HIV-associated classical Hodgkin lymphoma (HIV-cHL) is an important complication of HIV disease in the era of effective combination antiretroviral therapy (cART). Generally, newly diagnosed HIV-cHL should be managed with curative intent. With modern HIV therapeutics, HIV-cHL treatment outcomes are largely comparable to those of the background population with cHL (non–HIV-cHL). To achieve these outcomes, particular attention must be given to managing HIV. This management includes understanding HIV as a comorbid condition with a spectrum of impact that is unique to each patient. Meticulous attention to drug-drug interactions is required to avoid toxicity and pharmacokinetic effects that can undermine cure. Relapsed and refractory HIV-cHL poses additional therapeutic challenges. The standard management in this setting should also be based on that for non–HIV-cHL, and includes the use of salvage chemotherapy followed by autologous stem cell transplant in chemosensitive disease. The role of allogeneic hematopoietic stem cell transplant is less clear but may be useful in select cases. Newer agents with activity in cHL are being tested as part of primary and salvage therapy and are also highly relevant for HIV-cHL.

Medscape Continuing Medical Education online

This activity has been planned and implemented in accordance with the Essential Areas and policies of the Accreditation Council for Continuing Medical Education through the joint providership of Medscape, LLC and the American Society of Hematology.

Medscape, LLC is accredited by the ACCME to provide continuing medical education for physicians.

Medscape, LLC designates this Journal-based CME activity for a maximum of 1.0 AMA PRA Category 1 Credit(s)™. Physicians should claim only the credit commensurate with the extent of their participation in the activity.

All other clinicians completing this activity will be issued a certificate of participation. To participate in this journal CME activity: (1) review the learning objectives and author disclosures; (2) study the education content; (3) take the post-test with a 75% minimum passing score and complete the evaluation at http://www.medscape.org/journal/blood; and (4) view/print certificate. For CME questions, see page 1355.

Disclosures

The authors, Associate Editor Jacob M. Rowe, and CME questions author Laurie Barclay, freelance writer and reviewer, Medscape, LLC, declare no competing financial interests.

Learning objectives
  1. Describe the epidemiology of human immunodeficiency virus (HIV)-associated classical Hodgkin lymphoma, based on a review.

  2. Distinguish the clinical presentation and diagnosis of HIV-associated classical Hodgkin lymphoma.

  3. Discuss the clinical management of HIV-associated classical Hodgkin lymphoma.

Release date: February 19, 2015; Expiration date: February 19, 2016

HIV-associated classical Hodgkin lymphoma (HIV-cHL) is a serious complication of HIV. As with Kaposi sarcoma and non-Hodgkin lymphomas (NHLs), cHL risk is substantially elevated by HIV infection. However, unlike those tumors, HIV-cHL does not confer a diagnosis of AIDS. HIV-cHL may present with extranodal involvement that can lead to life-threatening organ dysfunction. Nonetheless, outcomes comparable to those seen in the general population are possible with standard curative-intent therapy and modern combination antiretroviral therapy (cART). In this article, we discuss the epidemiology, pathobiology, and clinical management of HIV-cHL.

A 44-year-old man was referred with relapsed HIV-cHL. He initially presented 20 months prior with supraclavicular swelling, night sweats, and weight loss. Excisional lymph node biopsy demonstrated Epstein-Barr virus (EBV)-associated cHL, mixed cellularity (MC) subtype (Figure 1). HIV serology revealed the previously undiagnosed infection. CD4+ T-cell count (CD4+ count) was 140 cells per mm3. He commenced tenofovir, lamivudine, and ritonavir-boosted atazanavir for HIV, as well as trimethoprim/sulfamethoxazole for Pneumocystis pneumonia prophylaxis. He had no history of opportunistic infections (OIs). 18Fluorodeoxyglucose (18FDG) positron emission tomography (PET) revealed 18FDG uptake in lymph nodes and bones (Figure 2A). He had advanced-stage, unfavorable disease (stage IVB and International Prognosis Score of 4 based on gender, stage, hemoglobin level of 10 mg/dL, and albumin level of 2.0 mg/dL). He received doxorubicin, bleomycin, vinblastine, and dacarbazine (ABVD) with pegfilgrastim and continued cART. His first cycle was complicated by grade 4 neutropenic fever lasting 2 weeks. During subsequent cycles, vinblastine and dacarbazine were dose reduced by 50%. Additionally, there were dose delays during several cycles due to neutropenia, and the patient developed grade 2 neuropathy. 18FDG-PET after 2 cycles showed uptake in the left axilla. A biopsy sample taken in response to a solitary radiographic lymph node abnormality after 6 28-day cycles showed reactive changes. He completed 8 cycles and achieved a complete response (CR). After therapy, CD4+ count was 173 cells per mm3.

Figure 1

Histopathology and immunohistochemistry of HIV-cHL. (A) Hematoxylin and eosin staining shows cHL, MC subtype. Immunostaining for (B) CD15, (C) CD30, and (D) EBV latent membrane protein 1 demonstrates Hodgkin Reed-Sternberg cells. (E) CD68 staining showing many (>5%)46  macrophages. (F) Hematoxylin and eosin staining at time of relapse shows cHL, MC subtype. Original magnification ×40.

Figure 1

Histopathology and immunohistochemistry of HIV-cHL. (A) Hematoxylin and eosin staining shows cHL, MC subtype. Immunostaining for (B) CD15, (C) CD30, and (D) EBV latent membrane protein 1 demonstrates Hodgkin Reed-Sternberg cells. (E) CD68 staining showing many (>5%)46  macrophages. (F) Hematoxylin and eosin staining at time of relapse shows cHL, MC subtype. Original magnification ×40.

Close modal
Figure 2

18FDG-PET in HIV-cHL. (A) Baseline 18FDG-PET. Volumetric image shows bulky intensely hypermetabolic cervical, mediastinal, and axillary lymph nodes and multiple focal bone lesions (representative vertebral lesion, red arrow). (B) Interim 18FDG-PET. At the end of cycle 2, coronal image focuses on a small suspicious lesion in left axilla (red arrow); diffuse bone uptake attributable to pegfilgrastim is also noted. After cycle 6, a biopsy sample of residual abnormalities in the left axilla showed reactive changes and no evidence of cHL. (C) End-of-therapy 18FDG-PET. Volumetric image shows resolution of 18FDG avid nodes. (D) Relapse 18FDG-PET. Volumetric image shows left axillary avid lymph node (red arrow) and other small nodes above the diaphragm.

Figure 2

18FDG-PET in HIV-cHL. (A) Baseline 18FDG-PET. Volumetric image shows bulky intensely hypermetabolic cervical, mediastinal, and axillary lymph nodes and multiple focal bone lesions (representative vertebral lesion, red arrow). (B) Interim 18FDG-PET. At the end of cycle 2, coronal image focuses on a small suspicious lesion in left axilla (red arrow); diffuse bone uptake attributable to pegfilgrastim is also noted. After cycle 6, a biopsy sample of residual abnormalities in the left axilla showed reactive changes and no evidence of cHL. (C) End-of-therapy 18FDG-PET. Volumetric image shows resolution of 18FDG avid nodes. (D) Relapse 18FDG-PET. Volumetric image shows left axillary avid lymph node (red arrow) and other small nodes above the diaphragm.

Close modal

Twelve months after completing chemotherapy, a new axillary lymph node was palpated. Computerized tomography (CT) revealed a 2.5-cm axillary lymph node and other new, smaller lymph nodes. Excisional biopsy revealed recurrent EBV-positive (EBV+) cHL lymphoma, MC subtype. CD4+ count was 145 cells per mm3 and HIV viral load remained undetectable. 18FDG-PET revealed no extranodal disease (Figure 2D). We reasoned that drug-drug interactions between HIV protease inhibitor (PI)-based therapy and vinblastine complicated his initial treatment, and that although he had recurrent disease at 12 months, it was likely chemosensitive owing to initial response despite prior dose reductions and cycle delays. Therefore, to reduce drug-drug interactions, we modified his cART, replacing ritonavir and atazanavir with efavirenz. Given his prior intolerance to therapy, we elected to employ dose-adjusted etoposide, doxorubicin, vincristine, cyclophosphamide, prednisone, and filgrastim rather than conventional cHL salvage. We anticipated that vincristine would be less affected by efavirenz than would be the case with vinblastine, and that dose intensity could be adjusted based on the prior cycle. Due to the low-dose prolonged infusions, we anticipated minimal cardiac toxicity from additional doxorubicin. After 2 cycles, CT showed no disease, and he proceeded to autologous stem cell transplant (auto-SCT) following carmustine, etoposide, cytarabine, and melphalan conditioning. CD4+ count 3 months posttransplant was 186 cells per mm3. The patient has been followed on cART for 7 years with no relapse, undetectable HIV with CD4+ count rising to >500 cells per mm3, and resolution of his neuropathy.

cHL in patients with HIV

cHL incidence is approximately 50 per 100 000 person-years among HIV-infected individuals, fivefold to 20-fold higher than in the background population.1-4  Nodular lymphocyte–predominant HL does not have an established association with HIV, although cases in HIV-infected people have been noted.3  Unlike HIV-associated NHL (HIV-NHL), in which incidence decreased by 50% after introduction of cART (mainly owing to decreases in immunoblastic histologic types),5,6  HIV-cHL risk has remained stable and burden of disease has increased.2,6,7  In the United States, cHL follows NHL, Kaposi sarcoma, lung cancer, and anal cancer as the fifth commonest tumor in the setting of HIV.6  The median age of 40 to 44 years, which after correcting for age distribution of the at-risk population, is older than that of the general population.8  Indeed, in the United States, 14% of the cHL cases in this age group are HIV associated. Between 2000 and 2010, a large proportion of cHL in African Americans (17%) and Hispanics (10%) was attributed to HIV,3  reflecting shifting demographics of the HIV epidemic in the United States. In Johannesburg, South Africa, where HIV seroprevalence is 20% in young adults, 60% of cHL cases are HIV related.9  As with NHL,10-13  over time there has been a pathobiological shift in cHL subtypes in patients with HIV. Prior to cART, MC was the commonest subtype.14-17  In the cART era, likely due to improved immunity among HIV-infected populations,2  the nodular sclerosis subtype now accounts for nearly 50% of cases.3 

CD4+ counts, initiation of cART, and HIV-cHL

The association between HIV-cHL and CD4+ count is complex. Median CD4+ count at cHL diagnosis is between 150 and 250 cells per mm3; however, HIV-cHL risk is elevated at all CD4+ counts.2,4,13  Additionally, besides being a manifestation of untreated HIV, lymphocytopenia is also a common manifestation of cHL.18  In case-control studies of HIV patients matched for clinical features including time on cART, the decrease in median CD4+ count for those who developed HIV-cHL was 100 cells per mm3 over 12 months prior to cHL diagnosis compared to a median increase of 35 cells per mm3 in controls.4,19  Indeed, a precipitous drop in CD4+ count may be a heralding event of HIV-cHL.4,20 

The period of highest risk of HIV-cHL diagnosis is in the months after initiating cART.19-22  Data from a large US Department of Veterans Affairs cohort suggest that the incidence may be as high as 250 per 100 000 person-years during the first year of cART, with a slow decline over 10 years with controlled HIV, and a substantially lower risk thereafter.21,23  It has been speculated that a certain amount of immune competence is required for cHL to develop. Alternatively, with cART initiation and control of HIV viremia, an inflammatory condition may occur that promotes or unmasks existing cHL.24  It is not possible to identify those destined to develop HIV-cHL on the basis of CD4+ dynamics alone.23  Vigilance for inflammatory, infectious, and malignant etiologies of immune reconstitution syndromes in all HIV patients initiating cART is recommended. Prompt evaluation may lead to improved outcomes.

Hodgkin Reed-Sternberg (HRS) cells, which make up a minority of tumor cells in cHL, are clonal, mature B cells that express activation markers CD15 and CD30.25-28  Distinct histologic patterns in the inflammatory background form the basis for cHL subcategorization. In HIV-cHL, HRS cells are usually EBV+. These EBV-infected HRS cells exhibit a type II latency pattern in which expression of EBV-encoded genes is largely limited to Epstein-Barr nuclear antigen 1 and latent membrane proteins (LMP1, LMP2A, LMP2B). By in situ staining for EBV-encoded small RNA, up to 80% of nodular sclerosis and MC HIV-cHL cases are EBV-associated compared with <40% of non–HIV-cHL.16,26,29  EBV-encoded genes modulate cellular signaling pathways and contribute to pathogenesis. For example, LMP125,28,30,31  is a constitutively active homolog of CD40 that upregulates nuclear factor-kappa B signaling and is essential for B-cell transformation in vitro,32  whereas LMP2A mimics B-cell receptor signaling and also contributes to proliferation.33  Epstein-Barr nuclear antigen 1 appears to influence cytokine networks by stimulating the production of chemokines such as CXCL10 and CCL20,34,35  thus attracting T-regulatory cells and inhibiting an immune response against the EBV+ cells.

Gene expression profiling has informed the pathogenetic role of the tumor microenvironment in EBV+ cHL.36,37  Furthermore, EBV+ cHL tends to group with high-risk gene expression profiles in cHL. Although EBV infection of HRS cells is not the only factor leading to a high-risk molecular signature,38  modulation by EBV of host cellular pathways may promote high-risk features that typify the clinical presentation. Other unique features in HIV-cHL (compared with non–HIV-cHL) include decreased nodal CD4+ T-cells and lack of CD4+ rosetting around HRS cells, which may parallel low circulating CD4+ counts. CD8+ cell infiltrates are preserved; however, cytotoxic granzyme B expression is decreased compared with non–HIV-cHL,25,28,39-42  suggesting a role for defective EBV-specific immunity in disease pathogenesis.43  HIV itself may influence the nodal microenvironment and the CD4+ T-cell fluctuations after cART initiation, possibly contributing to HIV-cHL pathogenesis. Recruitment of reconstituted CCR4+CD4+ T cells by HRS secretion of CCL17 is a provocative possibility, but as yet, the associations with EBV-infected HRS remain unclear.44,45 

Tumor infiltrating macrophages (TAMs), recruited in association with CD115 expression by HRS cells,37  are increasingly recognized as important in the pathophysiology of cHL.46-48  TAMs are prominent in EBV-associated cHL,47,48  including HIV-associated cases. Interestingly, the number of TAMs in HIV-cHL does not appear to differ significantly with cART,39  which, among other factors, may help explain why HIV-cHL risk does not appear to be substantially reduced by cART.42 

Clinical presentation, diagnosis, and staging

HIV-cHL often presents with advanced disease. Extranodal sites of disease, including bone marrow, liver, and spleen, as well as B symptoms are more common compared with the background population16,49  and can occur in the absence of enlarged lymph nodes. Diagnosis is established with biopsy samples of involved tissue, usually a lymph node. HRS cells are identified by morphologic characteristics, and immunostaining demonstrates them to be CD15+, CD30+. PAX5-positive and EBV-encoded small RNA–positive HRS cells support the diagnosis.

Clinical evaluation includes a history and physical, complete blood count with differential, albumin level, liver and kidney function, baseline pulmonary function test for patients undergoing bleomycin-containing regimens, and 18FDG-PET with integrated or concurrent CT of the neck, chest, abdomen, and pelvis.50 18FDG-PET must be interpreted with caution because HIV viremia and certain OIs can cause 18FDG-avid nodes.51-53  Although not validated in the setting of HIV-cHL, 18FDG-PET is both sensitive and specific for detecting bone marrow involvement in cHL. Revised cHL staging criteria recommend omitting the bone marrow biopsy in non–HIV-cHL,54  and we recommend omitting it in most HIV-related cases as well. Nonetheless, bone marrow biopsy may be diagnostically useful in patients with cytopenias, because employing Occam’s razor to unify the clinical findings within a single diagnosis can lead to missed diagnoses in HIV-infected patients. For example, reactive hemophagocytic lymphohistiocytosis in HIV-infected patients can be attributed to HIV-cHL in up to 20% of cases,55  and bone marrow biopsy in this setting can provide important information about the etiology of cytopenias. In the absence of specific risk stratification for HIV-cHL, patients should be classified as having early favorable, early unfavorable, or advanced disease using criteria for non–HIV-cHL.

Evaluation of HIV status and comorbidities

Given the high proportion of cHL attributed to HIV, we recommend asking all adults with cHL for permission to test for HIV during the initial clinical evaluation. Treating cHL in a patient with unrecognized HIV infection introduces undue risk, including treatment-related death, whereas treating cHL and known HIV allows for optimal management of both diseases.

Comorbid HIV has a broad spectrum of severity that requires evaluation during initial treatment planning. CD4+ count, HIV viral load, and, in some cases, HIV genotyping are integral to this assessment. Additional HIV-specific medical history includes duration of infection, HIV treatment history and response to therapy as measured by CD4+ count and HIV viral load, and complications of HIV or its therapy. The state of health prior to cancer development provides critical information. At the healthy end, with high CD4+ count and undetectable virus, HIV may be nearly inconsequential for initial cHL management. At the opposite end, where there is HIV-drug resistance and advanced CD4+ T-cell depletion, severe AIDS complications may render standard chemotherapy inadvisable. With improvements in HIV medicine, the latter scenario is increasingly rare, and it is essential that this determination be based on expert knowledge.

Although many patients will have undetectable HIV viral loads when on cART, some will have HIV viremia resulting from either ineffective therapy or no therapy. Patients continue to be diagnosed with HIV/AIDS after presenting late to medical attention for a variety of reasons, including lack of access to care, social stigma, and fear. HIV diagnosis made during a malignancy evaluation often presents an opportunity to successfully engage such patients in medical care, as illustrated in the case presentation. Treatment-experienced patients may have uncontrolled HIV viremia for several reasons. HIV mutations, especially in patients with a complex history of cART use, may confer resistance to one or more antiretroviral agents. These are sometimes found in de novo infection as well. HIV resistance affects short-term and long-term management and requires expert evaluation including HIV genotyping.56  Lack of viral suppression due to cART nonadherence is important to recognize and address.

We evaluate degree of immunosuppression on the basis of CD4+ count, history of OIs, and CD4+ count nadir. Patients with CD4+ counts >350 cells per mm3 are less likely to have HIV-specific complications during cHL therapy; those with CD4+ counts between 100 and 350 cells per mm3 have increased risk that can be mitigated with antimicrobial prophylaxis; and those with CD4+ counts <100 per mm3 are at highest risk and require additional prophylaxis throughout treatment and follow-up (Table 1). A history of prior OIs or a low CD4+ count nadir may indicate a more vulnerable immune status than reflected in contemporary CD4+ counts. We employ regular surveillance for treatment-related events (ie, infections, mucositis) and have a low threshold to initiate evaluation for infectious complications in all HIV-infected patients, particularly those with prior OIs or a current or nadir CD4+ count of <200 cells per mm3. We evaluate CD4+ counts each cycle, then every 3 months for the first year following chemotherapy.

Table 1

Suggested OI prophylaxis in patients with HIV and cHL undergoing chemotherapy

OIThreshold for prophylaxisProphylactic regimens
Pneumocystis jirovecii pneumonia All patients during chemotherapy; continue after therapy until a CD4+ count >200 cells per mm3 is sustained for 3-6 mo. Preferred 
 Trimethoprim/sulfamethoxazole, 800/160-mg tablet (Bactrim DS) Monday, Wednesday, and Friday  
 Trimethoprim/sulfamethoxazole, 400/80-mg tablet daily 
Alternatives 
 Pentamadine, 300 mg in 6 mL of sterile water via nebulizer inhaler, once monthly 
 Atovaquone, 1500-mg suspension once daily 
 Dapsone, 100-mg tablet daily; mild hemolysis may be seen in patients with glucose-6-phosphate dehydrogenase deficiency 
 Trimethoprim/sulfamethoxazole and atovaquone may also prevent toxoplasmosis. 
Herpes simplex virus 1/2 All patients with history of oral or anogenital herpes simplex virus. Follow general recommendations for secondary prophylaxis starting 3-6 mo after completion of therapy.  Valacyclovir, 1000-mg tablet daily 
 Famciclovir, 500-mg tablet twice daily 
 Acyclovir, 400-mg tablet twice daily 
 These agents may also provide prophylaxis against varicella-zoster virus reactivation. 
Candida albicans (especially oral thrush) Consider primary prophylaxis when CD4+ count is <100 cells per mm3, or secondary prophylaxis with history of mucosal candidiasis. Alternatively, can be treated on earliest symptoms, especially when CD4+ count is >200 cells per mm3 Fluconazole, 200-mg tablet once daily (or 3 times/wk); also prevents cryptococcal disease in HIV patients with CD4+ counts <100 cells per mm3; do not administer the day before or the day of chemotherapy 
 Nystatin oral suspension, 5 mL “swish and swallow” 2-4 times daily (may be included in oral mouthwashes used for management of mucositis) 
Atypical mycobacteria (ie, Mycobacterium avium complex or Mycobacterium intracellulareCD4+ count <100 cells per mm3. Consider a higher current CD4+ threshold in patients with recent history of a nadir <100 cells per mm3 or history of atypical mycobacterial infection.  Azithromycin, 1200 mg weekly 
OIThreshold for prophylaxisProphylactic regimens
Pneumocystis jirovecii pneumonia All patients during chemotherapy; continue after therapy until a CD4+ count >200 cells per mm3 is sustained for 3-6 mo. Preferred 
 Trimethoprim/sulfamethoxazole, 800/160-mg tablet (Bactrim DS) Monday, Wednesday, and Friday  
 Trimethoprim/sulfamethoxazole, 400/80-mg tablet daily 
Alternatives 
 Pentamadine, 300 mg in 6 mL of sterile water via nebulizer inhaler, once monthly 
 Atovaquone, 1500-mg suspension once daily 
 Dapsone, 100-mg tablet daily; mild hemolysis may be seen in patients with glucose-6-phosphate dehydrogenase deficiency 
 Trimethoprim/sulfamethoxazole and atovaquone may also prevent toxoplasmosis. 
Herpes simplex virus 1/2 All patients with history of oral or anogenital herpes simplex virus. Follow general recommendations for secondary prophylaxis starting 3-6 mo after completion of therapy.  Valacyclovir, 1000-mg tablet daily 
 Famciclovir, 500-mg tablet twice daily 
 Acyclovir, 400-mg tablet twice daily 
 These agents may also provide prophylaxis against varicella-zoster virus reactivation. 
Candida albicans (especially oral thrush) Consider primary prophylaxis when CD4+ count is <100 cells per mm3, or secondary prophylaxis with history of mucosal candidiasis. Alternatively, can be treated on earliest symptoms, especially when CD4+ count is >200 cells per mm3 Fluconazole, 200-mg tablet once daily (or 3 times/wk); also prevents cryptococcal disease in HIV patients with CD4+ counts <100 cells per mm3; do not administer the day before or the day of chemotherapy 
 Nystatin oral suspension, 5 mL “swish and swallow” 2-4 times daily (may be included in oral mouthwashes used for management of mucositis) 
Atypical mycobacteria (ie, Mycobacterium avium complex or Mycobacterium intracellulareCD4+ count <100 cells per mm3. Consider a higher current CD4+ threshold in patients with recent history of a nadir <100 cells per mm3 or history of atypical mycobacterial infection.  Azithromycin, 1200 mg weekly 

We screen for hepatitis B virus (HBV) and hepatitis C virus (HCV) with serology for hepatitis B surface antigen, antibody to hepatitis B core, antibody to hepatitis B surface antigen, and antibody to HCV. Patients with a positive hepatitis B surface antigen require an HBV viral load and treatment of HBV. We monitor persons who test positive for antibody to hepatitis B core closely for signs of liver disease. For positive HCV serology, we evaluate HCV RNA viral load. Sensitivity of HCV serology is suboptimal in HIV-infected patients, with 13% of seronegative patients having evidence of HCV on RNA testing. Therefore, HCV RNA testing is also advised in HCV-seronegative patients with a history of intravenous drug use, aspartate aminotransferase/alanine aminotransferase abnormalities, and/or thrombocytopenia.57  Given the advances in curative HCV therapy, we refer HCV-infected patients for treatment after completion of chemotherapy.

Primary chemotherapy for HIV-cHL

Historically, absent cART, long-term HIV-cHL outcomes were poor. As with HIV-NHL, low-dose chemotherapy strategies were sometimes used58,59 ; however, this method did not improve survival, and low-dose chemotherapy approaches are no longer appropriate. Adherence to chemotherapy dose and schedule in cHL therapy, even in the face of neutropenia (nonfebrile), is critical for optimizing the probability of cure for both non–HIV-cHL60  and HIV-cHL.61 

High cure rates in HIV-cHL in the cART era have recently been demonstrated. The German HIV-related Lymphoma Study Group prospectively evaluated a stage-adapted approach. Patients with early-stage favorable disease received 2 to 4 cycles of ABVD and involved-field radiation; patents with early unfavorable or advanced disease were largely treated with 6 to 8 cycles of standard BEACOPP (bleomycin, etoposide, cyclophosphamide, vincristine, procarbazine, and prednisone). Most received concurrent cART. The CR rates for patients with early favorable, early unfavorable, and advanced HIV-cHL were 96%, 100%, and 86%, respectively. The 2-year disease-free survival rate was 95% for early-stage disease and 89% for advanced-stage disease. The 2-year overall survival (OS) rate in the entire cohort was 90.7%.62  However, BEACCOPP was toxic; dose reductions and delays were common, and treatment-related mortality was 7%. Our approach is to avoid BEACOPP in HIV-cHL, with the possible exception of response-adapted therapeutic escalation. This approach is consistent with treatment practice for non–HIV-cHL in the United States, where BEACOPP is generally viewed as more likely to contribute to late treatment-related myelodysplastic syndrome or leukemia. Likewise, concerns for late radiotherapy effects, such as secondary cancers and cardiovascular disease, are also important for HIV-infected patients, a population potentially more vulnerable to these effects.

Drug-drug interactions must be taken into consideration when managing HIV-cHL. In a retrospective analysis, 2 Spanish study groups found potentially life-threatening adverse events with coadministration of the HIV-PI ritonavir, the backbone of many commonly used PI-based cART regimens. Of 61 patients treated with ABVD and ritonavir-based cART, 10% died of treatment-related infections and 41% required dose delays.63  Subsequently, ritonavir, through strong CYP3A4 inhibition, has been demonstrated to exert pharmacokinetic effects on vinblastine resulting in severe neutropenia and neuropathy.64,65  In contrast, a recent retrospective comparison of 93 HIV-infected and 131 HIV-unrelated patients treated with ABVD from 1997 to 2010 found similar outcomes in the 2 populations. What distinguishes this report from the previous is that 92% of HIV-infected patients received a PI-sparing cART regimen. Both groups received comparable cHL treatment. HIV-infected patients also received appropriate OI prophylaxis. Only 1 toxic death was noted among HIV-infected patents. Five-year disease-free survival and OS rates in HIV-infected vs uninfected patients, respectively, were 85% vs 87% and 81% vs 88%, with no statistically significant differences between groups. HIV was not a predictor for OS after correcting for International Prognosis Score.59 

Taken together, we currently recommend treating HIV-cHL as one would treat non–HIV-cHL, with careful management of HIV. The optimal care for the patient with HIV-cHL should be guided by current approaches for early favorable, early unfavorable, and advanced disease in the non-HIV setting. Accordingly, in the United States, ABVD remains the general standard-of-care first-line therapy for both early and advanced non–HIV-cHL, and existing evidence, although limited, generally supports ABVD as a standard in HIV-cHL.66,67  For bulky stage I and II disease in non–HIV-cHL, radiotherapy is commonly employed; however, there is interest in reducing or eliminating radiotherapy for at least some of these patients,68  and this treatment approach is the focus of ongoing National Cancer Institute (NCI)-sponsored trials. In our opinion, granulocyte colony-stimulating factors,69  especially for patients with CD4+ counts <200 cells per mm3, should be used more liberally than in HIV-unrelated cases for which primary neutropenic fever prophylaxis is not recommended.

Approach to cART and supportive care

We either continue cART or initiate cART as early as possible when managing patients with HIV-cHL. This practice is informed by ex-perience, because there are no prospective data to inform optimal timing of cART initiation in this setting. Available data raise the possibility of inadequate CR rates when concurrent cART is not employed.59,70-72  Nonetheless, when the only option is to use cART that negatively impacts the ability to administer curative-intent chemotherapy, we will defer cART until completion of treatment of HIV-cHL based on experience in treating HIV-NHL.12,73  With the increasing availability of antiretroviral agents and the option of several cHL regimens, this tactic has become increasingly unnecessary. In the setting of hepatic or renal dysfunction, we consider temporarily suspending or deferring cART to minimize polypharmacy. Drug-drug interactions may be particularly complicated in patients with low CD4+ counts and concurrent infections requiring treatment or in those with limited cART options.

For symptomatic patients with advanced HIV-cHL, starting chemotherapy as soon as possible is usually desired and often leads to improvement in both performance status and tumor-associated organ dysfunction. An appropriate cART regimen can be started after initial administration of ABVD. Occasionally, unanticipated adverse events occur when administering chemotherapy and concurrent cART. In such cases, we prioritize cHL in treatment decisions and avoid chemotherapy delays. If necessary, cART can be suspended. After chemotherapy, we may modify cART on the basis of patient and HIV-related factors. In addition to managing HIV, monitoring for OIs (especially oral candidiasis) and select use of OI prophylaxis (Table 1) are required in treating HIV-cHL. Optimal outcomes are fostered by collaborative teams with expertise in infectious disease, pharmacology, social support, and patient education. Additional specialists may be required.

A basic principle we follow is to eliminate, where possible, concomitant medications (including antiretroviral drugs) that constrain the ability to administer full-dose and on-schedule chemotherapy for cHL. Facilitating this approach are more than 35 US Food and Drug Administration–approved antiretroviral drugs and combined-pill combinations (Table 2). Regimens generally include a 3-drug combination of 2 NRTIs and 1 drug from another class. We avoid older NRTIs due to their excess toxicity and believe that ritonavir, used alone or as a pharmacologic booster, and other strong CYP3A4 inhibitors such as cobicistat are essentially contraindicated for coadministration during ABVD. We also avoid other PIs due to CYP3A4-inhibitory properties.61  Preferred cART regimens have minimal expected CYP3A4 effects and employ agents with low stand-alone toxicity. At present, a regimen of an integrase strand transfer inhibitor (dolutegravir or raltegravir) or certain nonnucleoside reverse transcriptase inhibitors (rilpivirine) along with an NRTI combination of emtricitabine and tenofovir (Truvada) or abacavir and lamivudine (Epzicom), is likely to have minimal clinically relevant adverse effects on cancer therapy for HIV-cHL. Additional regimens are feasible but require consideration of agents or combinations not listed in Table 2. In patients treated with cART, adherence should be assessed each cycle. We use a simple 7-day recall tool74,75  that entails asking the number of missed cART doses over the last 7 days. For those reporting less than 100% adherence, evaluation of the underlying reasons for nonadherence is required. We recommend HIV viral-load monitoring at each cycle until viral suppression is documented in patients initiating or changing cART and in patients with possible nonadherence.

Table 2

Select antiretroviral therapy agents with level AI evidence for treating HIV, potential CYP3A4 interactions, and other considerations for concurrent use with chemotherapy

Class/drugsDoseCYP3A4 interactionsSide effectsOther considerations
First-choice agents* 
 NRTIs   
  Emtricitabine/tenofovir (Truvada) 1 tablet (200 mg/300 mg) daily — Nephrotoxicity (<3%) Dose adjust for creatinine clearance <50 mL/min 
Dual anti-HBV activity, preferred in HBV coinfected patients 
AI NRTI in several NNRTI-, PI-, and INSTI-based regimens 
  Abacavir/lamivudine (Epzicom) 1 tablet (600 mg/300 mg) daily — Life-threatening hypersensitivity reactions in patients with HLA-B57*01 allele Pharmacogenomic testing for HLA-B57*01 required before use of abacavir 
Avoid Epzicom if creatinine clearance <50 mL/min; instead, use renally dosed individual agents 
AI NRTI in combination with dolutegravir 
 INSTIs    
  Dolutegravir (Tivicay) 1 tablet (50 mg) daily — Elevated AST/ALT Metabolized by UGT1A1; dose increase required with efavirenz, rifampin, or select ritonavir-based combinations, and possibly other UGT1A1 inducers 
Increase dose to 50 mg twice daily for patients with certain INSTI-related mutations 
AI in combination with dual NRTIs above 
  Raltegravir (Isentress) 1 tablet (400 mg) twice daily — Elevated AST/ALT Metabolized by UGT1A1; dose increase required with rifampin 
Elevated creatine phosphokinase AI in combination with emtricitabine/tenofovir 
Additional second-choice options 
 NNRTIs   
  Rilpivirine (Edurant) 1 tablet (25 mg) daily Weak induction Depressed mood, insomnia (<10%) Contraindicated in combination with strong CYP3A4 inducers or proton pump inhibitors 
AI in combination with emtricitabine/tenofovir (once-a-day combination; Complera) in patients with HIV viral load <100 000 copies per mL and CD4+ count >200 cells per mm3 
  Efavirenz (Sustiva; also included in once-a-day combination with emtricitabine/tenofovir (Atripla) 1 tablet (600 mg) daily; in Atripla, 1 tablet (600/300/ 200 mg) daily Strong induction Depressed mood (5%) and increased rate of suicidality CYP3A4 metabolized; dose adjust if used in combination with voriconazole or rifampin 
Nervous system symptoms (headache, insomnia, dizziness) <30%, general resolve within 2-4 wk AI as Atripla, or in combination with abacavir/lamivudine in patients with HIV viral load <100 000 copies per mL 
Rash (10%-15%)  
Contraindicated§ 
 PIs     
  Darunavir (Prezista) boosted by ritonavir (Norvir) 1 tablet (800 mg) daily combined with ritonavir
1 tablet (100 mg) daily 
Strong inhibition Gastrointestinal symptoms (10%-20%) AI in combination with emtricitibine/tenofovir 
Dyslipidemia (20%-25%) Increased dose recommended for cART-experienced patients or those with specific HIV mutations 
AST/ALT abnormalities (12%) Contraindicated with strong CYP3A4 inducers 
Rash (6%)  
  Atazanavir (Reyataz) boosted by ritonavir (Norvir) 1 tablet (300 mg) daily combined with ritonavir
1 tablet (100 mg) daily 
Strong inhibition Hyperbilirubinemia (44%) A1 in combination with NRTIs above 
Gastrointestinal symptoms (<5%) Take with food 
Dyslipidemia (24%) Concurrent strong CYP3A4 inducers contraindicated 
Rash (5%-7%) Atazanavir is strong UGT1A1 inhibitor, contraindicated with irinotecan 
 Cobicistat-boosted once-a-day regimens   
  Elvitegravir/cobicistat/emtricitabine/tenofovir (Stribild) 1 tablet (150 mg/150 mg/200 mg/300 mg) daily Strong inhibition Gastrointestinal symptoms (10%-20%) Take with food 
Headache (7%) Do not initiate if creatinine clearance <70 mL/min 
Nephrotoxicity (10%) Concurrent strong CYP3A4 inducers contraindicated 
Class/drugsDoseCYP3A4 interactionsSide effectsOther considerations
First-choice agents* 
 NRTIs   
  Emtricitabine/tenofovir (Truvada) 1 tablet (200 mg/300 mg) daily — Nephrotoxicity (<3%) Dose adjust for creatinine clearance <50 mL/min 
Dual anti-HBV activity, preferred in HBV coinfected patients 
AI NRTI in several NNRTI-, PI-, and INSTI-based regimens 
  Abacavir/lamivudine (Epzicom) 1 tablet (600 mg/300 mg) daily — Life-threatening hypersensitivity reactions in patients with HLA-B57*01 allele Pharmacogenomic testing for HLA-B57*01 required before use of abacavir 
Avoid Epzicom if creatinine clearance <50 mL/min; instead, use renally dosed individual agents 
AI NRTI in combination with dolutegravir 
 INSTIs    
  Dolutegravir (Tivicay) 1 tablet (50 mg) daily — Elevated AST/ALT Metabolized by UGT1A1; dose increase required with efavirenz, rifampin, or select ritonavir-based combinations, and possibly other UGT1A1 inducers 
Increase dose to 50 mg twice daily for patients with certain INSTI-related mutations 
AI in combination with dual NRTIs above 
  Raltegravir (Isentress) 1 tablet (400 mg) twice daily — Elevated AST/ALT Metabolized by UGT1A1; dose increase required with rifampin 
Elevated creatine phosphokinase AI in combination with emtricitabine/tenofovir 
Additional second-choice options 
 NNRTIs   
  Rilpivirine (Edurant) 1 tablet (25 mg) daily Weak induction Depressed mood, insomnia (<10%) Contraindicated in combination with strong CYP3A4 inducers or proton pump inhibitors 
AI in combination with emtricitabine/tenofovir (once-a-day combination; Complera) in patients with HIV viral load <100 000 copies per mL and CD4+ count >200 cells per mm3 
  Efavirenz (Sustiva; also included in once-a-day combination with emtricitabine/tenofovir (Atripla) 1 tablet (600 mg) daily; in Atripla, 1 tablet (600/300/ 200 mg) daily Strong induction Depressed mood (5%) and increased rate of suicidality CYP3A4 metabolized; dose adjust if used in combination with voriconazole or rifampin 
Nervous system symptoms (headache, insomnia, dizziness) <30%, general resolve within 2-4 wk AI as Atripla, or in combination with abacavir/lamivudine in patients with HIV viral load <100 000 copies per mL 
Rash (10%-15%)  
Contraindicated§ 
 PIs     
  Darunavir (Prezista) boosted by ritonavir (Norvir) 1 tablet (800 mg) daily combined with ritonavir
1 tablet (100 mg) daily 
Strong inhibition Gastrointestinal symptoms (10%-20%) AI in combination with emtricitibine/tenofovir 
Dyslipidemia (20%-25%) Increased dose recommended for cART-experienced patients or those with specific HIV mutations 
AST/ALT abnormalities (12%) Contraindicated with strong CYP3A4 inducers 
Rash (6%)  
  Atazanavir (Reyataz) boosted by ritonavir (Norvir) 1 tablet (300 mg) daily combined with ritonavir
1 tablet (100 mg) daily 
Strong inhibition Hyperbilirubinemia (44%) A1 in combination with NRTIs above 
Gastrointestinal symptoms (<5%) Take with food 
Dyslipidemia (24%) Concurrent strong CYP3A4 inducers contraindicated 
Rash (5%-7%) Atazanavir is strong UGT1A1 inhibitor, contraindicated with irinotecan 
 Cobicistat-boosted once-a-day regimens   
  Elvitegravir/cobicistat/emtricitabine/tenofovir (Stribild) 1 tablet (150 mg/150 mg/200 mg/300 mg) daily Strong inhibition Gastrointestinal symptoms (10%-20%) Take with food 
Headache (7%) Do not initiate if creatinine clearance <70 mL/min 
Nephrotoxicity (10%) Concurrent strong CYP3A4 inducers contraindicated 

Appropriate HIV therapy generally consists of an INSTI, NNRTI, or PI combined with 2 NTRIs, often in the form of a combination tablet.

Potential drug-drug interaction considerations are underlined.

*

First choice antiretroviral agents in combination with chemotherapy regimens containing CYP3A4 substrates are those that are not metabolized via CYP3A4 system.

—; no significant effect on CYP3A4 metabolism of other drugs.

Strong level of evidence (AI) in antiretroviral-naïve patients on the basis of randomized controlled trials; recommendations are in concordance with the Department of Health and Human Services Guidelines for the Use of Antiretroviral Agents in HIV-1-Infected Adults and Adolescents.56 

Use of these second-choice options is somewhat more likely to be limited by potential drug-drug interactions during chemotherapy.

§

Contraindicated in combination with vinblastine and other chemotherapy drugs heavily dependent on CYP3A4 metabolism; they may be useful after completion of chemotherapy.

Anti-HBV, antibody to HBV; AST/ALT, aspartate aminotransferase/alanine aminotransferase; HLA, human leukocyte antigen; INSTI, integrase strand transfer inhibitor; NNRTI, nonnucleoside reverse-transcriptase inhibitor; NRTI, nucleoside reverse-transcriptase inhibitor; UGT1A1, uridine diphosphate glucuronosyltransferase 1 polypeptide A1.

Treatment of relapsed and refractory HIV-cHL

The standard of care for relapsed cHL in HIV-uninfected patients is salvage therapy followed by high-dose chemotherapy and auto-SCT. HIV-infected patients have been shown to successfully undergo this modality.76-79  Treatment-related mortality is 3% to 5% across several conditioning regimens, and long-term outcomes in reported series appear comparable to HIV-negative patients.76-80  It is unclear whether late toxic events, such as secondary myelodysplastic syndrome, are different than in the background population.79 

We continue to individualize relapsed therapy according to the treatment history. The case presented here illustrates this philosophy. Currently, 2 clinical trials being conducted by the Blood and Marrow Clinical Trials Network (cosponsored by the NCI and the National Heart, Lung, and Blood Institute) for HIV-infected patients with hematologic cancers should be strongly considered for a patient such as the one highlighted here. One trial is evaluating auto-SCT; the other, allotransplant using preferentially (but not required) homologous CCR5-Δ32 HIV coreceptor donors, possibly rendering the new immune system impervious to HIV. These studies will provide feasibility data and insights into transplant options for HIV-cHL and other hematologic malignancies. Insights may also be gained into HIV reservoirs important toward efforts in HIV eradication. Given the near-universal EBV association, we consider performing allogeneic donor searches early for patients with relapsed or refractory HIV-cHL to expedite potential enrollment in allotransplant studies.

Ongoing efforts to decrease toxicity and improve survival in non–HIV-cHL and HIV-cHL are likely to lead to therapeutic advances for both populations. We strongly recommend inclusion of HIV-cHL patients in clinical trials of novel agents and allogeneic transplant to promote advances in the field.81  In HIV-cHL, it is not yet possible to identify those for whom ABVD is likely to fail. Improved prognostic and predictive markers are needed. Response-adapted therapy on the basis of early 18FDG-PET findings has been the subject of recent study in non–HIV-cHL. Negative and positive interim 18FDG-PET scans after 2 cycles of ABVD in advanced-stage non–HIV-cHL have been associated with 95% and 12% 2-year progression-free survival rates, respectively.82-84  The 18FDG-PET results can be falsely positive in HIV, sometimes necessitating confirmation of positive findings. Cycle-1 negative PET scan results may be even more useful in identifying those for whom more limited therapy can be given.85  Although FDG-PET imaging in HIV-cHL has not been validated, preliminary data suggest a high negative predictive value.86  Potentially increased false-positive rates in HIV-cHL may lead to less useful predictive values for positive interim scans. Therefore, strategies using negative 18FDG-PET results for treatment reduction may be the more fruitful research undertaking in HIV-cHL.

Novel therapeutics in primary and second-line cHL therapy is an area of active investigation. Brentuximab vedotin, a CD30-directed immunoconjugate of the antimitotic agent monomethyl auristatin E was approved by the US Food and Drug Administration for refractory cHL.87  Phase 3 studies evaluating the combination of brentuximab vedotin, doxorubicin, vinblastine, and dacarbazine88  as upfront therapy are underway in non–HIV-cHL. The AIDS Malignancy Consortium in collaboration with the NCI Cancer Therapy Evaluation Program is also testing this combination in HIV-cHL (NCT01771107). It is hopeful that outcomes of these studies will advance therapy regardless of HIV serostatus. Many additional immune modulatory (ie, checkpoint inhibition with anti–programmed-death 1 agents) and epigenetic approaches are currently being evaluated in cHL. Advances in these areas and improved understanding of disease biology will likely lead to future paradigm shifts in both non–HIV-cHL and HIV-cHL.

We thank Karen Aleman, Kathleen Wyvill, and the clinical staff at the National Institutes of Health Clinical Center for providing patient care; Stefania Pittaluga for histopathology consultation and images; Corina Millo and Millie Whatley for consultation and nuclear medicine images; and Alice Pau for consultation on antiretroviral pharmacology and guidelines.

This work was supported by the Intramural Research Program of the National Institutes of Health and the Cancer Therapy Evaluation Program of the National Cancer Institute.

Contribution: T.S.U. and R.F.L. wrote the manuscript, provided patient care, reviewed the case presentation, and performed the literature review; T.S.U. compiled the clinical data; and R.F.L. reviewed the clinical data.

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

Correspondence: Richard F. Little, 9609 Medical Center Dr, MSC 9739, Bethesda, MD 20892-9739; e-mail: richard_little@nih.gov.

1
Calabresi
 
A
Ferraresi
 
A
Festa
 
A
, et al. 
Brescia HIV Cancer Study Group
Incidence of AIDS-defining cancers and virus-related and non-virus-related non-AIDS-defining cancers among HIV-infected patients compared with the general population in a large health district of Northern Italy, 1999-2009.
HIV Med
2013
, vol. 
14
 
8
(pg. 
481
-
490
)
2
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
 
12
(pg. 
3786
-
3791
)
3
Shiels
 
MS
Koritzinsky
 
EH
Clarke
 
CA
Suneja
 
G
Morton
 
LM
Engels
 
EA
Prevalence of HIV Infection among U.S. Hodgkin lymphoma cases.
Cancer Epidemiol Biomarkers Prev
2014
, vol. 
23
 
2
(pg. 
274
-
281
)
4
Bohlius
 
J
Schmidlin
 
K
Boué
 
F
, et al. 
Collaboration of Observational HIV Epidemiological Research Europe
HIV-1-related Hodgkin lymphoma in the era of combination antiretroviral therapy: incidence and evolution of CD4⁺ T-cell lymphocytes.
Blood
2011
, vol. 
117
 
23
(pg. 
6100
-
6108
)
5
Besson
 
C
Goubar
 
A
Gabarre
 
J
, et al. 
Changes in AIDS-related lymphoma since the era of highly active antiretroviral therapy.
Blood
2001
, vol. 
98
 
8
(pg. 
2339
-
2344
)
6
Shiels
 
MS
Pfeiffer
 
RM
Gail
 
MH
, et al. 
Cancer burden in the HIV-infected population in the United States.
J Natl Cancer Inst
2011
, vol. 
103
 
9
(pg. 
753
-
762
)
7
Robbins
 
HA
Shiels
 
MS
Pfeiffer
 
RM
Engels
 
EA
Epidemiologic contributions to recent cancer trends among HIV-infected people in the United States.
AIDS
2014
, vol. 
28
 
6
(pg. 
881
-
890
)
8
Shiels
 
MS
Pfeiffer
 
RM
Engels
 
EA
Age at cancer diagnosis among persons with AIDS in the United States.
Ann Intern Med
2010
, vol. 
153
 
7
(pg. 
452
-
460
)
9
Wiggill
 
TM
Mantina
 
H
Willem
 
P
Perner
 
Y
Stevens
 
WS
Changing pattern of lymphoma subgroups at a tertiary academic complex in a high-prevalence HIV setting: a South African perspective.
J Acquir Immune Defic Syndr
2011
, vol. 
56
 
5
(pg. 
460
-
466
)
10
Little
 
RF
AIDS-related non-Hodgkin’s lymphoma: etiology, epidemiology, and impact of highly active antiretroviral therapy.
Leuk Lymphoma
2003
, vol. 
44
 
Suppl 3
(pg. 
S63
-
S68
)
11
Little
 
RF
Dunleavy
 
K
Update on the treatment of HIV-associated hematologic malignancies.
Hematology (Am Soc Hematol Educ Program)
2013
, vol. 
2013
 (pg. 
382
-
388
)
12
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
 
12
(pg. 
4653
-
4659
)
13
Gopal
 
S
Patel
 
MR
Yanik
 
EL
, et al. 
Temporal trends in presentation and survival for HIV-associated lymphoma in the antiretroviral therapy era.
J Natl Cancer Inst
2013
, vol. 
105
 
16
(pg. 
1221
-
1229
)
14
Frisch
 
M
Biggar
 
RJ
Engels
 
EA
Goedert
 
JJ
AIDS-Cancer Match Registry Study Group
Association of cancer with AIDS-related immunosuppression in adults.
JAMA
2001
, vol. 
285
 
13
(pg. 
1736
-
1745
)
15
Grulich
 
AE
Li
 
Y
McDonald
 
A
Correll
 
PK
Law
 
MG
Kaldor
 
JM
Rates of non-AIDS-defining cancers in people with HIV infection before and after AIDS diagnosis.
AIDS
2002
, vol. 
16
 
8
(pg. 
1155
-
1161
)
16
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
 
7
(pg. 
1758
-
1767
)
17
Hoffmann
 
C
Chow
 
KU
Wolf
 
E
, et al. 
Strong impact of highly active antiretroviral therapy on survival in patients with human immunodeficiency virus-associated Hodgkin’s disease.
Br J Haematol
2004
, vol. 
125
 
4
(pg. 
455
-
462
)
18
Hasenclever
 
D
Diehl
 
V
Armitage
 
JO
, et al. 
A prognostic score for advanced Hodgkin’s disease. International Prognostic Factors Project on Advanced Hodgkin’s Disease.
N Engl J Med
1998
, vol. 
339
 
21
(pg. 
1506
-
1514
)
19
Gotti
 
D
Danesi
 
M
Calabresi
 
A
, et al. 
Brescia HIV Cancer Study Group
Clinical characteristics, incidence, and risk factors of HIV-related Hodgkin lymphoma in the era of combination antiretroviral therapy.
AIDS Patient Care STDS
2013
, vol. 
27
 
5
(pg. 
259
-
265
)
20
Lanoy
 
E
Rosenberg
 
PS
Fily
 
F
, et al. 
FHDH-ANRS CO4
HIV-associated Hodgkin lymphoma during the first months on combination antiretroviral therapy.
Blood
2011
, vol. 
118
 
1
(pg. 
44
-
49
)
21
Kowalkowski
 
MA
Mims
 
MA
Day
 
RS
Du
 
XL
Chan
 
W
Chiao
 
EY
Longer duration of combination antiretroviral therapy reduces the risk of Hodgkin lymphoma: a cohort study of HIV-infected male veterans.
Cancer Epidemiol
2014
, vol. 
38
 
4
(pg. 
386
-
392
)
22
Simard
 
EP
Pfeiffer
 
RM
Engels
 
EA
Cumulative incidence of cancer among individuals with acquired immunodeficiency syndrome in the United States.
Cancer
2011
, vol. 
117
 
5
(pg. 
1089
-
1096
)
23
Kowalkowski
 
MA
Mims
 
MP
Amiran
 
ES
Lulla
 
P
Chiao
 
EY
Effect of immune reconstitution on the incidence of HIV-related Hodgkin lymphoma.
PLoS ONE
2013
, vol. 
8
 
10
pg. 
e77409
 
24
Gopal
 
S
Patel
 
MR
Achenbach
 
CJ
, et al. 
Lymphoma immune reconstitution inflammatory syndrome in the center for AIDS research network of integrated clinical systems cohort.
Clin Infect Dis
2014
, vol. 
59
 
2
(pg. 
279
-
286
)
25
Thompson
 
LD
Fisher
 
SI
Chu
 
WS
Nelson
 
A
Abbondanzo
 
SL
HIV-associated Hodgkin lymphoma: a clinicopathologic and immunophenotypic study of 45 cases.
Am J Clin Pathol
2004
, vol. 
121
 
5
(pg. 
727
-
738
)
26
Herbst
 
H
Steinbrecher
 
E
Niedobitek
 
G
, et al. 
Distribution and phenotype of Epstein-Barr virus-harboring cells in Hodgkin’s disease.
Blood
1992
, vol. 
80
 
2
(pg. 
484
-
491
)
27
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
 
4
(pg. 
1495
-
1505
)
28
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
 
7
(pg. 
2319
-
2326
)
29
Herndier
 
BG
Sanchez
 
HC
Chang
 
KL
Chen
 
YY
Weiss
 
LM
High prevalence of Epstein-Barr virus in the Reed-Sternberg cells of HIV-associated Hodgkin’s disease.
Am J Pathol
1993
, vol. 
142
 
4
(pg. 
1073
-
1079
)
30
Audouin
 
J
Diebold
 
J
Pallesen
 
G
Frequent expression of Epstein-Barr virus latent membrane protein-1 in tumour cells of Hodgkin’s disease in HIV-positive patients.
J Pathol
1992
, vol. 
167
 
4
(pg. 
381
-
384
)
31
Pallesen
 
G
Hamilton-Dutoit
 
SJ
Rowe
 
M
Young
 
LS
Expression of Epstein-Barr virus latent gene products in tumour cells of Hodgkin’s disease.
Lancet
1991
, vol. 
337
 
8737
(pg. 
320
-
322
)
32
Kaye
 
KM
Izumi
 
KM
Kieff
 
E
Epstein-Barr virus latent membrane protein 1 is essential for B-lymphocyte growth transformation.
Proc Natl Acad Sci USA
1993
, vol. 
90
 
19
(pg. 
9150
-
9154
)
33
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
 
10
(pg. 
3715
-
3721
)
34
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
 
1
(pg. 
195
-
204
)
35
Morales
 
O
Mrizak
 
D
François
 
V
, et al. 
Epstein-Barr virus infection induces an increase of T regulatory type 1 cells in Hodgkin lymphoma patients.
Br J Haematol
2014
, vol. 
166
 
6
(pg. 
875
-
890
)
36
Chetaille
 
B
Bertucci
 
F
Finetti
 
P
, et al. 
Molecular profiling of classical Hodgkin lymphoma tissues uncovers variations in the tumor microenvironment and correlations with EBV infection and outcome.
Blood
2009
, vol. 
113
 
12
(pg. 
2765
-
2775
)
37
Steidl
 
C
Diepstra
 
A
Lee
 
T
, et al. 
Gene expression profiling of microdissected Hodgkin Reed-Sternberg cells correlates with treatment outcome in classical Hodgkin lymphoma.
Blood
2012
, vol. 
120
 
17
(pg. 
3530
-
3540
)
38
Scott
 
DW
Chan
 
FC
Hong
 
F
, et al. 
Gene expression-based model using formalin-fixed paraffin-embedded biopsies predicts overall survival in advanced-stage classical Hodgkin lymphoma.
J Clin Oncol
2013
, vol. 
31
 
6
(pg. 
692
-
700
)
39
Kiyasu
 
J
Aoki
 
R
Tanaka
 
PY
, et al. 
FOXP3(+) regulatory and TIA-1(+) cytotoxic T lymphocytes in HIV-associated Hodgkin lymphoma.
Pathol Int
2012
, vol. 
62
 
2
(pg. 
77
-
83
)
40
Bosch Príncep
 
R
Lejeune
 
M
Salvadó Usach
 
MT
Jaén Martínez
 
J
Pons Ferré
 
LE
Alvaro Naranjo
 
T
Decreased number of granzyme B+ activated CD8+ cytotoxic T lymphocytes in the inflammatory background of HIV-associated Hodgkin’s lymphoma.
Ann Hematol
2005
, vol. 
84
 
10
(pg. 
661
-
666
)
41
Koulis
 
A
Trivedi
 
P
Ibrahim
 
H
Bower
 
M
Naresh
 
KN
The role of the microenvironment in human immunodeficiency virus-associated classical Hodgkin lymphoma.
Histopathology
2014
, vol. 
65
 
6
(pg. 
749
-
756
)
42
Hartmann
 
S
Jakobus
 
C
Rengstl
 
B
, et al. 
Spindle-shaped CD163+ rosetting macrophages replace CD4+ T-cells in HIV-related classical Hodgkin lymphoma.
Mod Pathol
2013
, vol. 
26
 
5
(pg. 
648
-
657
)
43
Birx
 
DL
Redfield
 
RR
Tosato
 
G
Defective regulation of Epstein-Barr virus infection in patients with acquired immunodeficiency syndrome (AIDS) or AIDS-related disorders.
N Engl J Med
1986
, vol. 
314
 
14
(pg. 
874
-
879
)
44
Ishida
 
T
Ishii
 
T
Inagaki
 
A
, et al. 
Specific recruitment of CC chemokine receptor 4-positive regulatory T cells in Hodgkin lymphoma fosters immune privilege.
Cancer Res
2006
, vol. 
66
 
11
(pg. 
5716
-
5722
)
45
Ma
 
Y
Visser
 
L
Roelofsen
 
H
, et al. 
Proteomics analysis of Hodgkin lymphoma: identification of new players involved in the cross-talk between HRS cells and infiltrating lymphocytes.
Blood
2008
, vol. 
111
 
4
(pg. 
2339
-
2346
)
46
Steidl
 
C
Lee
 
T
Shah
 
SP
, et al. 
Tumor-associated macrophages and survival in classic Hodgkin’s lymphoma.
N Engl J Med
2010
, vol. 
362
 
10
(pg. 
875
-
885
)
47
Kamper
 
P
Bendix
 
K
Hamilton-Dutoit
 
S
Honoré
 
B
Nyengaard
 
JR
d’Amore
 
F
Tumor-infiltrating macrophages correlate with adverse prognosis and Epstein-Barr virus status in classical Hodgkin’s lymphoma.
Haematologica
2011
, vol. 
96
 
2
(pg. 
269
-
276
)
48
Tan
 
KL
Scott
 
DW
Hong
 
F
, et al. 
Tumor-associated macrophages predict inferior outcomes in classic Hodgkin lymphoma: a correlative study from the E2496 Intergroup trial.
Blood
2012
, vol. 
120
 
16
(pg. 
3280
-
3287
)
49
Errante
 
D
Gabarre
 
J
Ridolfo
 
AL
, et al. 
Hodgkin’s disease in 35 patients with HIV infection: an experience with epirubicin, bleomycin, vinblastine and prednisone chemotherapy in combination with antiretroviral therapy and primary use of G-CSF.
Ann Oncol
1999
, vol. 
10
 
2
(pg. 
189
-
195
)
50
Cheson
 
BD
Pfistner
 
B
Juweid
 
ME
, et al. 
International Harmonization Project on Lymphoma
Revised response criteria for malignant lymphoma.
J Clin Oncol
2007
, vol. 
25
 
5
(pg. 
579
-
586
)
51
Brust
 
D
Polis
 
M
Davey
 
R
, et al. 
Fluorodeoxyglucose imaging in healthy subjects with HIV infection: impact of disease stage and therapy on pattern of nodal activation.
AIDS
2006
, vol. 
20
 
7
(pg. 
985
-
993
)
52
Scharko
 
AM
Perlman
 
SB
Pyzalski
 
RW
Graziano
 
FM
Sosman
 
J
Pauza
 
CD
Whole-body positron emission tomography in patients with HIV-1 infection.
Lancet
2003
, vol. 
362
 
9388
(pg. 
959
-
961
)
53
Sathekge
 
M
Maes
 
A
Kgomo
 
M
Pottel
 
H
Stolz
 
A
Van De Wiele
 
C
FDG uptake in lymph-nodes of HIV+ and tuberculosis patients: implications for cancer staging.
Q J Nucl Med Mol Imaging
2010
, vol. 
54
 
6
(pg. 
698
-
703
)
54
Barrington
 
SF
Mikhaeel
 
NG
Kostakoglu
 
L
, et al. 
Role of imaging in the staging and response assessment of lymphoma: consensus of the International Conference on Malignant Lymphomas Imaging Working Group.
J Clin Oncol
2014
, vol. 
32
 
27
(pg. 
3048
-
3058
)
55
Fardet
 
L
Lambotte
 
O
Meynard
 
JL
, et al. 
Reactive haemophagocytic syndrome in 58 HIV-1-infected patients: clinical features, underlying diseases and prognosis.
AIDS
2010
, vol. 
24
 
9
(pg. 
1299
-
1306
)
56
Panel on Antiretroviral Guidelines for Adults and Adolescents
 
Guidelines for the use of antiretroviral agents in HIV-1-infected adults and adolescents. Department of Health and Human Services. http://aidsinfo.nih.gov/contentfiles/adultandadolescentgl.pdf. Accessed June 1, 2014
57
Bharti
 
AR
Letendre
 
SL
Wolfson
 
T
, et al. 
Clinical variables identify seronegative HCV co-infection in HIV-infected individuals.
J Clin Virol
2011
, vol. 
52
 
4
(pg. 
328
-
332
)
58
Errante
 
D
Tirelli
 
U
Gastaldi
 
R
, et al. 
The Italian Cooperative Group on AIDS and Tumors (GICAT)
Combined antineoplastic and antiretroviral therapy for patients with Hodgkin’s disease and human immunodeficiency virus infection. A prospective study of 17 patients.
Cancer
1994
, vol. 
73
 
2
(pg. 
437
-
444
)
59
Levine
 
AM
Li
 
P
Cheung
 
T
, et al. 
Chemotherapy consisting of doxorubicin, bleomycin, vinblastine, and dacarbazine with granulocyte-colony-stimulating factor in HIV-infected patients with newly diagnosed Hodgkin’s disease: a prospective, multi-institutional AIDS clinical trials group study (ACTG 149).
J Acquir Immune Defic Syndr
2000
, vol. 
24
 
5
(pg. 
444
-
450
)
60
Boleti
 
E
Mead
 
GM
ABVD for Hodgkin’s lymphoma: full-dose chemotherapy without dose reductions or growth factors.
Ann Oncol
2007
, vol. 
18
 
2
(pg. 
376
-
380
)
61
Montoto
 
S
Shaw
 
K
Okosun
 
J
, et al. 
HIV status does not influence outcome in patients with classical Hodgkin lymphoma treated with chemotherapy using doxorubicin, bleomycin, vinblastine, and dacarbazine in the highly active antiretroviral therapy era.
J Clin Oncol
2012
, vol. 
30
 
33
(pg. 
4111
-
4116
)
62
Hentrich
 
M
Berger
 
M
Wyen
 
C
, et al. 
Stage-adapted treatment of HIV-associated Hodgkin lymphoma: results of a prospective multicenter study.
J Clin Oncol
2012
, vol. 
30
 
33
(pg. 
4117
-
4123
)
63
Xicoy
 
B
Ribera
 
JM
Miralles
 
P
, et al. 
Limited prognostic value of the International Prognostic Score in advanced stage human immunodeficiency virus infection-related Hodgkin lymphoma treated with the doxorubicin, bleomycin, vinblastine, and dacarbazine regimen.
Leuk Lymphoma
2009
, vol. 
50
 
10
(pg. 
1718
-
1720
)
64
Ezzat
 
HM
Cheung
 
MC
Hicks
 
LK
, et al. 
Incidence, predictors and significance of severe toxicity in patients with human immunodeficiency virus-associated Hodgkin lymphoma.
Leuk Lymphoma
2012
, vol. 
53
 
12
(pg. 
2390
-
2396
)
65
Corona
 
G
Vaccher
 
E
Spina
 
M
Toffoli
 
G
Potential hazard drug-drug interaction between boosted protease inhibitors and vinblastine in HIV patients with Hodgkin’s lymphoma.
AIDS
2013
, vol. 
27
 
6
(pg. 
1033
-
1035
)
66
Meyer
 
RM
Gospodarowicz
 
MK
Connors
 
JM
, et al. 
NCIC Clinical Trials Group; Eastern Cooperative Oncology Group
ABVD alone versus radiation-based therapy in limited-stage Hodgkin’s lymphoma.
N Engl J Med
2012
, vol. 
366
 
5
(pg. 
399
-
408
)
67
Viviani
 
S
Zinzani
 
PL
Rambaldi
 
A
, et al. 
Michelangelo Foundation; Gruppo Italiano di Terapie Innovative nei Linfomi; Intergruppo Italiano Linfomi
ABVD versus BEACOPP for Hodgkin’s lymphoma when high-dose salvage is planned.
N Engl J Med
2011
, vol. 
365
 
3
(pg. 
203
-
212
)
68
Meyer
 
RM
Hoppe
 
RT
Point/counterpoint: early-stage Hodgkin lymphoma and the role of radiation therapy.
Blood
2012
, vol. 
120
 
23
(pg. 
4488
-
4495
)
69
Kuritzkes
 
DR
Parenti
 
D
Ward
 
DJ
, et al. 
Filgrastim prevents severe neutropenia and reduces infective morbidity in patients with advanced HIV infection: results of a randomized, multicenter, controlled trial. G-CSF 930101 Study Group.
AIDS
1998
, vol. 
12
 
1
(pg. 
65
-
74
)
70
Berenguer
 
J
Miralles
 
P
Ribera
 
JM
, et al. 
Characteristics and outcome of AIDS-related Hodgkin lymphoma before and after the introduction of highly active antiretroviral therapy.
J Acquir Immune Defic Syndr
2008
, vol. 
47
 
4
(pg. 
422
-
428
)
71
Gérard
 
L
Galicier
 
L
Boulanger
 
E
, et al. 
Improved survival in HIV-related Hodgkin’s lymphoma since the introduction of highly active antiretroviral therapy.
AIDS
2003
, vol. 
17
 
1
(pg. 
81
-
87
)
72
Ribera
 
JM
Navarro
 
JT
Oriol
 
A
, et al. 
Prognostic impact of highly active antiretroviral therapy in HIV-related Hodgkin’s disease.
AIDS
2002
, vol. 
16
 
14
(pg. 
1973
-
1976
)
73
Dunleavy
 
K
Little
 
RF
Pittaluga
 
S
, et al. 
The role of tumor histogenesis, FDG-PET, and short-course EPOCH with dose-dense rituximab (SC-EPOCH-RR) in HIV-associated diffuse large B-cell lymphoma.
Blood
2010
, vol. 
115
 
15
(pg. 
3017
-
3024
)
74
Mannheimer
 
S
Friedland
 
G
Matts
 
J
Child
 
C
Chesney
 
M
The consistency of adherence to antiretroviral therapy predicts biologic outcomes for human immunodeficiency virus-infected persons in clinical trials.
Clin Infect Dis
2002
, vol. 
34
 
8
(pg. 
1115
-
1121
)
75
Mosam
 
A
Shaik
 
F
Uldrick
 
TS
, et al. 
A randomized controlled trial of highly active antiretroviral therapy versus highly active antiretroviral therapy and chemotherapy in therapy-naive patients with HIV-associated Kaposi sarcoma in South Africa.
J Acquir Immune Defic Syndr
2012
, vol. 
60
 
2
(pg. 
150
-
157
)
76
Krishnan
 
A
Molina
 
A
Zaia
 
J
, et al. 
Durable remissions with autologous stem cell transplantation for high-risk HIV-associated lymphomas.
Blood
2005
, vol. 
105
 
2
(pg. 
874
-
878
)
77
Spitzer
 
TR
Ambinder
 
RF
Lee
 
JY
, et al. 
Dose-reduced busulfan, cyclophosphamide, and autologous stem cell transplantation for human immunodeficiency virus-associated lymphoma: AIDS Malignancy Consortium study 020.
Biol Blood Marrow Transplant
2008
, vol. 
14
 
1
(pg. 
59
-
66
)
78
Re
 
A
Michieli
 
M
Casari
 
S
, et al. 
High-dose therapy and autologous peripheral blood stem cell transplantation as salvage treatment for AIDS-related lymphoma: long-term results of the Italian Cooperative Group on AIDS and Tumors (GICAT) study with analysis of prognostic factors.
Blood
2009
, vol. 
114
 
7
(pg. 
1306
-
1313
)
79
Balsalobre
 
P
Díez-Martín
 
JL
Re
 
A
, et al. 
Autologous stem-cell transplantation in patients with HIV-related lymphoma.
J Clin Oncol
2009
, vol. 
27
 
13
(pg. 
2192
-
2198
)
80
Bayraktar
 
UD
Ramos
 
JC
Petrich
 
A
, et al. 
Outcome of patients with relapsed/refractory acquired immune deficiency syndrome-related lymphoma diagnosed 1999-2008 and treated with curative intent in the AIDS Malignancy Consortium.
Leuk Lymphoma
2012
, vol. 
53
 
12
(pg. 
2383
-
2389
)
81
Persad
 
GC
Little
 
RF
Grady
 
C
Including persons with HIV infection in cancer clinical trials.
J Clin Oncol
2008
, vol. 
26
 
7
(pg. 
1027
-
1032
)
82
Gallamini
 
A
Hutchings
 
M
Rigacci
 
L
, et al. 
Early interim 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography is prognostically superior to international prognostic score in advanced-stage Hodgkin’s lymphoma: a report from a joint Italian-Danish study.
J Clin Oncol
2007
, vol. 
25
 
24
(pg. 
3746
-
3752
)
83
Hutchings
 
M
Loft
 
A
Hansen
 
M
, et al. 
FDG-PET after two cycles of chemotherapy predicts treatment failure and progression-free survival in Hodgkin lymphoma.
Blood
2006
, vol. 
107
 
1
(pg. 
52
-
59
)
84
Gallamini
 
A
Barrington
 
SF
Biggi
 
A
, et al. 
The predictive role of interim positron emission tomography for Hodgkin lymphoma treatment outcome is confirmed using the interpretation criteria of the Deauville five-point scale.
Haematologica
2014
, vol. 
99
 
6
(pg. 
1107
-
1113
)
85
Hutchings
 
M
Kostakoglu
 
L
Zaucha
 
JM
, et al. 
In vivo treatment sensitivity testing with positron emission tomography/computed tomography after one cycle of chemotherapy for Hodgkin lymphoma.
J Clin Oncol
2014
, vol. 
32
 
25
(pg. 
2705
-
2711
)
86
Okosun
 
J
Warbey
 
V
Shaw
 
K
, et al. 
Interim fluoro-2-deoxy-D-glucose-PET predicts response and progression-free survival in patients with Hodgkin lymphoma and HIV infection.
AIDS
2012
, vol. 
26
 
7
(pg. 
861
-
865
)
87
Younes
 
A
Gopal
 
AK
Smith
 
SE
, et al. 
Results of a pivotal phase II study of brentuximab vedotin for patients with relapsed or refractory Hodgkin’s lymphoma.
J Clin Oncol
2012
, vol. 
30
 
18
(pg. 
2183
-
2189
)
88
Younes
 
A
Connors
 
JM
Park
 
SI
, et al. 
Brentuximab vedotin combined with ABVD or AVD for patients with newly diagnosed Hodgkin’s lymphoma: a phase 1, open-label, dose-escalation study.
Lancet Oncol
2013
, vol. 
14
 
13
(pg. 
1348
-
1356
)
Sign in via your Institution