219
Views
39
CrossRef citations to date
0
Altmetric
Review

Primary effusion lymphoma: current perspectives

, &
Pages 3747-3754 | Published online: 28 Jun 2018

Abstract

Primary effusion lymphoma (PEL) is a rare and aggressive disease, affecting a unique population of patients who are often elderly or immunocompromised. PEL is associated with human herpesvirus type-8 infection and most commonly presents as malignant effusions of the body cavities. Patients diagnosed with PEL often have a compromised immune system from secondary conditions such as HIV. Chemotherapy has traditionally been the cornerstone of treatment for patients with a good performance status and no significant comorbidities. However, an optimal regimen does not exist. Most patients with PEL experience a relapse after frontline therapy within 6–8 months and subsequently require further treatment. In recent years, our understanding of the molecular drivers and environmental factors affecting the pathogenesis of PEL has expanded. This review will discuss the pathogenesis of PEL and various management approaches available in the frontline and relapsed setting as well as targeted agents that have shown promise in this disease.

Introduction

Originally referred to as body cavity lymphoma, primary effusion lymphoma (PEL) is a distinct B-cell non-Hodgkin lymphoma (NHL) with an aggressive phenotype. It is caused by human herpesvirus type 8 (HHV8), also referred to as Kaposi sarcoma-associated herpesvirus (KSHV).Citation1 This virus was initially described in association with AIDS-associated Kaposi sarcoma (KS) in 1994.Citation2 Subsequently, PEL was found to be associated with HHV8 in 1995 and was first reported as a unique neoplasm by the World Health Organization (WHO) classification in 2001.Citation3

Epidemiology

PEL is rare and accounts for ~4% of HIV-associated NHL and <1% of non-HIV-related lymphomas.Citation4 There is a male predominance of 6:1.Citation5 PEL typically presents in middle-aged patients infected with HIV or harboring other immunocompromised states, such as recipients of solid-organ transplants, patients with cirrhosis, and in the elderly, often in HHV8 endemic areas.Citation6Citation10 Epstein Bar virus (EBV) co-infection is commonly found (60%–90% of cases) although its role in the pathogenesis of PEL is not clear.Citation11 EBV-negative PELs are typically found in elderly HIV-negative patients from HHV8-endemic areas.Citation12

Pathogenesis

The gamma-herpesvirus HHV8 is found in association with a variety of malignancies including PEL, KS, a variant of Multicentric Castleman Disease, HHV8-positive diffuse large B-cell lymphoma (DLBCL), and germinotropic lymphoproliferative disease.Citation4,Citation13 It is universally implicated with the oncogenesis of PEL, infecting the B-cell during its latent phase and replicating during the lytic phase.Citation6,Citation14 In the latent phase of infection by HHV8, many viral transcripts are expressed promoting oncogenesis. These include latency-associated nuclear antigen (LANA), viral FLICE inhibitory protein (v-FLIP) and viral cyclin (vCyclin) and are implicated in the progression of HHV8-associated malignancies.Citation3 LANA maintains the latent phase of the virus, and additionally represses the tumor suppressor protein p53 and retinoblastoma protein, leading to cell growth and survival.Citation15,Citation16 It also may contribute to NOTCH dysregulation and tumor progression.Citation17 vCyclin and v-FLIP contribute to tumor growth via constitutively activating cyclin-dependent kinase 6 and the transcription factor nuclear factor kappa B (NF-κB) pathway, respectively, leading to tumor proliferation and inhibition of apoptosis while maintaining viral latency.Citation18 HHV8 additionally produces interleukin IL-6 (vIL-6) which is found in a high concentration in PEL-related effusions and induces VEGF increasing vascular permeability and augmenting the formation of PEL-related effusions.Citation19 Additionally, vIL-6 prevents apoptosis by suppressing proapoptotic cathepsin D. Other HHV8 genes expressed during the latent phase of the viral life cycle affect oncogenesis via cell binding, proliferation, apoptosis, angiogenesis, cytokine production, B-cell proliferation all leading to tumor growth.Citation4,Citation13 The lytic and reproductive phase of HHV8 leads to lysis and death of the infected cell therefore it behooves the disease and virus to remain in the latent phase to promote tumor growth and cell mortality.

Clinical presentation

As its name describes, PEL presents with malignant lymphomatous effusions in body cavities (pleural space, peritoneal cavity, pericardium). The clinical presentation in PEL is based upon disease location and quantity of the effusion. A common presentation is an immunocompromised man who complains of shortness of breath, leading to an imaging finding of a pleural effusion. Increased abdominal girth, lower extremity edema, and increases in abdominal pressures may lead one to discover accumulation of ascitic fluid from PEL. Pericardial involvement may present with cardiac tamponade with symptoms of dizziness, low blood pressure, and electrocardiogram changes. Some patients may also present with typical B symptoms of fever, weight loss, and night sweats. More rarely, presentations such as an extracavitary mass have also been described. Areas of involvement include organs adjacent to a cavitary space, regional lymph nodes, bone marrow, skin, central nervous system, and the gastrointestinal tract.Citation20Citation29

Diagnostic testing

Histopathological assessment

Initial diagnosis is made by hematopathologic assessment of the effusion at presentation. As morphologically described by the WHO, PEL cells bridge those of large-cell immunoblastic lymphoma, plasmablastic lymphoma, and anaplastic large-cell lymphomaCitation30 (). The cells are large, with moderate to abundant deeply basophilic cytoplasm, a large round to irregular nuclei, and prominent nucleoli. They express CD45, proving lymphoid origins, however, are a “null” lymphocyte phenotype as they do not exhibit typical B-cell or T-cell immunophenotype characteristics and are indeterminate by immunohistochemistry (IHC). CD30, a marker found in 70% of classical Hodgkin lymphoma. Reed-Sternberg cells are commonly observed in PEL, whereas CD15 is typically not expressed. Markers of plasma cell differentiation, including CD38 and CD138, are present; of note plasma-cell myeloma is typically CD45 negative.Citation31 human leukocyte antigen – antigen D related (HLA-DR), activation antigen, and epithelial membrane antigen are variably expressed.

Table 1 Pathologic features differentiating PEL from other aggressive lymphomas

Most importantly, the confirmation in diagnosis of PEL is dependent on identifying HHV8 viral infection in the nuclei of the malignant cells. This is evidenced via expression of LANA-1 by IHC stain or via DNA extraction and polymerase chain reaction amplification of HHV8.Citation31 This verification by itself can differentiate the null lymphocyte phenotype PEL from other lymphomas demonstrating strikingly similar clinical presentations and morphologic features.

Cell of origin and genetic alterations in PEL

Gene expression profiling reveals the PEL cell of origin to be closely related to postgerminal center late-differentiating B-cells and likely of plasmablastic derivation.Citation32 While clonal rearrangement of the heavy immunoglobulin gene is detected, demonstrating B-cell derivation, a recurrent cytogenetic abnormality or PEL-specific driver mutation has not been identified.Citation31 Typical NHL-related gross rearrangements or mutations of BCL2, c-Myc, and TP53 are not identified in PEL.

Gaidano et al found a high frequency of BCL6 5′ noncoding region nucleotide substitution mutations in PEL.Citation33 Bcl6 5′ mutations are markers of B-cell transition through the germinal center. Therefore, these mutations suggest the origin as postgerminal-center B-cells.Citation31 In addition, a frequent occurrence of complete or partial trisomy 12, trisomy 7, and abnormalities of bands 1q21-25 have been noted.Citation33 Interleukin 1 receptor-associated kinase 1 (IRAK1), together with its binding partner MYD88, mediate toll-like receptor signaling and reactivate HHV8 leading to prolonged survival of PEL in culture. Via X chromosome targeted sequencing of PEL exudate cells, Yang et al found IRAK1 constitutively phosphorylated (mutated) in PEL and required for survival of these tumor cells.Citation34 Of note, the IRAK1 mutation is a common, essential driver for Kaposi sarcoma.

Staging

As per the Lugano classification for NHL, all patients who present an effusion have stage IV disease at diagnosis.Citation35 Radiographic evaluation is akin to other aggressive NHL diagnoses including positron emission tomography-computed tomography. Additionally, a bone marrow biopsy and/or lumbar puncture may be considered if clinically indicated.

Prognosis

Given its resistance to cytotoxic therapies, treatment with PEL has generally been associated with a poor prognosis. Several factors have been evaluated as potential markers of prognosis. In a retrospective study of 28 HIV-positive patients with PEL, two independent predictors of decreased survival were identified by multivariate analysis: poor performance status and absence of combined antiretroviral therapy (c-ART) prior to PEL diagnosis.Citation36 In a different analysis of 104 patients with PEL, the number and location of affected cavities appeared to play a role in prognosis.Citation5 Specifically, the involvement of more than one body cavity was associated with an overall survival (OS) of 4 months in comparison to 18 months in patients with only one cavity involved.

Management

Given the low incidence of PEL, there are no large-scale randomized studies to guide treatment and management decisions. Most of the evidence is based on retrospective studies, case reports, and preclinical data.

Frontline treatment

Chemotherapy has traditionally been the cornerstone of treatment for patients with a good performance status and no significant comorbidities. However, an optimal regimen does not exist. As the majority of the PEL are diagnosed in the setting of HIV infection, the approach to their management is also based on HIV status.

Chemotherapy

Although there is no one standard regimen that is universally accepted for the frontline treatment of PEL, an aggressive lymphoma regimen is typically used. Examples include dose-adjusted (DA) EPOCH (etoposide, prednisolone, vincristine, cyclophosphamide, doxorubicin) or CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone).

The use of DA-EPOCH for PEL can be extrapolated from a study conducted at the National Cancer Institute in 39 patients with newly diagnosed AIDS-associated aggressive B-cell lymphoma.Citation37 The regimen produced an overall response rate (ORR) of 87%, 74% of which were complete remissions (CRs). At 52 months, the disease-free survival and OS were 92% and 60%, respectively. The treatment was well tolerated with grade 3 or 4 neutropenia, anemia, and thrombocytopenia occurring in 30%, 17%, and 21% of the cycles, respectively. Serious constipation or stomatitis occurred in less than 3% of cycles, and grade 3 peripheral neuropathies occurred in two patients. Though no randomized controlled trials exist for PEL, there are case reports with the use of DA-EPOCH in combination with ART. In one case report, an HIV patient with extracavitary PEL received four cycles of EPOCH which led to a complete response after four cycles that lasted for 14 months after completing treatment.Citation38

Simonelli et al conducted a retrospective analysis evaluating the efficacy of a CHOP-like regimen in which prednisone was omitted in the majority of the eight patients included in order to prevent the emergence or exacerbation of KS. Three patients achieved a CR (42%), and the median OS was 6 months.Citation39 Boulanger et al explored the efficacy of another multi-agent regimen, CHVp plus methotrexate, in another retrospective study of seven patients with AIDS-associated PEL. Patients received cyclophosphamide 650–700 mg/m2, doxorubicin 35–40 mg/m2, vincristine 0.8 mg/m2, or etoposide 80 mg/m2 on day 1 followed by 4-hour intravenous infusion of methotrexate 2.5–3 g/m2 on day 2 for 6–8 cycles every 21 days.Citation40 Three achieved a CR and remained in remission at 18, 26, and 78 months. Most of the patients had hematological toxicity with delayed clearance of methotrexate due to the presence of effusions. The use of methotrexate should be avoided as majority of the patients have an effusion that leads to decreased clearance and prolonged toxicity with methotrexate.

Rarely, PEL are CD20 positive. As with other B-cell lymphomas, treatment should incorporate rituximab-based chemoimmunotherapy. Two single-patient case reports have demonstrated the efficacy of RCHOP in this population, resulting in durable CR lasting 22 and 30 months.Citation41,Citation42 The use of R-EPOCH in an HIV-patient resulted in CR lasting 12 months.Citation43

Anti-retroviral therapy

Anti-retroviral treatment (ART) is an important component of the management in patients with lymphoproliferative malignancies in the setting of HIV. In a retrospective study of HIV-infected patients diagnosed with PEL, Boulanger et al demonstrated a shorter OS for patients with a poor performance status and untreated HIV infection prior to diagnosis.Citation36 Similarly, patients treated with a CHOP-like regimen without ART were unable to achieve a CR and had a shorter OS of 3 months.Citation39 ART given by itself can also lead to a CR, as evident by one case report in which a patient remained in CR at 14 months.Citation44

Treatment with ART should therefore be the mainstay of HIV-positive patients with PEL. In terms of choice of antiretroviral agents, some agents have more activity in PEL than others based on preclinical studies. Azidothymidine has been shown to sensitize primary effusion lymphoma cells to Kaposi sarcoma associated herpesvirus-specific CD4+ T cells and inhibits proliferation in PEL cell lines.Citation45 Similarly, preclinical studies with the protease inhibitor Lopinavir has shown induction of apoptosis of PEL cells via suppression of the NF-κB pathway. Interestingly, this effect, though present, was not as significant with other protease inhibitors like Ritonavir and Darunavir.Citation46 Hence, involving the infectious disease specialist in managing these patients is crucial.

Relapsed and refractory disease

Most patients with PEL experience a relapse after frontline therapy within 6–8 months and subsequently require further treatment.Citation11 Treatment options will depend upon the performance status, comorbidities, and goals of care based on the individual. Therapies that have been evaluated in patients include stem cell transplant, radiation, and targeted agents such as bortezomib.

Stem cell transplant

Autologous stem cell transplant (ASCT) has been attempted in a handful of patients. Waddington et al published a case report of an HIV-positive patient with PEL who received high-dose chemotherapy followed by autologous stem cell transplant in PEL.Citation47 The patient had persistent disease prior to ASCT. The transplant was unsuccessful as he developed recurrent accumulation of pleural fluid consistent with persistent disease indicating failure of ASCT. Won et al demonstrated the effectiveness of ASCT in an HIV-negative patient who proceeded to ASCT after achieving a CR with salvage chemotherapy (ifosfamide, carboplatin, and etoposide).Citation48 The patient remained in CR 12 months after completion of ASCT.

As the majority of patients with PEL have concurrent HIV infection, there has been great apprehension regarding the use of allogenic stem cell transplant (allo-SCT) in this population. Nevertheless, Bryant et al successfully performed a reduced intensity conditioning regimen followed by allo-SCT in one HIV-positive patient with PEL.Citation49 The patient achieved a CR and remained in CR for a period of 31 months after transplantation. There have been no other reports of the use of allo-SCT in PEL, therefore, the role for this therapy remains uncharted.

Radiation

When the goal of treatment shifts toward comfort in patients with refractory or relapsed disease, an important component is to identify if the site of disease is causing physical distress. Radiation by itself is inadequate in most instances for achieving a complete response as with other aggressive lymphomas; however, it can be useful to control the disease burden. In a case report by Cassoni et al, a multiply-refractory patient with PEL with a pleural-based mass was treated with localized radiation resulting in a sustained remission for 12 months.Citation70 Radiation can be considered for PEL with a solid component involved within one radiation field.

Antiviral therapies

As the malignant cells in PEL are infected with HHV8 virus, they theoretically can be targeted through antiviral therapy. Unfortunately, since the HHV8 virus is in a latent state, it is resistant to most antiviral agents. Klass and Offermann proposed a potential solution whereby induction of the HHV8 virus into the lytic phase via valproate followed by treatment with a lytic-phase effective anti-herpetic drug may lead to apoptosis of the infected cell.Citation50 Using an HHV-8 positive PEL cell line, they found lytic replication to be sensitive to ganciclovir, foscarnet and cidofovir, and resistant to acyclovir.Citation50,Citation51

The effectiveness of antiviral therapy in PEL has been demonstrated in a handful of case reports. An HIV patient who was refractory to frontline chemotherapy regimen of bortezomib, cyclophosphamide, Adriamycin, and prednisone experienced a CR with valganciclovir while continuing to receive c-ART.Citation52 The patient received treatment for 12 months but had achieved a CR by 6 months with eradication of the HHV8 virus. Seven months after completion of therapy, the patient continued on c-ART and remained disease free. Cidofovir has also been used in combination with ART and interferon. Hocqueloux et al published a case report of a patient with HIV-associated PEL treated with cidofovir and interferon (IFN)-α: cidofovir 5 mg/kg every 15 days in combination, IFN-α 3 million units 3 times a week.Citation53 A CR was achieved after 2 months of treatment; cidofovir was stopped at 3 months, whereas IFN-α was continued for 7 months. The patient remained in a complete remission for 24 months. The inferior activity of cidofovir is postulated to be due to poor penetration of the drug into the effusion.Citation54,Citation55 It has been shown to be highly active as a single agent when administered directly into the affected cavity. Luppi et al successfully treated three patients with HIV-negative, HHV8+ PEL with intracavitary cidofovir 2.5–5 mg/kg every week.Citation55 The first patient achieved a CR with two doses of intrapleural cidofovir and maintained a remission for 10 months. The second patient achieved a CR after 3 doses of intraperitoneal cidofovir, maintaining a remission for 5 months. The third patient achieved a CR after three doses of intrapleural cidofovir and remained treatment free for 15 months. This approach is limited to disease involving only one cavity but may be extrapolated to HIV-positive patients as well. There are no data for the use of intracavitary cidofovir injection in multiple cavities.

Talc pleurodesis

When PEL presents solely as an effusion, palliative approaches to decrease accumulation of fluid can be performed. One commonly utilized approach in other malignancies including malignant mesothelioma is pleurodesis. In this procedure, the visceral and parietal pleural layers are fused together so that there is no accumulation of fluid. Talc is a sclerosant which has been effectively used in this process. In addition to causing a fusion of the visceral and parietal pleura, it also has an apoptotic effect on mesothelioma cells by regulating the surface expression of the proto-oncogene c-myc and enhancing apoptosis.Citation56 A case series has shown that it is possible to achieve and maintain long-term remissions in patients with HIV-negative HHV8-associated PEL with video-assisted talc pleurodesis.Citation57 Remissions were achieved instantaneously after the procedure and lasting 50–60 months in the three patients evaluable. This is a reasonable option for elderly, frail individuals who have limited treatment options and are unable to tolerate aggressive treatments.

Targeted therapies

Proteasome inhibitors

Proteasomal activity is required for the survival of PEL cells and viral replication of KSHV cells. Preclinical data have shown that proteasome inhibitors reduce cell proliferation and induce apoptosis in KSHV-positive, EBV-positive and KSHV-positive, EBV-negative PEL cell lines.Citation54 As described previously, HHV8 viral latent transcripts also constitutively activate the NF-κB pathway, leading to tumor cell proliferation and survival. Bortezomib is a proteasome inhibitor currently approved in multiple myeloma and mantle cell lymphoma.Citation58 While single-agent bortezomib was not shown to be active in a small case series of three HIV-infected patients with relapsed/refractory PEL, it has promise in combination with other agents.Citation8 Siddiqi et al administered a combination of bortezomib, PEGylated liposomal doxorubicin, and rituximab to an HIV-negative patient with CD20 positive PEL, which resulted in a durable CR lasting at least 2 years.Citation59 Bortezomib has also been studied with the histone deacetylase inhibitor, vorinostat, in a murine PEL xenograft model. The combination of bortezomib and vorinostat led to KSHV lytic replication and cell death, thus demonstrating the synergy of vorinostat and bortezomib.Citation60

Brentuximab vedotin (BV)

BV is an antibody drug conjugate against CD30, currently approved in previously treated classical Hodgkin lymphoma and anaplastic large cell lymphoma.Citation61 As PEL is typically CD30 positive, Bhatt et al evaluated the role of BV in CD30 positive UM-PEL-1 and UM-PEL-3 cell lines and xenograft models.Citation62 BV induced G2 phase cell cycle arrest with intracellular delivery of the drug monomethyl auristatin E, inducing apoptosis of both the CD30 expressing cells and intratumoral cells lacking the target antigen. This effect was also observed in xenograft models inoculated with UM-PEL-1 and UM-PEL-3 cells. While there have been no clinical trials of BV in PEL, it has shown promise in other CD30+ lymphomas. Jacobsen et al administered BV to patients with CD30+ non-Hodgkin lymphoma patients, resulting in notable activity among the different histologies.Citation63 The ORR in DLBCL was 44%, including 17% complete remissions and a median duration of response of 16.6 months (range 2.7–22.7 months). Responses were also seen in patients with gray zone and posttransplant lymphoproliferative disorders. While the true efficacy in PEL is unclear, BV may be considered in select patients.

Preclinical studies

Preclinical data have provided insight into targeted therapies that may have efficacy in PEL. The mammalian target of rapamycin (mTOR), its activator AKT, and the target p70S6 kinases are frequently phosphorylated in PEL. Rapamycin, an inhibitor of mTOR, has been tested in a variety of PEL cell lines (BC-1, BC-3, JSC-1, BCBL-1, BCP-1, and VG-1), proving its ability to inhibit proliferation and induce apoptosis.Citation64 It has also been studied in xenograft models, confirming its ability to inhibit PEL tumor growth. PEL cell lines also secrete high levels of VEGF and L-6.Citation65 Given these findings, the VEGF inhibitor, bevacizumab, and the IL-6 inhibitor, tocilizumab, have also been studied in PEL cell lines (BCBL-1, BC-1, BC-3, TY-1) and xenograft mouse models.Citation66 Neither bevacizumab or tocilizumab were able to inhibit proliferation in the cell line studies, however, both agents were able to inhibit the development of malignant pleural effusion and provide a survival benefit in the xenograft models. These data support the exploration of the agents’ utility in the clinical setting.

The immunomodulatory agent, lenalidomide, was initially approved in multiple myeloma but has also shown activity in other lymphoid malignancies including mantle cell lymphoma.Citation67 It has been evaluated with arsenic trioxide, a potent agent in acute promyelocytic leukemia.Citation68 The combination resulted in inhibition of growth in PEL cell lines (BC-1 and BC-3) by upregulating p53 and inducing apoptosis.Citation69 Reduction of the expression of latent viral transcripts (LANA-1, LANA-2, v-FLIP, and v-Cyclin) and proteins (LANA-1/LANA-2) was also noted. In PEL xenograft models, lenalidomide and arsenic trioxide induced greater responses and OS than lenalidomide alone. These preclinical studies provide a rational for the use of these agents in the clinical setting. Unfortunately, given the rarity of this disease and the comorbidities associated with it, it is difficult to develop readily accessible trials for patients.

Conclusion

PEL is an uncommon and aggressive disease affecting a unique population of patients who are often elderly or immunocompromised. Most of the patients diagnosed with PEL have significant other comorbidities, such as HIV, along with a poor performance status which limit their enrollment in clinical trials. For the fit and healthy patients, intensive chemotherapy remains the preferred choice, however, the availability of various targeted agents may alter this approach. For the unfit individuals, palliative treatments or newer targeted agents should be considered. With the improvement in our understanding of PEL, the treatment landscape is evolving and promising. Despite our advances, for now the use of ART for HIV-positive patients will continue to remain the backbone of treatment.

Disclosure

The authors report no conflicts of interest in this work.

References

  • JarrettRFViruses and lymphoma/leukaemiaJ Pathol2006208217618616362996
  • ChangYCesarmanEPessinMSIdentification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcomaScience19942665192186518697997879
  • CesarmanEChangYMoorePSSaidJWKnowlesDMKaposi’s sarcoma-associated herpesvirus-like DNA sequences in AIDS-related body-cavity-based lymphomasN Engl J Med199533218118611917700311
  • KaplanLDHuman herpesvirus-8: Kaposi sarcoma, multicentric Castle-man disease, and primary effusion lymphomaHematol Am Soc Hematol Educ Program20132013103108
  • CastilloJJShumHLahijaniMWinerESButeraJNPrognosis in primary effusion lymphoma is associated with the number of body cavities involvedLeuk Lymphoma201253122378238222591071
  • ChenY-BRahemtullahAHochbergEPrimary effusion lymphomaOncologist200712556957617522245
  • BoulangerEHermineOFermandJ-PHuman herpesvirus 8 (HHV-8)-associated peritoneal primary effusion lymphoma (PEL) in two HIV-negative elderly patientsAm J Hematol2004761889115114607
  • BoulangerEAfonsoPVYahiaouiYAdle-BiassetteHGabarreJAgbalikaFHuman herpesvirus-8 (HHV-8)-associated primary effusion lymphoma in two renal transplant recipients receiving rapamycinAm J Transplant20088370771018261181
  • SaidJWTasakaTTakeuchiSPrimary effusion lymphoma in women: report of two cases of Kaposi’s sarcoma herpes virus-associated effusion-based lymphoma in human immunodeficiency virus-negative womenBlood1996888312431288874212
  • SasakiYIsegawaTShimabukuroAYonahaTYonahaHPrimary effusion lymphoma in an elderly HIV-negative patient with hemodialysis: importance of evaluation for pleural effusion in patients receiving hemodialysisCase Rep Nephrol Urol2014429510224987405
  • DunleavyKWilsonWHHow I treat HIV-associated lymphomaBlood2012119143245325522337719
  • SwerdlowSHCampoEHarrisNLWHO Classification of Tumours of Haematopoietic and Lymphoid TissuesRevised 4th ed2LyonInternational Agency for Research on Cancer2016
  • CarboneAGloghiniAKSHV/HHV8-associated lymphomasBr J Haematol20081401132417991301
  • SaidJKaposi’s sarcoma-associated herpesvirus (KSHV): a new viral pathogen associated with Kaposi’s sarcoma, primary effusion lymphoma, and multicentric Castleman’s diseaseWest J Med1997167137389265864
  • FriborgJJrKongWHottigerMONabelGJp53 inhibition by the LANA protein of KSHV protects against cell deathNature1999402676488989410622254
  • RadkovSAKellamPBoshoffCThe latent nuclear antigen of Kaposi sarcoma-associated herpesvirus targets the retinoblastoma-E2F pathway and with the oncogene Hras transforms primary rat cellsNat Med20006101121112711017143
  • DeCotiisJLLukacDMKSHV and the role of NOTCH receptor dysregulation in disease progressionPathogens201763E34
  • DirekzeSLamanHRegulation of growth signalling and cell cycle by Kaposi’s sarcoma-associated herpesvirus genesInt J Exp Pathol200485630531915566428
  • SakakibaraSTosatoGViral interleukin-6: role in Kaposi’s sarcoma-associated herpesvirus–associated malignanciesJ Interferon Cytokine Res2011311179180121767154
  • ElySAPowersJLewisDKaposi’s sarcoma-associated herpesvirus-positive primary effusion lymphoma arising in the suba-rachnoid spaceHum Pathol199930898198410452513
  • PielasinskiUSantonjaCRodríguez-PinillaSMRequenaLExtracavitary primary effusion lymphoma presenting as a cutaneous tumor: a case report and literature reviewJ Cutan Pathol201441974575324917369
  • InoueSMiyamotoTYoshinoTYamadoriIHagariYYamamotoOPrimary effusion lymphoma with skin involvementJ Clin Pathol200659111221122217071811
  • IbrahimUSaqibAMohammadFDingJHusseinSAtallahJPKSHV-associated extracavitary primary effusion lymphoma in an HIV seronegative patient: a case report and review of the literaturePostgrad Med2017129340240728122468
  • Kabiawu-AjiseOEHarrisJIsmailiNAmorosiEIbrahimSPrimary effusion lymphoma with central nervous system involvement in an HIV-negative homosexual maleActa Haematol20121282778222699313
  • ChadburnAHyjekEMathewSCesarmanESaidJKnowlesDMKSHV-positive solid lymphomas represent an extra-cavitary variant of primary effusion lymphomaAm J Surg Pathol200428111401141615489644
  • MedeirosBCManessLJBauerFARossJWKapurDUnusual presentation of “extracavitary” primary effusion lymphoma in previously unknown HIV diseaseConn Med2000641059159411100630
  • PantanowitzLWuZDezubeBJPihanGExtracavitary primary effusion lymphoma of the anorectumClin Lymphoma Myeloma20056214915216231856
  • KimYLeventakiVBhaijeeFJacksonCCMedeirosLJVegaFExtracavitary/solid variant of primary effusion lymphomaAnn Diagn Pathol201216644144622497739
  • SantonjaCMedina-PuenteCSerrano Del CastilloCCabello ÚbedaARodríguez-PinillaSMPrimary effusion lymphoma involving cerebrospinal fluid, deep cervical lymph nodes and adenoids. Report of a case supporting the lymphatic connection between brain and lymph nodesNeuropathology201737324925827862361
  • NadorRGCesarmanEChadburnAPrimary effusion lymphoma: a distinct clinicopathologic entity associated with the Kaposi’s sarcoma-associated herpes virusBlood19968826456568695812
  • BrimoFMichelRPKhetaniKAugerMPrimary effusion lymphoma: a series of 4 cases and review of the literature with emphasis on cytomorphologic and immunocytochemical differential diagnosisCancer2007111422423317554754
  • KleinUGloghiniAGaidanoGGene expression profile analysis of AIDS-related primary effusion lymphoma (PEL) suggests a plasmablastic derivation and identifies PEL-specific transcriptsBlood2003101104115412112531789
  • GaidanoGCapelloDCiliaAMGenetic characterization of HHV-8/KSHV-positive primary effusion lymphoma reveals frequent mutations of BCL6: implications for disease pathogenesis and histogenesisGenes Chromosomes Cancer199924116239892104
  • YangDChenWXiongJSherrodCJHenryDHDittmerDPInterleukin 1 receptor-associated kinase 1 (IRAK1) mutation is a common, essential driver for Kaposi sarcoma herpesvirus lymphomaProc Natl Acad Sci U S A201411144E4762E476825341731
  • ChesonBDFisherRIBarringtonSFRecommendations for initial evaluation, staging, and response assessment of Hodgkin and non-Hodgkin lymphoma: the Lugano classificationJ Clin Oncol201432273059306825113753
  • BoulangerEGérardLGabarreJPrognostic factors and outcome of human herpesvirus 8-associated primary effusion lymphoma in patients with AIDSJ Clin Oncol200523194372438015994147
  • LittleRFPittalugaSGrantNHighly effective treatment of acquired immunodeficiency syndrome-related lymphoma with dose-adjusted EPOCH: impact of antiretroviral therapy suspension and tumor biologyBlood2003101124653465912609827
  • El-AyassWYuE-MKarcherDSAragon-ChingJBComplete response to EPOCH in a patient with HIV and extracavitary primary effusion lymphoma involving the colon: a case report and review of literatureClin Lymphoma Myeloma Leuk201212214414722264773
  • SimonelliCSpinaMCinelliRClinical features and outcome of primary effusion lymphoma in HIV-infected patients: a single-institution studyJ Clin Oncol200321213948395414581418
  • BoulangerEDanielM-TAgbalikaFOksenhendlerECombined chemotherapy including high-dose methotrexate in KSHV/HHV8-associated primary effusion lymphomaAm J Hematol200373314314812827649
  • TerasakiYOkumuraHSaitoKHHV-8/KSHV-negative and CD20-positive primary effusion lymphoma successfully treated by pleural drainage followed by chemotherapy containing rituximabIntern Med200847242175217819075546
  • SuzukiKInoKSugawaraYMizutaniMSekineTKatayamaNProlonged survival in a patient with human herpesvirus-8-negative primary effusion lymphoma after combination chemotherapy with rituximabGan To Kagaku Ryoho2008354691694 In Japanese18408447
  • FosterWRBischinADorerRAboulafiaDMHuman herpesvirus type 8-associated large B-cell lymphoma: a nonserous extracavitary variant of primary effusion lymphoma in an HIV-infected man: a case report and review of the literatureClin Lymphoma Myeloma Leuk201616631132127234438
  • OksenhendlerEClauvelJPJouveshommeSDaviFMansourGComplete remission of a primary effusion lymphoma with antiretroviral therapyAm J Hematol1998573266
  • WilliamsonSJNicolSMStürzlMSabbahSHislopADAzidothymidine sensitizes primary effusion lymphoma cells to Kaposi sarcoma-associated herpesvirus-specific CD4+ T cell control and inhibits vIRF3 functionPLoS Pathog20161211e100604227893813
  • KariyaRTauraMSuzuSKaiHKatanoHOkadaSHIV protease inhibitor Lopinavir induces apoptosis of primary effusion lymphoma cells via suppression of NF-κB pathwayCancer Lett20143421525924012878
  • WaddingtonTWAboulafiaDMFailure to eradicate AIDS-associated primary effusion lymphoma with high-dose chemotherapy and autologous stem cell reinfusion: case report and literature reviewAIDS Patient Care STDs2004182677315006181
  • WonJ-HHanS-HBaeS-BSuccessful eradication of relapsed primary effusion lymphoma with high-dose chemotherapy and autologous stem cell transplantation in a patient seronegative for human immunodeficiency virusInt J Hematol200683432833016757433
  • BryantAMillikenSSuccessful reduced-intensity conditioning allogeneic HSCT for HIV-related primary effusion lymphomaBiol Blood Marrow Transplant200814560160218410904
  • KlassCMOffermannMKTargeting human herpesvirus-8 for treatment of Kaposi’s sarcoma and primary effusion lymphomaCurr Opin Oncol200517544745516093794
  • KedesDHGanemDSensitivity of Kaposi’s sarcoma-associated herpesvirus replication to antiviral drugs. Implications for potential therapyJ Clin Invest1997999208220869151779
  • MarquetJVelazquez-KennedyKLópezSBenitoABlanchardM-JGarcia-VelaJACase report of a primary effusion lymphoma successfully treated with oral valganciclovir after failing chemotherapyHematol Oncol201836131631928580733
  • HocquelouxLAgbalikaFOksenhendlerEMolinaJMLong-term remission of an AIDS-related primary effusion lymphoma with antiviral therapyAIDS200115228028211216942
  • HalfdanarsonTRMarkovicSNKalokheULuppiMA non-chemotherapy treatment of a primary effusion lymphoma: durable remission after intracavitary cidofovir in HIV negative PEL refractory to chemotherapyAnn Oncol200617121849185016766593
  • LuppiMTrovatoRBarozziPTreatment of herpesvirus associated primary effusion lymphoma with intracavity cidofovirLeukemia200519347347615674353
  • NasreenNMohammedKADowlingPAWardMJGalffyGAntonyVBTalc induces apoptosis in human malignant mesothelioma cells in vitroAm J Respir Crit Care Med20001612 Pt 159560010673205
  • BirsenRBoutboulDCrestaniBTalc pleurodesis allows long-term remission in HIV-unrelated Human Herpesvirus 8-associated primary effusion lymphomaLeuk Lymphoma20175881993199828084853
  • RaedlerLVelcade (Bortezomib) receives 2 new FDA indications: for retreatment of patients with multiple myeloma and for first-line treatment of patients with mantle-cell lymphomaAm Health Drug Benefits20158Spec Feature13514026629279
  • SiddiqiTJoyceRMA case of HIV-negative primary effusion lymphoma treated with bortezomib, pegylated liposomal doxorubicin, and rituximabClin Lymphoma Myeloma20088530030418854285
  • BhattSAshlockBMToomeyNLEfficacious proteasome/HDAC inhibitor combination therapy for primary effusion lymphomaJ Clin Invest201312362616262823635777
  • ADCETRIS® (brentuximab vedotin) | U.S. | Seattle Genetics http://www.seattlegenetics.com/products/adcetris-usAccessed February 24, 2018
  • BhattSAshlockBMNatkunamYCD30 targeting with brentuximab vedotin: a novel therapeutic approach to primary effusion lymphomaBlood201312271233124223838350
  • JacobsenEDSharmanJPOkiYBrentuximab vedotin demonstrates objective responses in a phase 2 study of relapsed/refractory DLBCL with variable CD30 expressionBlood201512591394140225573987
  • SinS-HRoyDWangLRapamycin is efficacious against primary effusion lymphoma (PEL) cell lines in vivo by inhibiting autocrine signalingBlood200710952165217317082322
  • AokiYTosatoGRole of vascular endothelial growth factor/vascular permeability factor in the pathogenesis of Kaposi’s sarcoma-associated herpesvirus-infected primary effusion lymphomasBlood199994124247425410590069
  • GotoHKudoEKariyaRTauraMKatanoHOkadaSTargeting VEGF and interleukin-6 for controlling malignant effusion of primary effusion lymphomaJ Cancer Res Clin Oncol2015141346547425304617
  • DimopoulosMAKastritisERajkumarSVTreatment of plasma cell dyscrasias with lenalidomideLeukemia20082271343135318509355
  • Trisenox (Arsenic trioxide) [package insert]Teva Pharmaceuticals USA, IncNorth Wales, PA 19454
  • HleihelRTawilNKaramMMerhiRABazarbachiAHajjHEArsenic trioxide and lenalidomide: a promising combination therapy for primary effusion lymphomaBlood2017130Suppl 12817
  • CassoniAAliUCaveJRemission after radiotherapy for a patient with chemotherapy-refractory HIV-associated primary effusion lymphomaJ Clin Oncol200826325297529918854561