Abstract
The observation in 1981 of the emergence of Kaposi sarcoma (KS) among young men who had sex with men was one of the first harbingers of the HIV epidemic. With advances in HIV care, the incidence of HIV-associated KS (HIV+KS) has decreased over time in the United States. However, it remains a persistent malignancy among some HIV-infected populations and is one of the most common tumors in sub-Saharan Africa. Because of the relapsing and remitting nature of this cancer, patients with HIV+KS can experience significant, long-term, morbidity. Patients with severe HIV+KS may also have concurrent lymphoproliferative syndromes, malignancies, and/or infections that can contribute to mortality. Several chemotherapy agents were explored in clinical trials for HIV+KS during the early stage of the epidemic. As HIV+KS emerges with CD4 lymphopenia and immunodysregulation, T-cell–sparing options are important to consider. Here, we explore the pathogenesis of HIV+KS and the current evidence for immunotherapy and therapies that potentially target KS pathogenesis. This review provides the current landscape of therapies for HIV+KS and highlights management issues for patients with HIV and cancer.
INTRODUCTION
In July 1981, the CDC reported the emergence of an aggressive form of Kaposi sarcoma (KS) among 26 young men who had sex with men, which was in contrast to the known epidemiologic characteristics of KS at the time.1 KS was previously a rare tumor found predominantly among elderly men of Mediterranean origin (classic KS). A more aggressive form of KS was also seen in sub-Saharan Africa (endemic KS), and KS was known to develop in immunosuppressed patients (iatrogenic KS). KS in the setting of HIV infection and AIDS is known as epidemic KS (HIV+KS).2 KS rates are elevated 500-fold among people living with HIV (PLWH) compared with the general population,3 and from the beginning of the HIV epidemic, KS was considered an AIDS-defining tumor.2 Epidemiologic studies suggested that KS was caused by a second transmissible agent, but for some time, this agent remained elusive. In 1994, the causative agent of KS, Kaposi sarcoma herpesvirus (KSHV, also known as human herpesvirus 8), was identified.4 KSHV has since been shown to cause primary effusion lymphoma (PEL), a form of multicentric Castleman disease (MCD) and an inflammatory cytokine syndrome (KICS).5-8
CONTEXT
Key Objective
Forty years ago, physicians were battling the unusual emergence of Kaposi sarcoma (KS) among young individuals subsequently diagnosed with HIV. Over time, there have been significant advances in the management of HIV worldwide with corresponding decreases in the rates of KS.
Knowledge Generated
We present the evidence of cancer therapies studied for HIV-associated KS (HIV+KS). Despite available US Food and Drug Administration–approved chemotherapies, there are immediate and long-term toxicities that need to be considered. We address investigations and treatments where HIV+KS occurs with other KS herpesvirus–associated diseases. We address the rationale for chemotherapy-sparing agents that are subject to ongoing clinical trials for participants with HIV+KS.
Relevance
In the 21st century, HIV+KS remains persistent and severe and can contribute to morbidity and mortality among young individuals worldwide who face health disparities. Moreover, it can occur concurrently with other diseases caused by KS herpesvirus and they can complicate the diagnosis and management of KS.
Over the past 40 years since the original report from the CDC, advances in HIV therapy with prompt initiation of antiretroviral therapy (ART) at HIV diagnosis have led to decreases in the rates of KS among PLWH in the United States and other countries.9-12 Between 2008 and 2016, the HIV+KS incidence rates decreased 3.2% per year in data from a US cancer registry.13 However, KSHV prevalence remains high in men who have sex with men and KS remains persistent, particularly among young Black men with HIV in the United States.14 Furthermore, in sub-Saharan Africa, KS continues to be a leading cause of morbidity and mortality because of high rates of HIV and endemic KSHV infection. KS is the most common tumor in men overall in certain countries in sub-Saharan Africa.15,16
This narrative review specifically focuses on management of HIV+KS, as it may have a more aggressive course than HIV-negative forms. HIV+KS can occur with concurrent KSHV-associated conditions, which can present diagnostic challenges as these diseases can mimic infections in an immunocompromised population. We address evidence for the current therapies, management of KS with concurrent KSHV-associated processes, and ongoing studies for HIV+KS.
METHODS
We searched PubMed for clinical trials, meta-analyses, and guidelines of major professional societies on the management of HIV+KS. Response rates from seminal prospective clinical trials investigating oncologic treatments (including chemotherapy, targeted therapy, and immunotherapy) of HIV+KS were highlighted. Studies that are open for enrollment for HIV+KS were identified from ClinicalTrials.gov.
DISCUSSION/OBSERVATIONS
Pathophysiology
KSHV is a linear double-stranded DNA gammaherpesvirus.17 KSHV can infect a wide variety of cell types, including B cells, monocytes, and endothelial cells. KS consists of KSHV-infected vascular or lymphatic endothelial cells; there is still uncertainty as to the exact cell of origin.
Like other herpesviruses, KSHV has latent and lytic replication programs. During latency, a few genes are expressed, including those encoded by open reading frame (ORF)71 (v-FLIP), ORF72 (v-cyclin), and ORF73 (latency-associated nuclear antigen) and several microRNAs.18-20 During lytic replication, all genes are expressed, resulting in production and release of progeny virions and death of the host cell. In KS, only a small proportion of infected cells express lytic genes. However, there is evidence that expression of one lytic gene, ORF74, that encodes a constitutively active viral G-protein–coupled receptor (vGPCR) is particularly important in KS pathogenesis.21,22 vGPCR is constitutively active and leads to production of vascular endothelial growth factor (VEGF), interleukin (IL)-6, IL-8, and tumor necrosis factor alpha, which promote KS through autocrine and paracrine pathways.21-23
Various KSHV-encoded genes promote proliferation of infected cells and dysregulate host innate immune pathways, including apoptosis and cell cycle arrest. Some KSHV-encoded genes are mimics of human genes with angiogenic and inflammatory properties. KSHV-encoded K3 and K5 ubiquinate surface MHC-1 and contribute to downregulation, making infected cells invisible to effector T cells.24-26 A homolog of human IL-6, known as viral IL-6, encoded by K2, induces the JAK and STAT pathway, leading to increased expression of angiogenic markers.27-29 Several KSHV genes also upregulate human IL-6, which contributes to the systemic symptoms and multiorgan dysfunction seen in MCD and PEL.30,31 KSHV v-FLIP activates the nuclear factor–κB, which contributes to the pathogenesis of KS, PEL, and MCD.19
Although many human cancers are considered clonal, the question whether KS is a clonal neoplasm, an oligoclonal process, or a multifocal vascular proliferation is still not completely resolved. One group of investigators studying women with advanced KS concluded that KS was a clonal cancer on the basis of the patterns of methylation in different KS lesions in the same individual.32 However, another group using a similar technique noted that clonality was noted in some lesions but not others.33 A more recent study investigating KSHV transcripts in KS lesions from both sexes concluded that most individual KS lesions were either oligoclonal or monoclonal and that different lesions in the same patient represented different oligoclonal or clonal expansions.34 This study is consistent with KS being a multifocal disease in which individual lesions arise separately rather than by metastases. However, additional research is needed to address these issues. Whatever the resolution, from a clinical perspective, KS acts like other neoplastic processes with its invasion of various organs and the devastating outcome of HIV+KS if untreated.
Clinical Presentation
Although HIV+KS is typically observed in PLWH who have < 200 CD4+ T cells/µL, some patients present with or experience a recurrence of KS at higher CD4+ T-cell counts.35 Patients with HIV+KS, like those with other epidemiologic forms of KS, often present with pigmented cutaneous KS lesions. There may be a spectrum of clinical presentations. Patients may initially present with a small focus of lesions that can be managed as an outpatient, but patients can also present with severe manifestations of cutaneous and visceral diseases with systemic symptoms or laboratory abnormalities requiring hospitalization. The nature of initial presentation influences additional investigations and management.
Cutaneous KS often presents in the lower extremities as flat, ulcerated, and/or fungating lesions that are frequently associated with edema (Fig 1). Patients may develop woody edema that can persist even after KS treatment, and long-standing edema can lead to recurrent skin infections and issues with mobility. Cutaneous lesions are often disseminated in patients with profound CD4+ T-cell lymphopenia; however, even a few isolated cutaneous lesions can cause significant morbidity if they are on the feet, face, or eye. In severe cases, KS lesions may also present in the oral cavity as nodular lesions, lymph nodes, or any visceral organ, most commonly the GI tract or pulmonary systems. In some cases, bone lesions are seen on imaging that may be intermittently symptomatic. Patients who report respiratory or GI symptoms or present with anemia may benefit from bronchoscopy and/or endoscopy in visualizing lesions, staging the disease, and investigating for opportunistic infections, especially if they are severely immunosuppressed. Cross-sectional imaging such as computed tomography, where available, showing visceral KS may be useful to determine the stage of disease. However, these tests should not delay treatment of HIV+KS in the presence of cutaneous KS. Biopsies may be obtained from the skin or GI system to confirm KS. Diagnostic characteristics of KS on biopsy include the presence of endothelial markers, spindle cells, and positive KSHV latency–associated nuclear antigen staining on immunohistochemistry.17 KS lesions are vascular and prone to bleeding, and biopsy of the airways is not advisable.
Diagnosis of Concurrent KSHV-Associated Processes and KS-Immune Reconstitution Inflammatory Syndrome
The presence of systemic symptoms such as fevers, night sweats, effusions, adenopathy, and laboratory abnormalities such as low albumin, elevated C-reactive protein, or pancytopenia should also prompt concern for concurrent KSHV-associated processes (MCD and/or PEL) that are often underdiagnosed and can occur in tandem with HIV+KS.6 This can pose a diagnostic challenge as these conditions can mimic infection in an immunocompromised host and can affect treatment and outcomes. Up to 50% of patients with MCD may have concurrent KS,36 but the prevalence of other KSHV-associated diseases among patients presenting with HIV+KS is unknown. Circulating markers concerning for a concurrent KSHV-associated process include elevated KSHV levels and IL-6 and IL-10 cytokine levels.6,8,31 KSHV viral load levels in the blood in patients with HIV+KS alone are often not elevated as compared with other KSHV-associated diseases.37 A large study among PLWH and KSHV-associated diseases demonstrated that an undetectable KSHV viral load excludes a diagnosis of MCD.38 Imaging in these cases should include a positron emission tomography-computed tomography to assess avid lymph nodes or splenomegaly that is suggestive of MCD.39 Diagnosis of concurrent KSHV-associated processes may require expert pathology review. Lymph nodes should be evaluated for MCD, and effusions should be sent for cytology and flow cytometry to diagnose PEL (Fig 2). In the absence of these histologic diagnoses, patients with KS who have symptoms of inflammation may have KICS.
A proportion of patients with HIV+KS may experience KS immune reconstitution inflammatory syndrome (KS-IRIS) within the first 12 weeks after ART initiation, resulting in worsening of HIV+KS or the onset of undiagnosed KS.41,42 However, this is a diagnosis of exclusion as the inflammatory symptoms may mimic infections or other KSHV-associated disorders, which if not appropriately diagnosed can result in high mortality. There is no standard therapy for KS-IRIS; continuation of ART and initiation of chemotherapy may result in improved outcomes.42 Although steroids are known to treat tuberculosis-IRIS, there have been cases where steroids led to the unmasking of KSHV-associated conditions, including KS, and steroids should probably be avoided when possible in patients with KS-IRIS.43
Treatment
Staging and principles of therapy.
KS is a vascular multicentric tumor, and therefore, the standard nomenclature from other cancer subtypes is not helpful, including the term metastases. HIV+KS is staged according to the extent of disease, CD4 count, and presence of systemic illness.44 Patients with HIV+KS limited to the skin and/or lymph nodes and/or minimal oral disease are considered as having T0 disease, and patients with edema, ulceration, extensive oral disease, or visceral KS are considered as having T1 disease (Fig 3). A stage-stratified approach to the treatment of HIV+KS is feasible where patients with minimal disease receive ART alone and those with T1 disease and extensive cutaneous disease or who otherwise require rapid disease resolution receive ART and chemotherapy.45
In the early studies of paclitaxel and liposomal doxorubicin (LD) in HIV+KS, only between 40% and 70% of participants were on concurrent ART (Table 1). With current management guidelines, all patients with HIV+KS should be initiated on ART. Most patients will be started a once-a-day pill of an integrase-inhibitor based therapy for HIV care, which generally will not interact with standard chemotherapy agents. Where possible, a multidisciplinary approach including oncologists, HIV specialists, and pharmacists from both disciplines should be involved to coordinate concomitant medications to avoid drug-drug interactions and manage opportunistic infections or other complications.
TABLE 1.
KSHV cannot be eradicated, patients with KS cannot be cured, and the goal of therapy is long-term control. There are no set number of cycles of systemic treatment for HIV+KS management.69 The duration of therapy is determined by the response to therapy. Cutaneous lesions can be evaluated by the total number, area of lesions, and number of nodular lesions. If there are a small number of lesions, six cycles or less of treatment may be sufficient. For those with many lesions or involvement of visceral disease, the number of cycles will depend on the response. Patients may require intermittent therapy for years. Repeat imaging, bronchoscopy, and/or endoscopy may be required to determine the response of visceral lesions. Physicians experienced in treating KS rarely use local approaches, such as panretin gel or radiotherapy. Although radiotherapy may play a role in obstructive KS lesions or cases where there are no systemic options available, it can delay wound healing and cause long-lasting induration and fibrosis.69 Except for biopsy, there is no role for surgical intervention or amputation in the management of HIV+KS.
Chemotherapy.
Several chemotherapy options were investigated in HIV+KS during the early stage of the HIV epidemic. Initially, combination chemotherapy regimens including doxorubicin, bleomycin, and vincristine were used. These combination regimens identified doxorubicin as the mainstay of treatment. Pegylated LD is doxorubicin encapsulated in long-circulating stealth liposomes. This formulation is thought to reduce uptake within the reticuloendothelial system and prolong circulation times. One study noted higher doxorubicin levels in KS lesions in those receiving LD as compared with those administered standard doxorubicin.70 Randomized controlled trials in the late 1990s comparing doxorubicin, bleomycin, and vincristine with LD at a dose of 20 mg/m2 once every 2 or 3 weeks demonstrated higher overall response rates and lower toxicity among those receiving LD46-50 (Table 1).
Paclitaxel was approved for use in HIV+KS in 1997. Overall response rates from phase II studies were between 56% and 71%.52-54 One randomized study compared paclitaxel administered every 2 weeks and LD every 3 weeks in 73 participants.50 The overall response rate was not statistically different between these agents (56% in paclitaxel and 46% in LD, P = .49). In general, paclitaxel is associated with more side effects such as fatigue, alopecia, and neuropathy and is generally used after LD. An important recent randomized study from 11 resource-limited settings in Brazil and sub-Saharan Africa highlighted the superiority of paclitaxel to etoposide or bleomycin plus vincristine in patients receiving ART.55
LD and paclitaxel remain the most used first-line chemotherapy options in HIV+KS. If patients have the re-emergence of KS after cessation of these therapies, patients can be retreated with the same agent. LD has a US Food and Drug Administration black box warning for cardiotoxicity at cumulative doses > 550 mg/m2. This is extrapolated from patients with advanced breast cancer; however, a study using endomyocardial biopsy of 10 patients with HIV+KS did not identify myocardial damage at cumulative LD doses of 440-840 mg/m2.71 Nonetheless, physicians considering cumulative doses above 550 mg/m2 should be aware of the potential for cardiotoxicity and weigh the potential risks against the benefits.
Other chemotherapy agents, such as vinorelbine, gemcitabine, and etoposide, have been investigated in HIV+KS56-58 in single-site phase II studies with none or only a proportion of participants on ART. These agents are less effective than LD or paclitaxel but can be considered in specific situations for persistent or relapsed HIV+KS per the National Comprehensive Cancer Network guidelines.69
Immunomodulatory agents and targeted therapies.
Initially on the basis of studies showing that thalidomide had antiangiogenic activity, other immunomodulatory agents, such as pomalidomide and lenalidomide, have been investigated in HIV+KS.59-61 We now know that this class of agents target cereblon, an E3 ubiquitin ligase,72,73 and downstream effects include enhancement of CD4+ and CD8+ T-cell costimulation, enhancement of natural killer cell activity, and modulation of tumor necrosis-α, IL-6, and VEGF. Preclinical studies have also shown that pomalidomide reverses the virus-induced downregulation of immune surface markers including MHC-1, ICAM-1, and B7-2 in several PEL cell lines, rendering cells more visible to the immune system. In two prospective trials, the response rate to pomalidomide among HIV+ participants was 60% and the response rate to lenalidomide among HIV+ participants was 40% (Table 1).59,60 There were increases in CD4+ T-cell counts from baseline to the first 4 weeks in the pomalidomide study. Data from this study led to approval of pomalidomide by the US Food and Drug Administration for the treatment of HIV+ and HIV-negative KS. The US National Center Institute–funded AIDS Malignancy Consortium (AMC) has initiated a trial of pomalidomide to assess its feasibility in sub-Saharan Africa and is planning a confirmatory study within the United States.
KS is notable for its aberrant angiogenesis with components of the VEGF pathway expressed in KS lesions; therefore, several agents that target angiogenesis have been investigated. Results so far have been mixed. Bevacizumab alone led to response rates of 31% and a 1-year progression-free survival of 70%.63 Bevacizumab in combination with LD led to a response rate of 56%, but side effects such as hypertension and bleeding events were noted and led to treatment cessation in several participants.74 Tyrosine kinase inhibitors that target angiogenesis and other proliferative pathways that are involved in KS pathogenesis have also been investigated. A small study of sorafenib demonstrated a poor response rate of 29%.65 Imatinib, an inhibitor of c-kit and platelet-derived growth factor receptor signaling, led to a response rate of 50% in 10 participants.66,75 However, in a larger phase II study, a partial response was seen in only 33% of participants.
Bortezomib, a proteosome inhibitor that inhibits the nuclear factor–κB pathway, is licensed for use in multiple myeloma. Additionally, bortezomib has been shown to result in KSHV lytic activation, which may lead to direct tumor killing by viral-mediated lysis. In a recent dose-escalation study of bortezomib by the AMC of 17 participants, the overall response rate was 60%.64 Five of six participants at the highest dose experienced a partial response (83%). Although bortezomib is known to increase KSHV lytic replication and increases in circulating KSHV plasma DNA were noted at the highest dose level, there were no associated inflammatory symptoms.
Immunotherapy.
Among PLWH, CD4+ T-cell loss may lead to depletion of virus-specific T cells and allows for proliferation of virus-infected cells, leading to virus-associated cancers. Chronic T-cell stimulation by HIV, oncogenic viruses, and tumor neoantigens can lead to an increase in immune checkpoint markers on T cells, leading to loss of cytotoxic responses and eventual T-cell exhaustion.76 Immune checkpoint markers include programmed death-1, and in the past 10 years, checkpoint inhibitors have revolutionized cancer therapy. However, exclusion of PLWH from cancer clinical trials because of concerns of toxicity, HIV activation, and potentially poor responses have led to a lag in their study and use in PLWH.77 Checkpoint inhibitor therapy may offer a promising chemotherapy-sparing option that avoids treatment-related decreases in CD4+ T-cell counts that may perpetuate HIV+KS. A retrospective case series identified nine patients with HIV+KS treated with immunotherapy (eight received nivolumab and one received pembrolizumab), resulting in a response rate of 67% and others experiencing stable disease.78 Responses were seen in those with low CD4+ T-cell counts and visceral disease, including a complete response in one patient with GI disease.
The first prospective study of pembrolizumab in PLWH and cancer included participants with CD4+ T-cell counts > 100 cells/µL. Six participants with HIV+KS were enrolled,68 and some tumor regression was noted in five of the six; however, no response met the threshold for a partial response. One participant with heavy pretreatment for HIV+KS and a history of elevated peripheral blood mononuclear cell–associated KSHV developed significant anasarca, effusions, and substantial KSHV viremia after two cycles of pembrolizumab, leading to the participant's death. Autopsy findings showed substantial KSHV activation, but not pathologic findings of MCD. The trial investigators were concerned about the emergence of KSHV lymphoproliferation possibly attributed to pembrolizumab. In the ongoing prospective KS cohort within this study, participants with active MCD or KICS, anemia, or thrombocytopenia are excluded because of safety concerns.
Treating KS with concurrent KSHV-associated processes.
When HIV+KS occurs in the presence of another KSHV-associated disease or diseases, it is important to treat the concurrent KSHV processes in tandem (Fig 3). Among patients with MCD and KS, rituximab and LD lead to remission of both conditions.79,80 Rituximab, which is used to treat KSHV-MCD, is known to exacerbate KS and LD can thwart this. After resolution of MCD symptoms, LD can be continued alone for KS. KICS is an IL-6 syndrome that occurs in patients with KS and KSHV infection without MCD pathology. Although tocilizumab, an anti–IL-6 receptor antibody, has been studied in patients with MCD and shown to induce transient improvement in the majority of patients,81 its use and the role of other IL-6 inhibitors in KICS are unknown. The optimal therapy for KICS and KS has not been identified, and patients should be referred for clinical trials. In patients who do not respond to therapy of KS, KICS, or MCD, physicians should consider undiagnosed PEL. In patients with a diagnosis of PEL and KS with or without MCD, treatment should include treatment of the PEL with curative intent, generally with combination anthracycline–based chemotherapy regimens used for other B-cell non-Hodgkin lymphomas.7 Clinical trial options should be considered, especially for patients experiencing relapsed or refractory disease.
Clinical trial approaches and future directions.
Building on existing evidence from clinical trials, many of the ongoing studies in HIV+KS include chemotherapy-sparing options. Confirmatory studies are planned in the United States and sub-Saharan Africa for pomalidomide to determine feasibility and efficacy. The HIV and AIDS Malignancy Branch of the US National Cancer Institute is conducting an ongoing study of pomalidomide in combination with LD in severe cases of KS among HIV-positive and HIV-negative participants (Table 2). This study also explores the use of pomalidomide and LD where KS occurs with other KSHV-associated processes such as KICS or MCD as a rituximab-sparing alternative. The HIV and AIDS Malignancy Branch is also initiating a study of pomalidomide in combination with nivolumab in virus-associated cancers, including HIV+KS. Other combination immunotherapy studies that investigate advanced cancers in PLWH include cabozantinib and nivolumab as well as nivolumab and ipilimumab.
TABLE 2.
Following the data from the bortezomib study in HIV+KS, there is an ongoing AMC trial using ixazomib, an oral proteasome inhibitor, in HIV+KS. Also, on the basis of an in vitro study, sEphB4-HSA, a soluble human receptor tyrosine kinase ephrin type-B receptor 4 fused to human serum albumin, which targets blood vessel maturation, cell migration, and adhesion, is being investigated by the AMC in HIV+KS. Cytokines that are involved in T-cell development and proliferation (IL-7 and interleukin 12 fused to NHS76 antibody [NHS-IL12], a congener of IL-12) are also the subject of ongoing studies. IL-12 has antiangiogenic effects mediated through the induction of interferon-γ, which may lead to an increase in interferon-inducible protein 10 (IP-10). IP-10 can downregulate KSHV-encoded vGPCR, which is important in KS pathogenesis. Studies of IL-12 alone and in combination with LD have previously shown safety and activity in HIV+KS.82,83 NHS-IL12 is a novel immunocytokine that is composed of two IL-12 heterodimers, each fused to an NHS76 antibody, which targets regions of tumor necrosis where DNA has become exposed. The study of NHS-IL12 also investigates it in combination with bintrafusp-alfa, a programmed cell death ligand-1 inhibitor fused to the soluble extracellular domain of human transforming growth factor β receptor II.
Prognosis and Health Disparity Issues
HIV+KS is a cancer that now often presents at the time of an initial HIV diagnosis.14,84 Data from a UK specialist center have demonstrated good outcomes in HIV+KS with a 5-year overall survival of 85% even in poor-risk disease.45 Even so, the prognosis of HIV+KS is often poor in resource-poor regions or in individuals with poor access to health care. In sub-Saharan Africa, KS continues to be a leading cause of mortality among patients diagnosed with HIV.16 Within the United States, projecting current HIV diagnosis rates in the future, it is expected that one in two Black men who have sex with men and one in four Latino men who have sex with men will be diagnosed with HIV during their lifetime.85 These populations also have a high prevalence of KSHV infection and are at risk for KS. Such individuals face stigma related to their HIV status, race, and sexual orientation. As a result, they also experience significant challenges in access to health care and are under-represented in cancer clinical trials. Efforts are ongoing to ensure inclusion of PLWH in cancer clinical trials. Where possible, patients should receive multidisciplinary care among oncology and HIV specialists including nurses, social workers, and outreach staff who are aware of the complexity and stigma that PLWH and cancer face.
In conclusion, in the past 40 years, there have been substantial advances in the understanding of HIV, KSHV, and HIV+KS. Moreover, systemic therapies for HIV+KS with ART have led to improvements in care of patients worldwide. However, HIV+KS continues to occur in the modern age, causing significant morbidity and mortality particularly among populations experiencing cancer health disparities.
Ramya Ramaswami
Research Funding: Celgene (Inst), Genentech (Inst), EMD Serono (Inst), CTI BioPharma Corp (Inst), Merck Serono (Inst)
Kathryn Lurain
Research Funding: Celgene (Inst), Merck (Inst), EMD Serono (Inst), CTI BioPharma Corp (Inst), Miltenyi Biotec (Inst), Janssen Oncology (Inst)
Robert Yarchoan
Research Funding: Celgene (Inst), Merck (Inst), Genentech (Inst), CTI BioPharma Corp (Inst), Janssen Research & Development (Inst), EMD Serono (Inst), Bristol Myers Squibb/Celgene (Inst)
Patents, Royalties, Other Intellectual Property: Patent related to increase in immune surface markers by pomalidomide, lenalidomide, and related drugs in patients with KSHV-associated diseases, Various patents on vasostatin (I), KSHV vIL-6 (I), internalization of surface markers (I), calreticulin (I), and calreticulin fragments (I), Various patents including patents on anti-HIV therapies and soluble IL-2 receptor; mostly expired, Patents on the treatment of Kaposi sarcoma with IL-12, Patent(s) on the treatment of HIV and hepatitis virus with an antiviral drug and a vaccine to prevent resistance.
No other potential conflicts of interest were reported.
SUPPORT
Supported by the Intramural Research Program of the National Cancer Institute, National Institutes of Health.
AUTHOR CONTRIBUTIONS
Conception and design: All authors
Financial support: Robert Yarchoan
Administrative support: Robert Yarchoan
Collection and assembly of data: Ramya Ramaswami
Manuscript writing: All authors
Final approval of manuscript: All authors
Accountable for all aspects of the work: All authors
AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST
Oncologic Treatment of HIV-Associated Kaposi Sarcoma 40 Years on
The following represents disclosure information provided by authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.
Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).
Ramya Ramaswami
Research Funding: Celgene (Inst), Genentech (Inst), EMD Serono (Inst), CTI BioPharma Corp (Inst), Merck Serono (Inst)
Kathryn Lurain
Research Funding: Celgene (Inst), Merck (Inst), EMD Serono (Inst), CTI BioPharma Corp (Inst), Miltenyi Biotec (Inst), Janssen Oncology (Inst)
Robert Yarchoan
Research Funding: Celgene (Inst), Merck (Inst), Genentech (Inst), CTI BioPharma Corp (Inst), Janssen Research & Development (Inst), EMD Serono (Inst), Bristol Myers Squibb/Celgene (Inst)
Patents, Royalties, Other Intellectual Property: Patent related to increase in immune surface markers by pomalidomide, lenalidomide, and related drugs in patients with KSHV-associated diseases, Various patents on vasostatin (I), KSHV vIL-6 (I), internalization of surface markers (I), calreticulin (I), and calreticulin fragments (I), Various patents including patents on anti-HIV therapies and soluble IL-2 receptor; mostly expired, Patents on the treatment of Kaposi sarcoma with IL-12, Patent(s) on the treatment of HIV and hepatitis virus with an antiviral drug and a vaccine to prevent resistance.
No other potential conflicts of interest were reported.
REFERENCES
- 1.Centers for Disease Control (CDC) : Kaposi's sarcoma and Pneumocystis pneumonia among homosexual men—New York City and California. MMWR Morb Mortal Wkly Rep 30:305-308, 1981 [PubMed] [Google Scholar]
- 2.Yarchoan R, Uldrick TS: HIV-associated cancers and related diseases. N Engl J Med 378:1029-1041, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hernández-Ramírez RU, Shiels MS, Dubrow R, et al. : Cancer risk in HIV-infected people in the USA from 1996 to 2012: A population-based, registry-linkage study. Lancet HIV 4:e495-e504, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Chang Y, Cesarman E, Pessin MS, et al. : Identification of herpesvirus-like DNA sequences in AIDS-associated Kaposi’s sarcoma. Science 266:1865-1869, 1994 [DOI] [PubMed] [Google Scholar]
- 5.Nador RG, Cesarman E, Chadburn A, et al. : Primary effusion lymphoma: A distinct clinicopathologic entity associated with the Kaposi's sarcoma-associated herpes virus. Blood 88:645-656, 1996 [PubMed] [Google Scholar]
- 6.Ramaswami R, Lurain K, Polizzotto MN, et al. : Characteristics and outcomes of KSHV-associated multicentric Castleman disease with or without other KSHV diseases. Blood Adv 5:1660-1670, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Lurain K, Polizzotto MN, Aleman K, et al. : Viral, immunologic, and clinical features of primary effusion lymphoma. Blood 133:1753-1761, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Polizzotto MN, Uldrick TS, Wyvill KM, et al. : Clinical features and outcomes of patients with symptomatic Kaposi sarcoma herpesvirus (KSHV)-associated inflammation: Prospective characterization of KSHV inflammatory cytokine syndrome (KICS). Clin Infect Dis 62:730-738, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Peprah S, Engels EA, Horner M-J, et al. : Kaposi sarcoma incidence, burden and prevalence in United States people with HIV, 2000-2015. Cancer Epidemiol Biomarkers Prev 30:1627-1633, 2021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.INSIGHT START Study Group, Lundgren JD, Babiker AG, et al. : Initiation of antiretroviral therapy in early asymptomatic HIV infection. N Engl J Med 373:795-807, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Coghill AE, Pfeiffer RM, Shiels MS, et al. : Excess mortality among HIV-infected individuals with cancer in the United States. Cancer Epidemiol Biomarkers Prev 26:1027-1033, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Shiels MS, Islam JY, Rosenberg PS, et al. : Projected cancer incidence rates and burden of incident cancer cases in HIV-infected adults in the United States through 2030. Ann Intern Med 168:866-873, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Luo Q, Satcher Johnson A, Hall HI, et al. : Kaposi sarcoma rates among persons living with human immunodeficiency virus in the United States: 2008-2016. Clin Infect Dis, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.White DL, Oluyomi A, Royse K, et al. : Incidence of AIDS-related Kaposi sarcoma in all 50 United States from 2000 to 2014. J Acquir Immune Defic Syndr 81:387-394, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Semeere A, Wenger M, Busakhala N, et al. : A prospective ascertainment of cancer incidence in sub-Saharan Africa: The case of Kaposi sarcoma. Cancer Med 5:914-928, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bray F, Ferlay J, Soerjomataram I, et al. : Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin 68:394-424, 2018 [DOI] [PubMed] [Google Scholar]
- 17.Cesarman E, Damania B, Krown SE, et al. : Kaposi sarcoma. Nat Rev Dis Primers 5:9, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Schulz TF, Cesarman E: Kaposi sarcoma-associated herpesvirus: Mechanisms of oncogenesis. Curr Opin Virol 14:116-128, 2015 [DOI] [PubMed] [Google Scholar]
- 19.An J, Sun Y, Sun R, et al. : Kaposi's sarcoma-associated herpesvirus encoded vFLIP induces cellular IL-6 expression: The role of the NF-kappaB and JNK/AP1 pathways. Oncogene 22:3371-3385, 2003 [DOI] [PubMed] [Google Scholar]
- 20.Cai XLS, Zhang Z, Gonzalez CM, et al. : Kaposi's sarcoma-associated herpesvirus expresses an array of viral microRNAs in latently infected cells. Proc Natl Acad Sci USA 102:5570-5575, 2005 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bais C, Santomasso B, Coso O, et al. : G-protein-coupled receptor of Kaposi's sarcoma-associated herpesvirus is a viral oncogene and angiogenesis activator. Nature 391:86-89, 1998 [DOI] [PubMed] [Google Scholar]
- 22.Montaner S, Sodhi A, Ramsdell AK, et al. : The Kaposi's sarcoma-associated herpesvirus G protein-coupled receptor as a therapeutic target for the treatment of Kaposi's sarcoma. Cancer Res 66:168-174, 2006 [DOI] [PubMed] [Google Scholar]
- 23.Arvanitakis L, Geras-Raaka E, Varma A, et al. : Human herpesvirus KSHV encodes a constitutively active G-protein-coupled receptor linked to cell proliferation. Nature 385:347-350, 1997 [DOI] [PubMed] [Google Scholar]
- 24.Ishido S, Wang C, Lee BS, et al. : Downregulation of major histocompatibility complex class I molecules by Kaposi's sarcoma-associated herpesvirus K3 and K5 proteins. J Virol 74:5300-5309, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Haque M, Ueda K, Nakano K, et al. : Major histocompatibility complex class I molecules are down-regulated at the cell surface by the K5 protein encoded by Kaposi's sarcoma-associated herpesvirus/human herpesvirus-8. J Gen Virol 82:1175-1180, 2001 [DOI] [PubMed] [Google Scholar]
- 26.Coscoy L, Ganem D: Kaposi's sarcoma-associated herpesvirus encodes two proteins that block cell surface display of MHC class I chains by enhancing their endocytosis. Proc Natl Acad Sci USA 97:8051-8056, 2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Molden J, Chang Y, You Y, et al. : A Kaposi's sarcoma-associated herpesvirus-encoded cytokine homolog (vIL-6) activates signaling through the shared gp130 receptor subunit. J Biol Chem 272:19625-19631, 1997 [DOI] [PubMed] [Google Scholar]
- 28.Aoki Y, Narazaki M, Kishimoto T, et al. : Receptor engagement by viral interleukin-6 encoded by Kaposi sarcoma-associated herpesvirus. Blood 98:3042-3049, 2001 [DOI] [PubMed] [Google Scholar]
- 29.Aoki Y, Yarchoan R, Wyvill K, et al. : Detection of viral interleukin-6 in Kaposi sarcoma-associated herpesvirus-linked disorders. Blood 97:2173-2176, 2001 [DOI] [PubMed] [Google Scholar]
- 30.Aoki Y, Tosato G, Fonville TW. et al. : Serum viral interleukin-6 in AIDS-related multicentric Castleman disease. Blood 97:2526-2527, 2001 [DOI] [PubMed] [Google Scholar]
- 31.Polizzotto M, Uldrick T, Hu D, et al. : Clinical manifestations of Kaposi sarcoma herpesvirus lytic activation: Multicentric Castleman disease (KSHV–MCD) and the KSHV inflammatory cytokine syndrome. Front Microbiol 3:73, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rabkin CS, Bedi G, Musaba E, et al. : AIDS-related Kaposi's sarcoma is a clonal neoplasm. Clin Cancer Res 1:257, 1995 [PubMed] [Google Scholar]
- 33.Gill P, Tsai Y, Rao AP, et al. : Clonality in Kaposi's sarcoma. N Engl J Med 337:570-572, 1997 [DOI] [PubMed] [Google Scholar]
- 34.Duprez R, Lacoste V, Brière J, et al. : Evidence for a multiclonal origin of multicentric advanced lesions of Kaposi sarcoma. J Natl Cancer Inst 99:1086-1094, 2007 [DOI] [PubMed] [Google Scholar]
- 35.Maurer T, Ponte M, Leslie K: HIV-associated Kaposi's sarcoma with a high CD4 count and a low viral load. N Engl J Med 357:1352-1353, 2007 [DOI] [PubMed] [Google Scholar]
- 36.Bower M: How I treat HIV-associated multicentric Castleman disease. Blood 116:4415-4421, 2010 [DOI] [PubMed] [Google Scholar]
- 37.Jary A, Leducq V, Palich R, et al. : Usefulness of Kaposi's sarcoma-associated herpesvirus (KSHV) DNA viral load in whole blood for diagnosis and monitoring of KSHV-associated diseases. J Clin Microbiol 56:1-4, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Haq IU, Dalla Pria A, Papanastasopoulos P, et al. : The clinical application of plasma Kaposi sarcoma herpesvirus viral load as a tumour biomarker: Results from 704 patients. HIV Med 17:56-61, 2016 [DOI] [PubMed] [Google Scholar]
- 39.Polizzotto MN, Millo C, Uldrick TS, et al. : 18F-fluorodeoxyglucose positron emission tomography in Kaposi sarcoma herpesvirus-associated multicentric castleman disease: Correlation with activity, severity, inflammatory and virologic parameters. J Infect Dis 212:1250-1260, 2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.BioRender : Biorender.com
- 41.Letang E, Lewis JL, Bower M, et al. : Immune reconstitution inflammatory syndrome associated with Kaposi sarcoma: Higher incidence and mortality in Africa than in the UK. AIDS 27:1603-1613, 2013 [DOI] [PubMed] [Google Scholar]
- 42.Bower M, Nelson M, Young AM, et al. : Immune reconstitution inflammatory syndrome associated with Kaposi's sarcoma. J Clin Oncol 23:5224-5228, 2005 [DOI] [PubMed] [Google Scholar]
- 43.Manion M, Uldrick T, Polizzotto MN, et al. : Emergence of Kaposi's sarcoma herpesvirus-associated complications following corticosteroid use in TB-IRIS. Open Forum Infect Dis 5:ofy217, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Krown SE, Testa MA, Huang J: AIDS-related Kaposi's sarcoma: Prospective validation of the AIDS clinical trials group staging classification. AIDS Clinical Trials Group Oncology Committee. J Clin Oncol 15:3085-3092, 1997 [DOI] [PubMed] [Google Scholar]
- 45.Bower M, Dalla Pria A, Coyle C, et al. : Prospective stage-stratified approach to AIDS-related Kaposi's sarcoma. J Clin Oncol 32:409-414, 2014 [DOI] [PubMed] [Google Scholar]
- 46.Northfelt DW, Dezube BJ, Thommes JA, et al. : Pegylated-liposomal doxorubicin versus doxorubicin, bleomycin, and vincristine in the treatment of AIDS-related Kaposi's sarcoma: Results of a randomized phase III clinical trial. J Clin Oncol 16:2445-2451, 1998 [DOI] [PubMed] [Google Scholar]
- 47.Stewart S, Jablonowski H, Goebel FD, et al. : Randomized comparative trial of pegylated liposomal doxorubicin versus bleomycin and vincristine in the treatment of AIDS-related Kaposi's sarcoma. International Pegylated Liposomal Doxorubicin Study Group. J Clin Oncol 16:683-691, 1998 [DOI] [PubMed] [Google Scholar]
- 48.Martin-Carbonero L, Barrios A, Saballs P, et al. : Pegylated liposomal doxorubicin plus highly active antiretroviral therapy versus highly active antiretroviral therapy alone in HIV patients with Kaposi's sarcoma. AIDS 18:1737-1740, 2004 [DOI] [PubMed] [Google Scholar]
- 49.Cooley T, Henry D, Tonda M, et al. : A randomized, double-blind study of pegylated liposomal doxorubicin for the treatment of AIDS-related Kaposi's sarcoma. Oncologist 12:114-123, 2007 [DOI] [PubMed] [Google Scholar]
- 50.Cianfrocca M, Lee S, Von Roenn J, et al. : Randomized trial of paclitaxel versus pegylated liposomal doxorubicin for advanced human immunodeficiency virus-associated Kaposi sarcoma: Evidence of symptom palliation from chemotherapy. Cancer 116:3969-3977, 2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Saville MW, Lietzau J, Pluda JM, et al. : Treatment of HIV-associated Kaposi's sarcoma with paclitaxel. Lancet 346:26-28, 1995 [DOI] [PubMed] [Google Scholar]
- 52.Welles L, Saville MW, Lietzau J, et al. : Phase II trial with dose titration of paclitaxel for the therapy of human immunodeficiency virus-associated Kaposi's sarcoma. J Clin Oncol 16:1112-1121, 1998 [DOI] [PubMed] [Google Scholar]
- 53.Gill PS, Tulpule A, Espina BM, et al. : Paclitaxel is safe and effective in the treatment of advanced AIDS-related Kaposi's sarcoma. J Clin Oncol 17:1876-1883, 1999 [DOI] [PubMed] [Google Scholar]
- 54.Tulpule A, Groopman J, Saville MW, et al. : Multicenter trial of low-dose paclitaxel in patients with advanced AIDS-related Kaposi sarcoma. Cancer 95:147-154, 2002 [DOI] [PubMed] [Google Scholar]
- 55.Krown SE, Moser CB, MacPhail P, et al. : Treatment of advanced AIDS-associated Kaposi sarcoma in resource-limited settings: A three-arm, open-label, randomised, non-inferiority trial. Lancet 395:1195-1207, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Nasti G, Errante D, Talamini R, et al. : Vinorelbine is an effective and safe drug for AIDS-related Kaposi's sarcoma: Results of a phase II study. J Clin Oncol 18:1550-1557, 2000 [DOI] [PubMed] [Google Scholar]
- 57.Busakhala NW, Waako PJ, Strother MR, et al. : Randomized phase IIA trial of gemcitabine compared with bleomycin plus vincristine for treatment of Kaposi's sarcoma in patients on combination antiretroviral therapy in western Kenya. J Glob Oncol 4:1-9, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Evans SR, Krown SE, Testa MA, et al. : Phase II evaluation of low-dose oral etoposide for the treatment of relapsed or progressive AIDS-related Kaposi's sarcoma: An AIDS Clinical Trials Group clinical study. J Clin Oncol 20:3236-3241, 2002 [DOI] [PubMed] [Google Scholar]
- 59.Polizzotto MN, Uldrick TS, Wyvill KM, et al. : Pomalidomide for symptomatic Kaposi's sarcoma in people with and without HIV infection: A phase I/II study. J Clin Oncol 34:4125-4131, 2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Pourcher V, Desnoyer A, Assoumou L, et al. : Phase II trial of lenalidomide in HIV-infected patients with previously treated Kaposi's sarcoma: Results of the ANRS 154 Lenakap Trial. AIDS Res Hum Retroviruses 33:1-10, 2017 [DOI] [PubMed] [Google Scholar]
- 61.Fife K, Howard MR, Gracie F, et al. : Activity of thalidomide in AIDS-related Kaposi's sarcoma and correlation with HHV8 titre. Int J STD AIDS 9:751-755, 1998 [DOI] [PubMed] [Google Scholar]
- 62.Little RF, Wyvill KM, Pluda JM, et al. : Activity of thalidomide in AIDS-related Kaposi's sarcoma. J Clin Oncol 18:2593-2602, 2000 [DOI] [PubMed] [Google Scholar]
- 63.Uldrick TS, Wyvill KM, Kumar P, et al. : Phase II study of bevacizumab in patients with HIV-associated Kaposi's sarcoma receiving antiretroviral therapy. J Clin Oncol 30:1476-1483, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Reid EG, Suazo A, Lensing SY, et al. : Pilot trial AMC-063: Safety and efficacy of bortezomib in AIDS-associated Kaposi sarcoma. Clin Cancer Res 26:558-565, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Uldrick TS, Gonçalves PH, Wyvill KM, et al. : A phase Ib study of sorafenib (BAY 43-9006) in patients with Kaposi sarcoma. Oncologist 22:505-e49, 2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Koon HB, Bubley GJ, Pantanowitz L, et al. : Imatinib-induced regression of AIDS-related Kaposi's sarcoma. J Clin Oncol 23:982-989, 2005 [DOI] [PubMed] [Google Scholar]
- 67.Koon HB, Krown SE, Lee JY, et al. : Phase II trial of imatinib in AIDS-associated Kaposi's sarcoma: AIDS Malignancy Consortium Protocol 042. J Clin Oncol 32:402-408, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Uldrick TS, Goncalves PH, Abdul-Hay M, et al. : Assessment of the safety of pembrolizumab in patients with HIV and advanced cancer-A phase 1 study. JAMA Oncol 5:1332-1339, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.National Comprehensive Cancer Network : AIDS-Related Kaposi Sarcoma (Version 1.2018), 2018 [DOI] [PubMed] [Google Scholar]
- 70.Northfelt DW, Martin FJ, Working P, et al. : Doxorubicin encapsulated in liposomes containing surface-bound polyethylene glycol: Pharmacokinetics, tumor localization, and safety in patients with AIDS-related Kaposi's sarcoma. J Clin Pharmacol 36:55-63, 1996 [DOI] [PubMed] [Google Scholar]
- 71.Berry G, Billingham M, Alderman E, et al. : The use of cardiac biopsy to demonstrate reduced cardiotoxicity in AIDS Kaposi’s sarcoma patients treated with pegylated liposomal doxorubicin. Ann Oncol 9:711-716, 1998 [DOI] [PubMed] [Google Scholar]
- 72.Zhu YX, Kortuem KM, Stewart AK: Molecular mechanism of action of immune-modulatory drugs thalidomide, lenalidomide and pomalidomide in multiple myeloma. Leuk Lymphoma 54:683-687, 2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lopez-Girona A, Mendy D, Ito T, et al. : Cereblon is a direct protein target for immunomodulatory and antiproliferative activities of lenalidomide and pomalidomide. Leukemia 26:2326-2335, 2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Ramaswami R, Uldrick TS, Polizzotto MN, et al. : A pilot study of liposomal doxorubicin combined with bevacizumab followed by bevacizumab monotherapy in patients with advanced Kaposi sarcoma. Clin Cancer Res 25:4238-4247, 2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Koon HB, Fingleton B, Lee JY, et al. : Phase II AIDS Malignancy Consortium trial of topical halofuginone in AIDS-related Kaposi sarcoma. J Acquir Immune Defic Syndr 56:64-68, 2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Lurain K, Ramaswami R, Yarchoan R, et al. : Anti-PD-1 and anti-PD-L1 monoclonal antibodies in people living with HIV and cancer. Curr HIV/AIDS Rep 17:547-556, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Reuss JE, Stern D, Foster JC, et al. : Assessment of Cancer Therapy Evaluation Program advocacy and inclusion rates of people living with HIV in anti-PD1/PDL1 clinical trials. JAMA Netw Open 3:e2027110, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Galanina N, Goodman AM, Cohen PR, et al. : Successful treatment of HIV-associated Kaposi sarcoma with immune checkpoint blockade. Cancer Immunol Res 6:1129-1135, 2018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Uldrick TS, Polizzotto MN, Aleman K, et al. : Rituximab plus liposomal doxorubicin in HIV-infected patients with KSHV-associated multicentric Castleman disease. Blood 124:3544-3552, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.National Comprehensive Cancer Network : B-Cell Lymphomas. Version 4.2021, 2021 [Google Scholar]
- 81.Ramaswami R, Lurain K, Peer CJ, et al. : Tocilizumab in patients with symptomatic Kaposi sarcoma herpesvirus-associated multicentric Castleman disease. Blood 135:2316-2319, 2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Little RF, Aleman K, Kumar P, et al. : Phase 2 study of pegylated liposomal doxorubicin in combination with interleukin-12 for AIDS-related Kaposi sarcoma. Blood 110:4165-4171, 2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Little RF, Pluda JM, Wyvill KM, et al. : Activity of subcutaneous interleukin-12 in AIDS-related Kaposi sarcoma. Blood 107:4650-4657, 2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Boscoe FP, Johnson CJ, Sherman RL, et al. : The relationship between area poverty rate and site-specific cancer incidence in the United States. Cancer 120:2191-2198, 2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Centers for Disease Control:. Lifetime Risk of HIV Diagnosis, 2016 [Google Scholar]