Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Eur J Haematol ; 112(5): 723-730, 2024 May.
Article in English | MEDLINE | ID: mdl-38155405

ABSTRACT

OBJECTIVE: To describe cases of Kaposi's sarcoma-associated herpesvirus (KSHV)-associated multicentric Castleman's disease (MCD) and primary effusion lymphoma (PEL) in patients with HIV from a large, safety-net hospital system in Dallas, Texas, USA. METHODS: We conducted a retrospective review of patients with HIV-associated PEL and/or MCD. RESULTS: Twelve patients with PEL and 10 patients with MCD were identified. All patients were male and 17 of 20 were men who have sex with men; 66.7% of PEL patients and 50% of MCD patients had concurrent KS at the time of diagnosis; 42% of patients with PEL and 20% of patients with MCD died during the follow-up period. We noted improved survival in our cohort compared to previous studies, particularly in our PEL patients with a median survival of 11.4 months compared to 3-6-month median survival historically. Median follow-up time for MCD patients was 17.5 months. This improved survival is despite suboptimal antiretroviral therapy (ART) adherence at diagnosis, with only 50% of patients on ART at the time of MCD/PEL diagnosis. CONCLUSION: These data highlight the importance of early recognition of PEL and MCD, and the larger-scale efforts needed to better understand the pathogenetic drivers of clinical outcomes in patients affected by KSHV-related diseases.


Subject(s)
Castleman Disease , HIV Infections , Herpesvirus 8, Human , Lymphoma, Primary Effusion , Sarcoma, Kaposi , Sexual and Gender Minorities , Humans , Male , Female , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/diagnosis , Sarcoma, Kaposi/epidemiology , HIV , Homosexuality, Male , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/epidemiology , Lymphoma, Primary Effusion/etiology , Safety-net Providers , Castleman Disease/complications , Castleman Disease/diagnosis , HIV Infections/complications
2.
Mol Carcinog ; 60(8): 567-581, 2021 08.
Article in English | MEDLINE | ID: mdl-34101920

ABSTRACT

The sesquiterpene lactone parthenolide is a major component of the feverfew medicinal plant, Tanacetum parthenium. Parthenolide has been extensively studied for its anti-inflammatory and anticancer properties in several tumor models. Parthenolide's antitumor activities depend on several mechanisms but it is mainly known as an inhibitor of the nuclear factor-κB (NF-κB) pathway. This pathway is constitutively activated and induces cell survival in primary effusion lymphoma (PEL), a rare aggressive AIDS-related lymphoproliferative disorder that is commonly caused by the human herpesvirus 8 (HHV-8) infection. The aim of this study is to evaluate the targeted effect of Parthenolide both in vitro and in vivo. Herein, parthenolide significantly inhibited cell growth, induced G0 /G1 cell cycle arrest, and induced massive apoptosis in PEL cells and ascites. In addition, parthenolide inhibited the NF-ĸB pathway suppressing IĸB phosphorylation and p65 nuclear translocation. It also reduced the expression of the DNA methylase inhibitor (DNMT1). Parthenolide induced HHV-8 lytic gene expression without inhibiting latent viral gene expression. Importantly, DMAPT, the more soluble parthenolide prodrug, promoted delay in ascites development and prolonged the survival of PEL xenograft mice. This study supports the therapeutic use of parthenolide in PEL and encourages its further clinical development.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Lymphoma, Primary Effusion/drug therapy , Sesquiterpenes/pharmacology , Animals , Apoptosis/drug effects , Biomarkers , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Disease Models, Animal , Disease Susceptibility , Drug Evaluation, Preclinical , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
3.
Blood ; 136(19): 2175-2187, 2020 11 05.
Article in English | MEDLINE | ID: mdl-32518949

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive malignancy with poor prognosis even under chemotherapy. Kaposi sarcoma-associated herpesvirus (KSHV), one of the human oncogenic viruses, is the principal causative agent. Currently, there is no specific treatment for PEL; therefore, developing new therapies is of great importance. Sphingolipid metabolism plays an important role in determining the fate of tumor cells. Our previous studies have demonstrated that there is a correlation between sphingolipid metabolism and KSHV+ tumor cell survival. To further develop sphingolipid metabolism-targeted therapy, after screening a series of newly synthesized ceramide analogs, here, we have identified compounds with effective anti-PEL activity. These compounds induce significant PEL apoptosis, cell-cycle arrest, and intracellular ceramide production through regulation of ceramide synthesizing or ceramide metabolizing enzymes and dramatically suppress tumor progression without visible toxicity in vivo. These new compounds also increase viral lytic gene expression in PEL cells. Our comparative transcriptomic analysis revealed their mechanisms of action for inducing PEL cell death and identified a subset of novel cellular genes, including AURKA and CDCA3, controlled by sphingolipid metabolism, and required for PEL survival with functional validation. These data provide the framework for the development of promising sphingolipid-based therapies against this virus-associated malignancy.


Subject(s)
Aurora Kinase A/metabolism , Ceramides/pharmacology , Herpesvirus 8, Human/pathogenicity , Lymphoma, Primary Effusion/drug therapy , Sarcoma, Kaposi/complications , Sphingolipids/pharmacology , Animals , Apoptosis , Aurora Kinase A/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Survival , Ceramides/chemistry , Female , Gene Expression Profiling , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Sarcoma, Kaposi/virology , Tumor Cells, Cultured , Virus Replication , Xenograft Model Antitumor Assays
5.
Blood Adv ; 3(14): 2105-2117, 2019 07 23.
Article in English | MEDLINE | ID: mdl-31300418

ABSTRACT

Genome-wide CRISPR/Cas9 screens represent a powerful approach to studying mechanisms of drug action and resistance. Cereblon modulating agents (CMs) have recently emerged as candidates for therapeutic intervention in primary effusion lymphoma (PEL), a highly aggressive cancer caused by Kaposi's sarcoma-associated herpesvirus. CMs bind to cereblon (CRBN), the substrate receptor of the cullin-RING type E3 ubiquitin ligase CRL4CRBN, and thereby trigger the acquisition and proteasomal degradation of neosubstrates. Downstream mechanisms of CM toxicity are incompletely understood, however. To identify novel CM effectors and mechanisms of CM resistance, we performed positive selection CRISPR screens using 3 CMs with increasing toxicity in PEL: lenalidomide (LEN), pomalidomide (POM), and CC-122. Results identified several novel modulators of the activity of CRL4CRBN The number of genes whose inactivation confers resistance decreases with increasing CM efficacy. Only inactivation of CRBN conferred complete resistance to CC-122. Inactivation of the E2 ubiquitin conjugating enzyme UBE2G1 also conferred robust resistance against LEN and POM. Inactivation of additional genes, including the Nedd8-specific protease SENP8, conferred resistance to only LEN. SENP8 inactivation indirectly increased levels of unneddylated CUL4A/B, which limits CRL4CRBN activity in a dominant negative manner. Accordingly, sensitivity of SENP8-inactivated cells to LEN is restored by overexpression of CRBN. In sum, our screens identify several novel players in CRL4CRBN function and define pathways to CM resistance in PEL. These results provide rationale for increasing CM efficacy on patient relapse from a less-efficient CM. Identified genes could finally be developed as biomarkers to predict CM efficacy in PEL and other cancers.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , CRISPR-Cas Systems , Clustered Regularly Interspaced Short Palindromic Repeats , Lymphoma, Primary Effusion/etiology , Adaptor Proteins, Signal Transducing/metabolism , Cullin Proteins/metabolism , Drug Resistance, Neoplasm/genetics , Endopeptidases/genetics , Gene Knockdown Techniques , Genome-Wide Association Study , Humans , Lenalidomide/adverse effects , Lenalidomide/pharmacology , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Models, Biological , Thalidomide/analogs & derivatives , Thalidomide/pharmacology , Ubiquitin-Protein Ligases
6.
Clin Lymphoma Myeloma Leuk ; 19(9): e513-e520, 2019 09.
Article in English | MEDLINE | ID: mdl-31253594

ABSTRACT

BACKGROUND: Primary effusion lymphoma (PEL) is a rare type of non-Hodgkin lymphoma. The limited disease-free survival after chemotherapy has resulted in a poor prognosis. The outcomes data for high-dose therapy followed by autologous hematopoietic cell transplantation (auto-HCT) for PEL are limited owing to the rarity of the disease. PATIENTS AND METHODS: The present study included 9 patients with PEL from 2 major academic centers. Of these patients, 4 had received auto-HCT after high-dose therapy. Of the 9 patients, 8 (89%) had immunodeficiency (7 with human immunodeficiency virus seropositivity; 1, a solid organ transplant recipient) at the diagnosis. Human herpesvirus-8 by immunohistochemistry was positive in 8 patients. Anthracycline-based combination chemotherapy was used as first-line treatment in 7 patients; 4 underwent auto-HCT after attaining first complete remission. RESULTS: The median follow-up of the surviving patients was 25 months (95% confidence interval [CI], 8%-29%). The 2-year progression-free and overall survival for the 8 patients who had received treatment was 58% (95% CI, 22%-95%) and 73% (95% CI, 41%-100%), respectively. The 2-year progression-free and overall survival for the patients who had received auto-HCT was 50% (95% CI, 1%-99%) and 75% (95% CI, 33%-100%), respectively. Of the 4 auto-HCT recipients, all had been in first complete remission at the time of autografting. The cumulative incidence of relapse was 50% (95% CI, 19%-100%). No deaths were attributable to auto-HCT at 2 years after autografting. CONCLUSION: Despite the small sample size, our data have shown that consolidative auto-HCT is safe and effective and should be considered for eligible patients with PEL after demonstration of an objective response to induction chemotherapy. However, the high relapse rate remains a concern and warrants the development of new strategies to mitigate post-transplantation relapse.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematopoietic Stem Cell Transplantation , Lymphoma, Primary Effusion/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biopsy , Combined Modality Therapy , Comorbidity , Disease Management , Female , Follow-Up Studies , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Immunohistochemistry , Immunophenotyping , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/mortality , Male , Middle Aged , Prognosis , Recurrence , Survival Analysis , Transplantation, Autologous , Treatment Outcome
7.
J Virol ; 92(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29743366

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) can cause several human cancers, including primary effusion lymphoma (PEL), which frequently occur in immunocompromised patients. KSHV-infected patients often suffer from polymicrobial infections caused by opportunistic bacterial pathogens. Therefore, it is crucial to understand how these coinfecting microorganisms or their secreted metabolites may affect KSHV infection and the pathogenesis of virus-associated malignancies. Quorum sensing (QS), a cell density-based intercellular communication system, employs extracellular diffusible signaling molecules to regulate bacterial virulence mechanisms in a wide range of bacterial pathogens, such as Pseudomonas aeruginosa, which is one of the most common opportunistic microorganisms found in immunocompromised individuals. In this study, we evaluated and compared the influence on PEL growth and the host/viral interactome of the major QS signaling molecules [N-(3-oxododecanoyl)-l-homoserine lactone (OdDHL), N-butyrylhomoserine lactone (BHL), and 2-heptyl-3-hydroxy-4-quinolone (PQS)] in conditioned medium from wild-type (wt) and QS mutant laboratory strains as well as clinical isolates of P. aeruginosa Our data indicate that P. aeruginosa coinfection may facilitate virus dissemination and establishment of new infection and further promote tumor development through effectively inducing viral lytic gene expression by its QS systems.IMPORTANCE Currently, most studies about KSHV infection and/or virus-associated malignancies depend on pure culture systems or immunodeficient animal models. However, the real situation should be much more complicated in KSHV-infected immunocompromised patients due to frequent polymicrobial infections. It is important to understand the interaction of KSHV and coinfecting microorganisms, especially opportunistic bacterial pathogens. Here we report for the first time that P. aeruginosa and its quorum-sensing signaling molecules display a complicated impact on KSHV-associated lymphoma growth as well as the intracellular host/viral gene expression profile. Our data imply that targeting of coinfecting pathogens is probably necessary during treatment of virus-associated malignancies in these immunocompromised patients.


Subject(s)
4-Butyrolactone/analogs & derivatives , Gene Expression Regulation , Herpesvirus 8, Human/physiology , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/pathology , Quinolones/metabolism , Quorum Sensing , 4-Butyrolactone/metabolism , Bacterial Load , Herpesviridae Infections/etiology , Herpesviridae Infections/metabolism , Herpesviridae Infections/pathology , Humans , Lymphoma, Primary Effusion/metabolism , Pseudomonas Infections/complications , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , Signal Transduction , Viral Proteins/genetics , Viral Proteins/metabolism , Virulence
8.
Int J Hematol ; 108(3): 329-334, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29516290

ABSTRACT

Primary effusion lymphoma (PEL) is a rare type of extranodal lymphoma, typically of a B-cell origin, which presents as lymphomatous effusion with no nodal enlargement or tumor masses. The development PEL is universally associated with human herpes virus-8 (HHV-8) infection. Cases of HHV-8-negative primary lymphomatous effusion have recently been reported and referred to as HHV-8-unrelated PEL-like lymphoma. Some cases of this disease have been reported in iatrogenic immunocompromised patients. The mechanisms responsible for the inhibitory effects of the discontinuation of immunosuppressants other than methotrexate (MTX) against the disease, which have been demonstrated for MTX-associated lymphoproliferative disorders, have not yet been elucidated. We describe a case of PEL-like lymphoma that developed in the course of antisynthetase syndrome and was treated with tacrolimus. A single dose of systemic chemotherapy did not improve lymphomatous effusion, whereas the discontinuation of tacrolimus resulted in the long-term remission of this disease.


Subject(s)
Epstein-Barr Virus Infections/complications , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/etiology , Myositis/complications , Tacrolimus/administration & dosage , Tacrolimus/adverse effects , Withholding Treatment , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Female , Herpesvirus 8, Human , Humans , Immunocompromised Host , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/pathology , Middle Aged , Prednisolone/administration & dosage , Treatment Outcome , Vincristine/administration & dosage
10.
Mol Cancer Ther ; 16(11): 2627-2638, 2017 11.
Article in English | MEDLINE | ID: mdl-28847988

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive subtype of non-Hodgkin lymphoma caused by Kaposi's sarcoma-associated herpesvirus (KSHV) infection. Currently, treatment options for patients with PEL are limited. Oncolytic viruses have been engineered as anticancer agents and have recently shown increased therapeutic promise. Similarly, lytic activation of endogenous viruses from latently infected tumor cells can also be applied as a cancer therapy. In theory, such a therapeutic strategy would induce oncolysis by viral replication, while simultaneously stimulating an immune response to viral lytic cycle antigens. We examined the combination of the FDA-approved drug ingenol-3-angelate (PEP005) with epigenetic drugs as a rational therapeutic approach for KSHV-mediated malignancies. JQ1, a bromodomain and extra terminal (BET) protein inhibitor, in combination with PEP005, not only robustly induced KSHV lytic replication, but also inhibited IL6 production from PEL cells. Using the dosages of these agents that were found to be effective in reactivating HIV (as a means to clear latent virus with highly active antiretroviral therapy), we were able to inhibit PEL growth in vitro and delay tumor growth in a PEL xenograft tumor model. KSHV reactivation was mediated by activation of the NF-κB pathway by PEP005, which led to increased occupancy of RNA polymerase II onto the KSHV genome. RNA-sequencing analysis further revealed cellular targets of PEP005, JQ1, and the synergistic effects of both. Thus, combination of PEP005 with a BET inhibitor may be considered as a rational therapeutic approach for the treatment of PEL. Mol Cancer Ther; 16(11); 2627-38. ©2017 AACR.


Subject(s)
Azepines/administration & dosage , Diterpenes/administration & dosage , Lymphoma, Primary Effusion/drug therapy , Sarcoma, Kaposi/therapy , Triazoles/administration & dosage , Animals , Cell Line, Tumor , DNA Replication/drug effects , Herpesvirus 8, Human/drug effects , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/pathogenicity , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/genetics , Lymphoma, Primary Effusion/virology , Mice , NF-kappa B/genetics , Oncolytic Virotherapy/methods , Oncolytic Viruses/drug effects , Oncolytic Viruses/genetics , Oncolytic Viruses/pathogenicity , Sarcoma, Kaposi/complications , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/virology , Virus Replication/drug effects , Xenograft Model Antitumor Assays
11.
Ann Clin Lab Sci ; 47(3): 344-348, 2017 May.
Article in English | MEDLINE | ID: mdl-28667038

ABSTRACT

Post-transplant lymphoproliferative disorders (PTLD) are emergent complications of organ transplantation occurring in 2% to 10% of transplanted patients. Epstein-Barr virus (EBV) infections are considered the most important factors for the development of these heterogeneous disorders. Primary effusion lymphoma (PEL) is a lymphoproliferative disorder predominantly described in patients with advanced AIDS and it is almost universally associated with human herpesvirus 8 (HHV8). In rare case, PEL also occurs in HHV8-negative patient, in the setting of hepatitis B and C virus infection. However, all these cases showed pan B-cell markers to be positive. Here, we report a case of PTLD presented as HHV8-negative and HIV-negative primary effusion lymphoma lacking near all lymphoid markers except PAX5 on immunohistochemistry, which created a diagnostic challenge. The diagnosis requires multiple approaches including molecular and genetic tests.


Subject(s)
Liver Transplantation , Lymphoma, Primary Effusion/genetics , Lymphoproliferative Disorders/genetics , Chromosomes, Human, Pair 14 , Chromosomes, Human, Pair 8 , Humans , Immunohistochemistry , Liver Transplantation/adverse effects , Lymphoma, Primary Effusion/etiology , Lymphoproliferative Disorders/etiology , Male , Middle Aged , PAX5 Transcription Factor/metabolism , Translocation, Genetic
12.
Cell Host Microbe ; 22(1): 61-73.e7, 2017 Jul 12.
Article in English | MEDLINE | ID: mdl-28704654

ABSTRACT

The human tumor viruses Epstein-Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) establish persistent infections in B cells. KSHV is linked to primary effusion lymphoma (PEL), and 90% of PELs also contain EBV. Studies on persistent KSHV infection in vivo and the role of EBV co-infection in PEL development have been hampered by the absence of small animal models. We developed mice reconstituted with human immune system components as a model for KSHV infection and find that EBV/KSHV dual infection enhanced KSHV persistence and tumorigenesis. Dual-infected cells displayed a plasma cell-like gene expression pattern similar to PELs. KSHV persisted in EBV-transformed B cells and was associated with lytic EBV gene expression, resulting in increased tumor formation. Evidence of elevated lytic EBV replication was also found in EBV/KSHV dually infected lymphoproliferative disorders in humans. Our data suggest that KSHV augments EBV-associated tumorigenesis via stimulation of lytic EBV replication.


Subject(s)
Coinfection , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/pathogenicity , Herpesvirus 8, Human/physiology , Herpesvirus 8, Human/pathogenicity , Neoplasms/virology , Animals , B-Lymphocytes/virology , Cell Line, Tumor , Cytokines/blood , DNA, Viral/analysis , Disease Models, Animal , Epstein-Barr Virus Infections/blood , Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/virology , Genes, Viral/genetics , Herpesviridae Infections/blood , Herpesviridae Infections/immunology , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , High-Throughput Nucleotide Sequencing , Humans , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/virology , Mice , Spleen/pathology , Spleen/virology , Survival Rate , Virus Replication
14.
Cancer Res Treat ; 49(1): 274-278, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27283030

ABSTRACT

Primary effusion lymphoma (PEL) is a rare type of non-Hodgkin's lymphoma arising from a B-cell lineage characterized by the formation of malignant effusion in body cavities without evidence of a detectable tumor. The effusion contains tumor cells universally infected with human herpesvirus 8 (HHV8), which is the critical factor differentiating PEL from HHV8-unrelated PEL-like lymphoma (PEL-LL). This report describes a 77-year-old male patient with pleural effusion and ascites, containing lymphoma cells expressing a B-cell phenotype, but without markers of HHV8 in immunocytochemical analysis. The patient was diagnosed with PEL-LL and treated with six cycles of rituximab plus cyclophosphamide, doxorubicin, vincristine, and prednisolone (R-CHOP), which resulted in a complete remission. The patient is currently disease-free 15 months post-treatment. To the best of our knowledge, this is the first report on administration of R-CHOP in a PEL-LL patient in South Korea.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Herpesviridae Infections/complications , Herpesvirus 8, Human , Lymphoma, Primary Effusion/drug therapy , Lymphoma, Primary Effusion/etiology , Age Factors , Aged , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biomarkers , Biopsy , Cyclophosphamide/adverse effects , Cyclophosphamide/therapeutic use , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Humans , Lymphoma, Primary Effusion/diagnosis , Male , Multimodal Imaging/methods , Prednisone/adverse effects , Prednisone/therapeutic use , Remission Induction , Rituximab , Treatment Outcome , Vincristine/adverse effects , Vincristine/therapeutic use
15.
Leukemia ; 31(3): 614-624, 2017 03.
Article in English | MEDLINE | ID: mdl-27538487

ABSTRACT

Primary effusion lymphoma (PEL) is an incurable malignancy that develops in immunodeficient patients as a consequence of latent infection of B-cells with Kaposi's sarcoma-associated herpes virus (KSHV). Malignant growth of KSHV-infected B cells requires the activity of the transcription factor nuclear factor (NF)-κB, which controls maintenance of viral latency and suppression of the viral lytic program. Here we show that the KSHV proteins K13 and K15 promote NF-κB activation via the protease mucosa-associated lymphoid tissue lymphoma translocation protein-1 (MALT1), a key driver of NF-κB activation in lymphocytes. Inhibition of the MALT1 protease activity induced a switch from the latent to the lytic stage of viral infection, and led to reduced growth and survival of PEL cell lines in vitro and in a xenograft model. These results demonstrate a key role for the proteolytic activity of MALT1 in PEL, and provide a rationale for the pharmacological targeting of MALT1 in PEL therapy.


Subject(s)
Caspases/metabolism , Herpesviridae Infections/complications , Herpesviridae Infections/virology , Herpesvirus 8, Human/physiology , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/pathology , Neoplasm Proteins/metabolism , Virus Latency , Animals , Biomarkers , Caspases/genetics , Cell Line , Cell Survival/genetics , Disease Models, Animal , Enzyme Activation , Flow Cytometry , Gene Silencing , Host-Pathogen Interactions , Humans , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , NF-kappa B/metabolism , Neoplasm Proteins/genetics , Protein Binding , Viral Proteins/metabolism , Virus Activation , Xenograft Model Antitumor Assays
16.
Clin Lymphoma Myeloma Leuk ; 16 Suppl: S175-80, 2016 08.
Article in English | MEDLINE | ID: mdl-27521315

ABSTRACT

BACKGROUND: Primary effusion lymphoma (PEL) is a rare malignancy usually associated with HIV infection. Management and outcomes are poorly understood. METHODS: The medical records of all patients diagnosed with HIV-associated PEL at our institution between 1999 and 2014 were reviewed. Patients were followed till death, treatment failure or loss of follow-up. RESULTS: Twelve patients with PEL were identified during the 15 year study period; 9 had HIV infection. All 9 were male; median age was 45 years. All presented with local symptoms and were diagnosed with PEL a median of 11 years after HIV diagnosis. Location was pleural (3), pericardial (3), peritoneal (1) and extracavitatory (2). By definition, all had Ann Arbor stage 4 at diagnosis. Median follow-up was 34 months. Two patients had poor performance status and were unable to get chemotherapy. Seven patients had a complete remission (CR) and two died within 1 month of diagnosis. The median CD4 levels at PEL diagnosis in patients with poor versus good outcomes were 54 cells/mm3 (range, 26-82 cells/mm3) and 211 cells/mm3 (range, 73-800 cells/mm3). In contrast, the median lactate dehydrogenase (LDH) levels at PEL diagnosis with poor versus good prognosis were 1074 U/L (range, 703-1445 U/L) and 283 U/L (range, 156-760 U/L). CONCLUSIONS: Given its rarity, our knowledge of PEL relies solely on case reports and case series. Prompt HAART and chemotherapy may be effective in HIV- associated PEL and good outcomes are possible. LDH and CD4 may be possible prognostic factors in PEL.


Subject(s)
HIV Infections/complications , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/therapy , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antiretroviral Therapy, Highly Active , CD4 Lymphocyte Count , Comorbidity , HIV Infections/drug therapy , HIV Infections/immunology , HIV Infections/virology , Humans , Immunohistochemistry , Lymphoma, Primary Effusion/mortality , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Retrospective Studies , Treatment Outcome , Viral Load
17.
Pediatr Blood Cancer ; 63(9): 1674-6, 2016 09.
Article in English | MEDLINE | ID: mdl-27186682

ABSTRACT

Primary effusion lymphoma (PEL) is a rare lymphoma that occurs more frequently in immunocompromised adults and has a poor survival. We report a 9-year-old female with combined immunodeficiency with an Epstein-Barr virus positive/human herpes virus 8 negative PEL-like lymphoma. The treatment with systemic chemotherapy for non-Hodgkin lymphoma, zidovudine, and interferon-α failed to control disease progression. This is the first reported pediatric case of PEL-like lymphoma. Increased diagnostic awareness and more effective treatment strategies are needed for this rare lymphoma.


Subject(s)
Immunologic Deficiency Syndromes/complications , Lymphoma, Primary Effusion/etiology , Child , Female , Humans , Immunologic Deficiency Syndromes/congenital , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/drug therapy
18.
Clin Lymphoma Myeloma Leuk ; 16(6): 311-21, 2016 06.
Article in English | MEDLINE | ID: mdl-27234438

ABSTRACT

BACKGROUND: Primary effusion lymphoma (PEL) is a rare non-Hodgkin lymphoma subtype primarily seen in human immunodeficiency virus (HIV)-infected individuals with low CD4(+) cell counts and elevated HIV viral loads. It has always been associated with human herpesvirus type 8 (HHV-8) and in 80% of cases has also been associated with Epstein-Barr virus (EBV). Less commonly, PEL has presented in patients with advanced age and other conditions associated with an altered immunity, including malignancy, liver cirrhosis, and immunosuppressive medications. It is a tumor of B-cell lineage; however, it shows a "null" phenotype, rarely expressing pan-B cell surface antigens. It will usually express CD45, CD30, CD38, CD138, and MUM1 and is characterized by lymphomatous effusions in body cavities but not lymphadenopathy. It is an aggressive lymphoma, with an average median survival of < 1 year. HHV-8-associated large B-cell lymphoma (HHV-8-LBL) is a second variant of PEL that is both solid and extracavitary. It has immunoblastic and/or anaplastic morphologic features and a distinct immunohistochemical staining pattern. It could also have a different clinical presentation than that of classic PEL. MATERIALS AND METHODS: We describe the case of a 57-year-old HIV-infected man who presented with a slow-growing and asymptomatic abdominal mass. Examination of an excisional biopsy specimen showed malignant large cells with prominent cytoplasm that were positive for pan-B cell antigen CD20, HHV-8, and EBV and negative for CD138, CD10, BCL-6, CD3, and CD30. The Ki-67 labeling index was 90%. The diagnosis was stage IIIA HHV-8-LBL, and he was treated with 6 cycles of R-EPOCH (rituximab, etoposide, vincristine, doxorubicin, cyclophosphamide, and prednisone) infusion chemotherapy. At 12 months after treatment, he was in complete remission. We also performed a Medline and Embase search to better understand the clinical findings of our patient and the unique attributes of HHV-8-LBL. Focusing our search on English language studies, we identified 83 cases of HHV-8-LBL without an effusion component. We compared these 83 cases with 118 reported cases of classic PEL. RESULTS: The median age of the patients with HHV-8-LBL was 41 years (range, 24-77), and 96% of the cases were associated with HIV. The median age of the patients with classic PEL was 41 years (range, 26-86), and 96% of the cases were associated with HIV. Of those with HHV-8-LBL, 31 of 61 (51%) had a pre-existing diagnosis of acquired immunodeficiency syndrome (AIDS) and 47 of 63 (75%) were coinfected with EBV. In contrast, 69 of 96 patients (72%) with classic PEL had a pre-existing AIDS diagnosis and 40 of 49 (82%) were coinfected with EBV. The mean CD4(+) count of the HHV-8-LBL patients was 256 cells/µL (range, 18-1126 cells/µL) compared with 139 cells/µL (range, 2-557 cells/µL) in the classic PEL patients. The median survival time for both groups was similar at 5.5 months (range, 25 days to ≥ 25 months) for patients with HHV-8-LBL and 4 months (range, 2 days to ≥ 113 months) for those with classic PEL. More patients with HHV-8-LBL were alive at the last follow-up point (59% vs. 18%). The percentage of patients achieving complete remission was 54% (30 of 56) and 36% (32 of 89) for HHV-8-LBL and classic PEL, respectively. CONCLUSION: Our patient's high CD4(+) cell count, the lack of a pre-existing AIDS diagnosis, and the excellent response to chemotherapy highlights that HHV-8-LBL might have distinct clinical features and possibly a better response to chemotherapy than classic PEL. HHV-8-LBL should be included in the differential diagnosis of HIV patients with solid lesions. It is essential that patients' Centers for Disease Control and Prevention HIV clinical status and HIV viral load at the diagnosis of PEL and HHV-8-LBL be reported and that the reported clinical results include longer term follow-up data. Only then will a more complete clinical picture of this little-appreciated and little-understood PEL variant be defined.


Subject(s)
Herpesviridae Infections/complications , Herpesvirus 8, Human , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/etiology , Adult , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Biopsy , Coinfection , HIV Infections/complications , HIV Infections/diagnosis , HIV Infections/immunology , HIV Infections/virology , Homosexuality, Male , Humans , Immunohistochemistry , Immunophenotyping , Lymphoma, Primary Effusion/drug therapy , Male , Middle Aged , Neoplasm Staging , Positron Emission Tomography Computed Tomography , Tomography, X-Ray Computed , Treatment Outcome , Young Adult
19.
Leuk Lymphoma ; 57(7): 1625-32, 2016 07.
Article in English | MEDLINE | ID: mdl-26727336

ABSTRACT

Human herpesvirus-8-unrelated primary effusion lymphoma characterized by lymphomatous effusion without nodal lesions occasionally exhibits spontaneous remission. To elucidate the factors associated with a good prognosis, this study analyzed the clinical parameters of four patients treated in the department and 109 patients reported in case reports. The median age was 71 years and the median overall survival was 20 months. Patients possessing two independent favorable factors, an elderly status (≥ 70 years) and low serum lactate dehydrogenase (< 500 IU/L) showed a markedly higher 1-year survival than patients lacking either of the two factors in the absence of chemotherapy (94% vs 20%, p = 3 × 10(-5)), which was similarly observed in the chemotherapy group (94% vs 51%, p = 0.002). The use of rituximab was also a strong predictor of survival (89% vs 49%, p = 7 × 10(-6)). Elderly patients not exhibiting an increased lactate dehydrogenase may represent a benign sub-group of effusion lymphoma, which do not require chemotherapy to achieve remission.


Subject(s)
Herpesviridae Infections/complications , Herpesvirus 8, Human , Lactate Dehydrogenases/blood , Lymphoma, Primary Effusion/blood , Lymphoma, Primary Effusion/etiology , Age Factors , Aged , Aged, 80 and over , Biomarkers , Biopsy , Chromosome Aberrations , Chromosome Banding , Coinfection , Female , Herpesviridae Infections/virology , Humans , Immunophenotyping , Lymphoma, Primary Effusion/diagnosis , Lymphoma, Primary Effusion/drug therapy , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Risk Factors
20.
Viruses ; 6(9): 3472-86, 2014 Sep 19.
Article in English | MEDLINE | ID: mdl-25243371

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV, also named Human herpesvirus 8 HHV-8) is the cause of Kaposi sarcoma (KS), the most common malignancy in HIV-infected individuals worldwide, primary effusion lymphoma (PEL) and multicentric Castleman disease (MCD). KSHV is a double-stranded DNA virus that encodes several homologues of cellular proteins. The structural similarity between viral and host proteins explains why some viral homologues function as their host counterparts, but sometimes at unusual anatomical sites and inappropriate times. In other cases, structural modification in the viral proteins can suppress or override the function of the host homologue, contributing to KSHV-related diseases. For example, viral IL-6 (vIL-6) is sufficiently different from human IL-6 to activate gp130 signaling independent of the α subunit. As a consequence, vIL-6 can activate many cell types that are unresponsive to cellular IL-6, contributing to MCD disease manifestations. Here, we discuss the molecular biology of KSHV homologues of cellular products as conduits of virus/host interaction with a focus on identifying new strategies for therapy of KS and other KSHV-related diseases.


Subject(s)
Castleman Disease/etiology , Herpesviridae Infections/complications , Herpesviridae Infections/virology , Herpesvirus 8, Human/genetics , Lymphoma, Primary Effusion/etiology , Sarcoma, Kaposi/etiology , Cytokines/genetics , Cytokines/metabolism , Herpesviridae Infections/genetics , Humans , Inflammation Mediators/metabolism , NF-kappa B/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...