (MInT) Group.
Lancet Oncol
2006
, vol. 
7
 (pg. 
379
-
391
)
141
Spina
 
M
Jaeger
 
U
Sparano
 
JA
, et al. 
Rituximab plus infusional cyclophosphamide, doxorubicin, and etoposide (R-CDE) in HIV-associated non-Hodgkin's lymphoma: pooled results from three phase II trials.
Blood
2005
, vol. 
105
 (pg. 
1891
-
1897
)
142
Spina
 
M
Simonelli
 
C
Vaccher
 
E
, et al. 
Long-term follow-up of rituxiamb plus infusional cyclophosphamide, doxorubicin and etoposide (CDE) in combination with HAART in HIV-related non-Hodgkin's lymphomas (NHL)
Ann Oncol
2008
, vol. 
19
 
Proocedings of the 10th International Conference on Malignant Lymphoma.
4–7 June 2008
Lugano, Switzerland
suppl 4
pg. 
iv151
  
Abstract 226
143
Boué
 
F
Gabarre
 
J
Gisselbrecht
 
C
, et al. 
Phase II trial of CHOP plus rituximab in patients with HIV-associated non-Hodgkin's lymphoma.
J Clin Oncol
2006
, vol. 
24
 (pg. 
4123
-
4128
)
144
Ribera
 
JM
Oriol
 
A
Morgades
 
M
, et al. 
Safety and efficacy of cyclophosphamide, adriamycin, vincristine, prednisone and rituximab in patients with human immunodeficiency virus-associated diffuse large B-cell lymphoma: results of a phase II trial.
Br J Hematol
2008
, vol. 
140
 (pg. 
411
-
419
)
145
Kaplan
 
LD
Lee
 
JY
Ambinder
 
RF
, et al. 
Rituximab does not improve clinical outcome in a randomized phase 3 trial of CHOP with or without rituximab in patients with HIV-associated non-Hodgkin lymphoma: AIDS-Malignancies Consortium Trial 010.
Blood
2005
, vol. 
106
 (pg. 
1538
-
1543
)
146
Levine
 
AM
Lee
 
J
Kaplan
 
L
Liebes L
 
F
Sparano
 
JA
Efficacy and toxicity of concurrent rituximab plus infusional EPOCH in HIV-associated lymphoma: AIDS Malignancy Consortium Trial 034
J Clin Oncol
2008
, vol. 
26
 
Prooceding of the 44th American Society of Clinical Oncology
May 30th–June 3rd 2008
Chicago
15S
pg. 
460s
  
Abstract 8527
147
Lim
 
ST
Karim
 
R
Nathwani
 
BN
Tulpule
 
A
Espina
 
B
Levine
 
AM
AIDS-related Burkitt's lymphoma versus diffuse large-cell lymphoma in the pre-highly active antiretroviral therapy (HAART) and HAART eras: significant differences in survival with standard chemotherapy.
J Clin Oncol
2005
, vol. 
23
 (pg. 
4430
-
4438
)
148
Spina
 
M
Simonelli
 
C
Talamini
 
R
Tirelli
 
U
Patients with HIV with Burkitt's lymphoma have a worse outcome than those with diffuse large-cell lymphoma also in the highly active antiretroviral therapy era.
J Clin Oncol
2005
, vol. 
23
 (pg. 
8132
-
8133
)
149
Wang
 
ES
Straus
 
DJ
Teruya-Feldstein
 
J
, et al. 
Intensive chemotherapy with cyclophosphamide, doxorubicin, high-dose methotrexate/ifosfamide, etoposide, and high-dose cytarabine (CODOX-M/IVAC) for human immunodeficiency virus-associated Burkitt lymphoma.
Cancer
2003
, vol. 
98
 (pg. 
1196
-
1205
)
150
Oriol
 
A
Ribera
 
JM
Esteve
 
J
, et al. 
Lack of influence of human immunodeficiency virus infection status in the response to therapy and survival of adult patients with mature B-cell lymphoma or leukemia: results of the PETHEMA-LAL3/97 study.
Haematologica
2003
, vol. 
88
 (pg. 
445
-
453
)
151
Oriol
 
A
Ribera
 
JM
Bergua
 
J
, et al. 
High-dose chemotherapy and immunotherapy in adult Burkitt lymphoma: comparison of results in human immunodeficiency virus-infected and noninfected patients.
Cancer
2008
, vol. 
113
 (pg. 
117
-
125
)
152
Boulanger
 
E
Gerard
 
L
Gabarre
 
J
, et al. 
Prognostic factors and outcome of human herpesvirus 8-associated primary effusion lymphoma in patients with AIDS.
J Clin Oncol
2005
, vol. 
23
 (pg. 
4372
-
4380
)
153
Spina
 
M
Gaidano
 
G
Carbone
 
A
Capello
 
D
Tirelli
 
U
Highly active antiretroviral therapy in human herpesvirus-8-related body-cavity-based lymphoma.
AIDS
1998
, vol. 
12
 (pg. 
955
-
956
)
154
Halfdanarson
 
TR
Markovic
 
SN
Kalokhe
 
U
Luppi
 
M
A non-chemotherapy treatment of a primary effusion lymphoma: durable remission after intracavitary cidofovir in HIV negative PEL refractory to chemotherapy.
Ann Oncol
2006
, vol. 
17
 (pg. 
1849
-
1850
)
155
Ghosh
 
SK
Wood
 
C
Boise
 
LH
, et al. 
Potentiation of TRAIL-induced apoptosis in primary effusion lymphoma through azidothymidine-mediated inhibition of NF-kappa B.
Blood
2003
, vol. 
101
 (pg. 
2321
-
2327
)
156
Riedel
 
DJ
Gonzalez-Cuyar
 
LF
Zhao
 
XF
Redfield
 
RR
Gilliam
 
BL
Plasmablastic lymphoma of the oral cavity: a rapidly progressive lymphoma associated with HIV infection.
Lancet Infect Dis
2008
, vol. 
8
 (pg. 
261
-
267
)
157
Schichman
 
SA
McClure
 
R
Schaefer
 
RF
Mehta
 
P
HIV and plasmablastic lymphoma manifesting in sinus, testicles, and bones: a further expansion of the disease spectrum.
Am J Hematol
2004
, vol. 
77
 (pg. 
291
-
295
)
158
Spina
 
M
Vaccher
 
E
Juzbasic
 
S
, et al. 
Human Immunodeficiency virus-related non-Hodgkin lymphoma: activity of infusional cyclophosphamide, doxorubicin and etoposide as second-line chemotherapy in 40 patients.
Cancer
2001
, vol. 
1
 (pg. 
200
-
206
)
159
Gabarre
 
J
Marcelin
 
AG
Azar
 
N
High-dose therapy plus autologous hematopoietic stem cell transplantation for human immunodeficiency virus (HIV)-related lymphoma: results and impact on HIV disease.
Haematologica
2004
, vol. 
89
 (pg. 
1100
-
1108
)
160
Krishnan
 
AY
Molina
 
A
Zaia
 
J
, et al. 
Durable remissions with autologous stem cell transplantation for high-risk HIV-associated lymphomas.
Blood
2005
, vol. 
105
 (pg. 
874
-
878
)
161
Re
 
A
Cattaneo
 
C
Michieli
 
M
, et al. 
High-dose therapy and autologous peripheral-blood stem-cell transplantation as salvage treatment for HIV-associated lymphoma in patients receiving highly active antiretroviral therapy.
J Clin Oncol
2003
, vol. 
21
 (pg. 
4423
-
4427
)
162
Krishnan
 
A
Zaia
 
J
Alvarnas
 
J
, et al. 
Autologous stem cell transplant (ASCT) for AIDS-related lymphoma (ARL).
Basic, Epidemiological and Clinical Reserch
Proocedings of the 11th International Conference on Malignancies and other Immunodeficiencies (ICMAOI)
October 6–7, 2008
Bethesda, MD
 
Abstract 041
Sign in via your Institution