Abstract
Persons living with human immunodeficiency virus (PLWH) carry increased risk for developing malignancies, including glioblastoma. Despite extensive investigations, both human immunodeficiency virus (HIV) and glioblastoma are incurable. Treatment for a patient with combined glioblastoma and HIV remains an unexplored need. Preliminary evidence suggests that immunotherapy may be effective for the simultaneous treatment of both HIV and cancer by reversing HIV latency and T cell exhaustion. We present a case of glioblastoma in a PLWH who was treated with pembrolizumab. Treatment was well tolerated and safe with a mixed response. Our patient did not develop any opportunistic infections, immune-related adverse events, or worsening of his immunodeficiency. To our knowledge, this is the first reported case of a PLWH and glioblastoma treated with immunotherapy.
Keywords: : cancer disparities, glioblastoma, HIV, HIV latency, HIV reservoir, human immunodeficiency virus, immunotherapy, pembrolizumab, PD-1, programmed cell death 1 inhibitor
Plain Language Summary
Persons living with human immunodeficiency virus (PLWH) are at increased risk for cancers, including glioblastoma. Despite extensive research, both human immunodeficiency virus (HIV) and glioblastoma are incurable. The optimal treatment for concurrent HIV and glioblastoma is unknown. Early evidence suggests that immunotherapy can deplete residual HIV and restore immune function. We present a case of glioblastoma in a PLWH treated with immunotherapy. Treatment was well tolerated and safe. To our knowledge, this is the first reported case of a PLWH and glioblastoma treated with immunotherapy.
Plain language summary
Article highlights.
Human immunodeficiency virus cannot be fully eradicated
Full eradication of the virus is hampered by reservoirs of latent HIV residing within memory CD4+ T cells.
Persons living with human immunodeficiency virus carry an increased risk for cancer and treatment for these patients is an unmet need
Immunotherapy appears safe and effective in persons living with HIV with cancer.
Immunotherapy for glioblastoma is under investigation.
Preliminary evidence suggests that immunotherapy may also reverse HIV latency.
1. Background
Despite the advent of highly active antiretroviral therapy (HAART), human immunodeficiency virus (HIV) remains an incurable disease [1]. Though HIV-1 RNA viral load levels (VL) can be undetectable in persons living with HIV (PLWH), PLWH can only achieve functional cure [2,3]. Full eradication of the virus cannot be achieved due to reservoirs of latent HIV residing within memory CD4+ T cells [2,3]. This intracellular location provides a selective advantage for the virus to persist because it is inaccessible by HAART [3,4,5,6,7]. The mechanism by which this occurs is driven by a positive tropism between latent HIV and programmed cell death protein (PD)-1 [6,8]. In PLWH on HAART, PD-1 is overexpressed on the surface of memory CD4+ T cells and draws latent HIV into the cells [6,8]. Accordingly, PD-1 expression correlates with the extent of these reservoirs [9,10]. Permanent suppression of this pool of transcriptionally silent HIV is one treatment approach under exploration [3]. However, there remains a potential for latent HIV to reactivate, as evidenced by the remarkable case of the ‘Mississippi Child’ wherein an HIV-infected child in remission subsequently relapsed from reactivation of dormant HIV [3,5,11]. Depletion of HIV from these reservoirs may represent a better approach to eliminate the risk for reactivation [3,4,12]. Histone deacetylase and PD-1 inhibitors are tumor-directed therapies repurposed as HIV latent reversal agents to force HIV out of intracellular hiding where it can be eliminated by HAART [4,12,13,14].
Beyond the challenges of disease control with HIV, PLWH also carry an increased risk of developing malignancies compared with the general population [15,16,17]. This increased risk is driven by impaired immune surveillance [16,18]. Understanding the underlying mechanism of this immunodeficiency is essential to understanding how cancer treatment in PLWH can be addressed. First, HIV preferentially destroys CD4+ helper T cells, whose function is to activate the CD8+ T effector cells that clear virally infected cells and tumor cells [19]. Second, the function of CD8+ T cells is further impaired by the chronic immune activation from HIV [16,18]. Chronic immune activation upregulates PD-1 expression in previously primed CD8+ T cells, which leads to T cell exhaustion [16,18]. The loss of CD8+ T cell function from both these mechanisms then permit neoplastic development in PLWH [16,18]. Last, irrespective of HIV, cancers are marked by immunosuppression and T cell exhaustion [20]. On the basis of this understanding, the optimal therapy would inhibit the interaction between PD-1 and its ligands [6,8,9,17]. This blockade would concurrently address HIV latency and T cell exhaustion [17,21,22,23]. For these reasons, PD-1 inhibitors, such as pembrolizumab, have been the focus of studies for PLWH and cancer.
Early limited results are promising, but none of these studies included glioblastoma, a non-AIDS defining malignancy [13,16,17,21,25]. Malignancies, including glioblastoma, have been traditionally excluded from HIV clinical trials [13,17,27]. Correspondingly, PLWH have historically been excluded from glioblastoma trials [13,14,28,29,30,31,32,33,34]. Cancer care disparities in PLWH are now being recognized with efforts underway to bridge this knowledge gap [32,33,34,35]. We present a case of a well-controlled PLWH with progressive glioblastoma who was treated with pembrolizumab. We hypothesized that PD-1 inhibition could potentially reverse both HIV latency and T cell exhaustion to treat HIV and glioblastoma concurrently.
2. Case presentation
A 39-year-old male PLWH presented with headache. His neurologic exam was unremarkable. His outside lab lymphocyte subset panel 5 revealed that the plasma VL was 82,900 copies/ml and the absolute CD4+ count was 299 cells/ml3 (Figure 1). HAART was initiated. Brain MRI with and without contrast showed left parietal ring enhancing intra-axial mass (Figure 2A), and he underwent subtotal resection of the tumor.
Figure 1.
Timeline of patient's viral load level and CD4+ counts pre and post-pembrolizumab therapy. The viral load was undetectable prior to his first and second pembrolizumab infusions. Following these infusions, low-level viremia was detected, suggestive of HIV latency reversal and the purging of quiescent HIV out of the CD4+ T cells into the peripheral blood.
Figure 2.
Contrast-enhanced T1-weighted brain MRI axial sections, show masses in the left parietal, left temporal and left frontal lobes pre-pembrolizumab (A–C) and post three infusions of pembrolizumab (D–F). (A) 4.2 cm left parietal enhancing mass. (B) 3.2 cm left temporal enhancing mass. (C) 0.8 cm left frontal enhancing mass. (D) Interval regression (3.2 cm) of the left parietal mass. (E) Interval growth (6.0 cm) of the left temporal mass. (F) Interval enlargement (1.7 cm) of the left frontal heterogeneously enhancing mass.
Histologic sections showed a diffusely infiltrating glial neoplasm with extensive necrosis, composed of highly pleomorphic cells with bizarre, irregular nuclei, smudgy chromatin and variable amounts of eosinophilic cytoplasm intermixed with poorly differentiated cells with polygonal hyperchromatic nuclei and scant cytoplasm in the background of abundant geographic necrosis and numerous regions with microvascular proliferation (Figures 3A–C). On immunohistochemistry (IHC), tumor cells were patchy positive with GFAP, showed increased expression of p53 (suggestive of TP53 mutation) and had a variable Ki-67 proliferative index ranging from 20 to 80% in some areas (Figures 3D–F). IHC for CD3 and CD20 highlighted scattered T- and B-cells, respectively, but were negative in tumor cells (Figures 3G-H). In situ hybridization for EBER (Epstein-Barr virus [EBV]-encoded small RNA) was also negative. Next-generation sequencing (NGS) performed at a commercial laboratory showed a hot spot mutation in the promoter region of the TERT gene and a damaging missense mutation in the TP53 tumor suppressor gene. There were no mutations in ATRX, IDH1, IDH2, H3-3A and H3C3 genes (by NGS), and EGFRvIII was absent (by real-time PCR). Fluorescence in situ hybridization showed deletion of PTEN gene with no evidence of MET, MYCN, PDGFRA amplifications, BRAF rearrangement or 1p/19q codeletion. RNA sequencing was negative for RAF1, FGFR1/2/3 and NTRK1/2/3 fusions. PD-L1 LDT expression was detected by IHC in 60% Tumor Cells/Total Viable Tumor Cells. While significant atypia in a relatively young patient raised the possibility of a hypermutated tumor, tumor mutational burden (TMB) was low (5 mutations/Mb), and there was no microsatellite instability. MGMT promoter was unmethylated. A final integrated diagnosis of Glioblastoma, IDH-wildtype was determined based on both morphologic features and molecular findings [36,37].
Figure 3.
Histologic sections show a high-grade glioma (A) in the background of extensive necrosis (arrow). (B) Some tumor cells show abundant eosinophilic cytoplasm and bizarre nuclei. Microvascular proliferation (arrows) is present. (C) In other areas, immature-appearing tumor cells with scant cytoplasm and polygonal, hyperchromatic nuclei are arranged around vessels. (D) GFAP immunohistochemical stain is patchy and positive supporting the diagnosis of a glioma. (E) P53 stain is positive in more than 90% of the tumor cells, suggestive of TP53 gene mutation. (F) Ki-67 proliferative index is increased, reaching 80% in some areas. (G) CD3 stain highlights scattered small T-lymphocytes and (H) CD20 stain highlights scattered small B-lymphocytes. All scale bars are 100 microns.
Temozolomide was withheld for neutropenia and his tumor was treated with radiation (60 Gy in 30 fractions) alone. On Day 208, surveillance brain MRI with and without contrast showed 0.8 cm dural-based left frontal and 3.2 cm left temporal enhancing masses (Figures 2B–C). His neutropenia resolved, but he was not amenable to chemotherapy. Additional treatment options were offered, including pembrolizumab and lenvatinib [38]. He expressed a preference for intravenous route of administration, and he decided to proceed with pembrolizumab (200 mg every 3 weeks) and bevacizumab (10 mg/kg every 2 weeks). On Day 230, he received his first pembrolizumab infusion. His pre-pembrolizumab plasma VL was undetectable and his CD4+ count was 408 cells/ml3. On Day 240, his VL increased to 52 copies/ml, which subsequently dropped to undetectable levels on Day 246. At the time of his second pembrolizumab infusion on Day 252, his VL remained undetectable and his CD4+ count rose to 808 cells/ml3. At the time of his last bevacizumab infusion as well as his third and last pembrolizumab infusion on Day 276, he developed recurrent low-level viremia (VL: 94 copies/ ml) and his CD4+count dropped to 173 cells/ml3. He reported compliance with HAART. No opportunistic infections developed from the CD4+count decrease below 200 cells/ml3. On Day 290, his brain MRI demonstrated continued regression of the left parietal tumor, but interval enlargement of both left frontal and left temporal masses (Figures 2D–F). On Day 315, his VL was again undetectable and his CD4+ count increased to 661 cells/ml3. He was initiated on dexamethasone 4 mg twice daily with improvement in his headaches and further pembrolizumab was held. He experienced no immune-related adverse events from the pembrolizumab and no adverse events from bevacizumab. The decision was made to offer neurosurgical intervention for the 6 cm left temporal tumor for symptomatic relief and tissue diagnosis. The patient decided to proceed with resection and further bevacizumab was held in anticipation for surgery. Unfortunately, on Day 323, 11 months from his initial diagnosis, he passed away from causes unrelated to his HIV and glioblastoma. Ethical guidelines set out by the declaration of Helsinki were followed in the preparation of this report, and the patient provided written consent.
3. Discussion
Immunotherapy, such as PD-1 inhibitors, has revolutionized the landscape of treatment for many cancers. It is widely accepted that the inhibitory action of PD-1 augments the immune response and restores the function of CD8+ T cells to induce apoptosis in both virally infected cells and tumor cells [6,8,9,17,20]. However, PD-1 inhibitors have shown limited success for the treatment of glioblastoma due to its immunosuppressive tumor microenvironment (TME), with tumor infiltrating lymphocytes representing only <1%–2% of the TME [39]. Though high TMB may be useful as a predictive marker for response to immunotherapy in specific solid cancers, evidence suggests that high TMB is not associated with immunotherapy response for glioblastoma [40]. Conversely, TMB may be inversely associated with immunotherapy efficacy [41]. Our patient's TMB was low, which has been associated with prolonged survival in recurrent glioblastoma patients treated with immunotherapy [41]. Limited evidence exists for the use of PD-1 inhibitors in HIV and comorbid cancer. Even less evidence exists for its use in HIV and glioblastoma. When comorbid HIV and glioblastoma occur in the same patient, there are added complexities that must be taken into consideration. Immunodeficiency from the virus coupled with immunodeficiency from the tumor may impair the ability to mount a response to therapy [13,14,16,28,29,30,31,32,33,34,42,43]. Furthermore, drug–drug interactions between HAART and chemotherapy can exacerbate depressed CD4+ counts in a PLWH [44,45]. To mitigate these barriers, PD-1 inhibitors are being explored for PLWH and cancer. PD-1 inhibitors offer immune reconstitution without the conventional myelosuppression that is associated with chemotherapy and can potentially dually treat HIV and glioblastoma with no added risk of toxicity, opportunistic infections, or worsening immunosuppression [16,42,43,44,45,46].
Evidence suggests that PD-1 inhibitors do not cause detrimental effect on the VL in PLWH and malignancy [17,47]. Low level viremia bearing no clinical consequence has been reported, but a recent study suggests that low level viremia may be due to latency reversal [12,13,17]. In our patient with undetectable VL prior to pembrolizumab therapy, we observed two occurrences of low level viremia during pembrolizumab therapy. On both occasions, his VL subsequently returned to undetectable levels. This raises the possibility that low level viremia is a surrogate marker for HIV latency reversal and can be explained by quiescent HIV being purged out of the CD4+ T cells into peripheral blood giving rise to viremia then the subsequent elimination by HAART returning the VL back to down to undetectable levels [5,6,13].
PD-1 inhibitors have also not been shown to detrimentally affect CD4+ counts in PLWH and malignancy and do not increase the risk for opportunistic infections [17,21,47,48]. Our case corroborates these findings. CD4+ counts can steadily increase or decrease, but can also fluctuate [47,49,50,51], as was observed in our patient. Baseline to best response CD4+ count showed a single fold increase from 408 cells/ml3 to 808 cells/ml3. Though his CD4+ count fell below 200 cells/ml3 on one occasion, no opportunistic infections were unmasked.
PD-1 inhibitors are considered to be overall safe and well tolerated in PLWH and malignancy [13,17,21,25]. There are no apparent added risks for the development of immune-related adverse events (irAEs) in this patient population [17]. Reported risk factors include a higher number of cycles (median = 4 cycles) and higher cumulative dose (median = 600 mg) and [52]. In our patient, there were no irAEs following 3 administered cycles of pembrolizumab with a cumulative dose of 600 mg.
In addition to safety, preliminary evidence shows that pembrolizumab may be effective in PLWH with durable response [17,21,22,47,49,53,54,55,56,57]. Moreover, the combination of pembrolizumab and bevacizumab may be synergistic [58]. Anti-angiogenic therapy upregulates the expression of adhesion molecules, thereby enhancing the interaction of CD8+ effector T cells allowing CD8+ T cells to cross the blood-brain barrier [58]. However, this combinatorial therapy produced a prolonged response in a case of metastatic glioblastoma, but it remains to be determined if this will translate to improved overall survival [58,59].
In our patient, tumor regression (1/fourfold decrease) was noted in the intraparenchymal left parietal tumor, the original site of disease. Though it is possible this was a result of bevacizumab-induced pseudoresponse [60], there was no rebound growth 1.5 months after bevacizumab was discontinued. However, conflicting responses to pembrolizumab have been reported [17]. Our case also substantiates these findings. In our patient, we observed a mixed tumor response with tumor enlargement in the new dural-based left frontal and left temporal tumors with 1.1-fold and 3.6-fold increases compared with baseline, respectively. The paradoxical regression of the intraparenchymal tumor and the enlargement of the dural-based tumors was unexpected. It is widely accepted that glioblastomas are immunologically cold tumors and trafficking of CD8+ T cells beyond the blood–brain barrier is a known obstacle of PD-1 inhibitors [48,61,62]. The dural-based location is expected to provide an advantage by ameliorating the limitation of the blood-brain barrier [63,64]. We reason that these dural-based lesions were not previously treated as was the previously radiated left parietal lesion, which may have effected synergistic therapeutic benefit [65]. Further, it remains unknown if the enlargement of the left frontal and left temporal tumors were a result of true progression or pseudoprogression. Immunotherapy-induced inflammation can mimic true progression and can occur in MGMT unmethylated glioblastoma, though the probability is lower compared with methylated glioblastoma [21,47,66,67]. In the absence of tissue diagnosis due to our patient's untimely death from unrelated causes, a final diagnosis could not be made.
Our case adds to the mounting evidence supporting the use of immunotherapy in PLWH and cancer. In our patient, pembrolizumab was well tolerated, safe and at least partially effective with no apparent increased risk for toxicity, opportunistic infections, or worsening immunosuppression. To our knowledge, this is the first reported case of the use of pembrolizumab in a PLWH and glioblastoma.
There are limitations to this study. A single case is not generalizable to the entire patient population. Given our patient's untimely death, the duration for follow-up was short and the true efficacy of pembrolizumab alone could not be further assessed. The retrospective aspect of this study allowed for trending HIV-1 RNA, but not CD4+ T cell-associated HIV RNA [13,68]. There was a possibility that the observed low-level viremia was a result of HAART non-compliance as opposed to reversal of HIV latency. However, this scenario is less likely given the subsequent return of VL to undetectable levels and subsequent rise in CD4+ count.
4. Conclusion
The long-term survival of persons living with HIV is altering the clinical spectrum for the development of non-AIDS defining malignancies, including glioblastoma. Immunotherapy may potentially treat HIV and glioblastoma concurrently. Pembrolizumab was well tolerated, safe and effective in our patient. Further studies are needed to support the safety and efficacy of immunotherapy in persons living with HIV and cancer.
Author contributions
C Yuen – study concept, data collection, analysis, interpretation, manuscript drafting, revision and final approval. S Bao - data collection, analysis and final approval. M Pekmezci – data collection, analysis, interpretation, manuscript drafting, revision and final approval. F Mo – data collection and final approval. X-T Kong – manuscript revision and final approval.
Financial disclosure
The authors have no financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
Competing interests disclosure
X-T Kong: Received honorarium from Zai Lab for invited speeches for symposiums prior to July of 2021. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Writing disclosure
No writing assistance was utilized in the production of this manuscript.
Ethical conduct of research
The Community Regional Medical Center Institutional Review Board approved this study. Consent for publication: Written consent was obtained from the patient and written consent for the final publication was obtained from the patient's next of kin.
Data availability statement
Availability of data and material: The datasets from this study are available from the corresponding author upon reasonable request.
Funding: No funding was received for this manuscript.
References
Papers of special note have been highlighted as: • of interest; •• of considerable interest
- 1.Cillo AR, Mellors JW. Which therapeutic strategy will achieve a cure for HIV-1? Curr Opin Virol. 2016;18:14–19. doi: 10.1016/j.coviro.2016.02.001 [DOI] [PubMed] [Google Scholar]
- 2.Marcus JL, Leyden WA, Alexeeff SE, et al. Comparison of overall and comorbidity-free life expectancy between insured adults with and without HIV infection, 2000–2016. JAMA Netw Open. 2020;3(6):e207954. doi: 10.1001/jamanetworkopen.2020.7954 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Dahabieh MS, Battivelli E, Verdin E. Understanding HIV latency: the road to an HIV cure. Annu Rev Med. 2015;66:407–421. doi: 10.1146/annurev-med-092112-152941 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Describes the pathogenesis of HIV latency.
- 4.Van Lint C, Emiliani S, Ott M, Verdin E. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. EMBO J. 1996;15(5):1112–1120. doi: 10.1002/j.1460-2075.1996.tb00449.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lopes JR, Chiba DE, Dos Santos JL. HIV latency reversal agents: a potential path for functional cure? Eur J Med Chem. 2021;213:113213. doi: 10.1016/j.ejmech.2021.113213 [DOI] [PubMed] [Google Scholar]; •• Describes the role of therapeutics to potentially reverse HIV latency.
- 6.Fromentin R, DaFonseca S, Costiniuk CT, et al. PD-1 blockade potentiates HIV latency reversal ex vivo in CD4(+) T cells from ART-suppressed individuals. Nat Commun. 2019;10(1):814. doi: 10.1038/s41467-019-08798-7 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Describes the potential role of PD-1 inhibitors to reverse HIV latency.
- 7.Scully EP, Rutishauser RL, Simoneau CR, et al. Inconsistent HIV reservoir dynamics and immune responses following anti-PD-1 therapy in cancer patients with HIV infection. Ann Oncol. 2018;29(10):2141–2142. doi: 10.1093/annonc/mdy259 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Chomont N, El-Far M, Ancuta P, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med. 2009;15(8):893–900. doi: 10.1038/nm.1972 [DOI] [PMC free article] [PubMed] [Google Scholar]; • Describes HIV latency and intra-cellular persistence.
- 9.Day CL, Kaufmann DE, Kiepiela P, et al. PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature. 2006;443(7109):350–354. doi: 10.1038/nature05115 [DOI] [PubMed] [Google Scholar]; • Describes molecular mechanisms that may contribute to HIV latency.
- 10.Hatano H. Immune activation and HIV persistence: considerations for novel therapeutic interventions. Curr Opin HIV AIDS. 2013;8(3):211–216. doi: 10.1097/COH.0b013e32835f9788 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Describes possible therapies to reverse HIV latency.
- 11.Luzuriaga K, Gay H, Ziemniak C, et al. Viremic relapse after HIV-1 remission in a perinatally infected child. N Engl J Med. 2015;372(8):786–788. doi: 10.1056/NEJMc1413931 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Guihot A, Marcelin AG, Massiani MA, et al. Drastic decrease of the HIV reservoir in a patient treated with nivolumab for lung cancer. Ann Oncol. 2018;29(2):517–518. doi: 10.1093/annonc/mdx696 [DOI] [PubMed] [Google Scholar]; •• This publication describes reversal of HIV latency with immunotherapy in cancer patient.
- 13.Uldrick TS, Adams SV, Fromentin R, et al. Pembrolizumab induces HIV latency reversal in people living with HIV and cancer on antiretroviral therapy. Sci Transl Med. 2022;14(629):eabl3836. doi: 10.1126/scitranslmed.abl3836 [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Describes reversal of HIV latency with immunotherapy in cancer patients.
- 14.Johnson DB, Sullivan RJ, Menzies AM. Immune checkpoint inhibitors in challenging populations. Cancer. 2017;123(11):1904–1911. doi: 10.1002/cncr.30642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hajjar M, Lacoste D, Brossard G, et al. Non-acquired immune deficiency syndrome-defining malignancies in a hospital-based cohort of human immunodeficiency virus-infected patients: Bordeaux, France, 1985–1991. Groupe d'Epidemiologie Clinique du SIDA en Aquitaine. J Natl Cancer Inst. 1992;84(20):1593–1595. doi: 10.1093/jnci/84.20.1593 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Choy W, Lagman C, Lee SJ, et al. Impact of human immunodeficiency virus in the pathogenesis and outcome of patients with glioblastoma multiforme. Brain Tumor Res Treat. 2016;4(2):77–86. doi: 10.14791/btrt.2016.4.2.77 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Uldrick TS, Goncalves PH, Abdul-Hay M, et al. Assessment of the safety of pembrolizumab in patients with HIV and advanced cancer-a Phase I study. JAMA Oncol. 2019;5(9):1332–1339. doi: 10.1001/jamaoncol.2019.2244 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Hall JR, Short SC. Management of glioblastoma multiforme in HIV patients: a case series and review of published studies. Clin Oncol (R Coll Radiol). 2009;21(8):591–597. doi: 10.1016/j.clon.2009.04.006 [DOI] [PubMed] [Google Scholar]
- 19.Iwai Y, Ishida M, Tanaka Y, et al. Involvement of PD-L1 on tumor cells in the escape from host immune system and tumor immunotherapy by PD-L1 blockade. Proc Natl Acad Sci USA. 2002;99(19):12293–12297. doi: 10.1073/pnas.192461099 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Woroniecka K, Fecci PE. T-cell exhaustion in glioblastoma. Oncotarget. 2018;9(82):35287–35288. doi: 10.18632/oncotarget.26228 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Chang E, Sabichi AL, Kramer JR, et al. Nivolumab treatment for cancers in the HIV-infected population. J Immunother. 2018;41(8):379–383. doi: 10.1097/CJI.0000000000000240 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lavole A, Guihot A, Veyri M, et al. PD-1 blockade in HIV-infected patients with lung cancer: a new challenge or already a strategy? Ann Oncol. 2018;29(4):1065–1066. doi: 10.1093/annonc/mdx817 [DOI] [PubMed] [Google Scholar]
- 23.Ostios-Garcia L, Faig J, Leonardi GC, et al. Safety and efficacy of PD-1 inhibitors among HIV-positive patients with non-small cell lung cancer. J Thorac Oncol. 2018;13(7):1037–1042. doi: 10.1016/j.jtho.2018.03.031 [DOI] [PubMed] [Google Scholar]
- 25.Rasmussen TA, Rajdev L, Rhodes A, et al. Impact of anti-PD-1 and anti-CTLA-4 on the human immunodeficiency virus (HIV) reservoir in people living with HIV with cancer on antiretroviral therapy: the AIDS Malignancy Consortium 095 study. Clin Infect Dis. 2021;73(7):e1973–e1981. doi: 10.1093/cid/ciaa1530 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Swindells S, Andrade-Villanueva JF, Richmond GJ, et al. Long-acting cabotegravir and rilpivirine for maintenance of HIV-1 suppression. N Engl J Med. 2020;382(12):1112–1123. doi: 10.1056/NEJMoa1904398 [DOI] [PubMed] [Google Scholar]
- 28.Rodrigues LF, Camacho A, Spohr T. Secondary glioblastoma metastasis outside the central nervous system in a young HIV-infected patient. Ther Adv Med Oncol. 2020;12:1758835920923432. doi: 10.1177/1758835920923432 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Mendez Valdez MJ, Lu VM, Kim E, et al. Glioblastoma multiforme in patients with human immunodeficiency virus: an integrated review and analysis. J Neurooncol. 2022;159(3):571–579. doi: 10.1007/s11060-022-04095-4 [DOI] [PubMed] [Google Scholar]
- 30.Wang T, Gao T, Niu X, et al. Clinical characteristics and prognostic analysis of glioma in human immunodeficiency virus-infected patients. World Neurosurg. 2018;114:e218–e223. doi: 10.1016/j.wneu.2018.02.168 [DOI] [PubMed] [Google Scholar]
- 31.Ludmir EB, Espinoza AF, Jethanandani A, et al. Incidence and correlates of HIV exclusion criteria in cancer clinical trials. Int J Cancer 2020;146(8):2362–2364. doi: 10.1002/ijc.32800 [DOI] [PubMed] [Google Scholar]
- 32.Suneja G, Coghill A. Cancer care disparities in people with HIV in the United States. Curr Opin HIV AIDS. 2017;12(1):63–68. doi: 10.1097/COH.0000000000000320 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Corrigan KL, Wall KC, Bartlett JA, Suneja G. Cancer disparities in people with HIV: a systematic review of screening for non-AIDS-defining malignancies. Cancer. 2019;125(6):843–853. doi: 10.1002/cncr.31838 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Uldrick TS, Ison G, Rudek MA, et al. Modernizing clinical trial eligibility criteria: recommendations of the American Society of Clinical Oncology-Friends of Cancer Research HIV working group. J Clin Oncol. 2017;35(33):3774–3780. doi: 10.1200/JCO.2017.73.7338 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Reid E, Suneja G, Ambinder RF, et al. Cancer in people living with HIV, Version 1.2018, NCCN Clinical Practice Guidelines in Oncology. J Natl Compr Canc Netw. 2018;16(8):986–1017. doi: 10.6004/jnccn.2018.0066 [DOI] [PubMed] [Google Scholar]
- 36.Stichel D, Ebrahimi A, Reuss D, et al. Distribution of EGFR amplification, combined chromosome 7 gain and chromosome 10 loss, and TERT promoter mutation in brain tumors and their potential for the reclassification of IDHwt astrocytoma to glioblastoma. Acta Neuropathol. 2018;136(5):793–803. doi: 10.1007/s00401-018-1905-0 [DOI] [PubMed] [Google Scholar]
- 37.World Health O, International Agency for Research on C, Board WHOCoTE . Central nervous system tumours. Lyon: International Agency for Research on Cancer, World Health Organization Lyon; 2021. English. [Google Scholar]
- 38.Gomez-Roca C, Yanez E, Im S-A, et al. LEAP-005: a Phase II multicohort study of lenvatinib plus pembrolizumab in patients with previously treated selected solid tumors—Results from the colorectal cancer cohort. J Clin Oncol. 2021;39(Suppl. 3):9 4. doi: 10.1200/JCO.2021.39.3_suppl.94 [DOI] [Google Scholar]
- 39.AlHarbi M, Ali Mobark N, AlMubarak L, et al. Durable response to nivolumab in a pediatric patient with refractory glioblastoma and constitutional biallelic mismatch repair deficiency. Oncologist. 2018;23(12):1401–1406. doi: 10.1634/theoncologist.2018-0163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Gershon R, Polevikov A, Karepov Y, et al. Frequencies of 4 tumor-infiltrating lymphocytes potently predict survival in glioblastoma, an immune desert. Neuro Oncol. 2024;26(3):473–487. doi: 10.1093/neuonc/noad204 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Gromeier M, Brown MC, Zhang G, et al. Very low mutation burden is a feature of inflamed recurrent glioblastomas responsive to cancer immunotherapy. Nat Commun. 2021;12(1):352. doi: 10.1038/s41467-020-20469-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Oliveira VC, Gomes T, Ferreira LC, et al. Glioblastoma multiforme in an HIV-infected patient: an unexpected diagnosis. J Int Assoc Provid AIDS Care. 2014;13(5):411–413. doi: 10.1177/2325957414536230 [DOI] [PubMed] [Google Scholar]
- 43.Blumenthal DT, Raizer JJ, Rosenblum MK, et al. Primary intracranial neoplasms in patients with HIV. Neurology. 1999;52(8):1648–1651. doi: 10.1212/WNL.52.8.1648 [DOI] [PubMed] [Google Scholar]
- 44.Torres HA, Mulanovich V. Management of HIV infection in patients with cancer receiving chemotherapy. Clin Infect Dis. 2014;59(1):106–114. doi: 10.1093/cid/ciu174 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Lurain K, Ramaswami R, Yarchoan R, Uldrick TS. Anti-PD-1 and Anti-PD-L1 monoclonal antibodies in people living with HIV and cancer. Curr HIV/AIDS Rep. 2020;17(5):547–556. doi: 10.1007/s11904-020-00525-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Gauchotte G, Peyre M, Pouget C, et al. Prognostic value of histopathological features and loss of H3K27me3 immunolabeling in anaplastic meningioma: a multicenter retrospective study. J Neuropathol Exp Neurol. 2020;79(7):754–762. doi: 10.1093/jnen/nlaa038 [DOI] [PubMed] [Google Scholar]
- 47.Sahin IH, Kane SR, Brutcher E, et al. Safety and efficacy of immune checkpoint inhibitors in patients with cancer living with HIV: a perspective on recent progress and future needs. JCO Oncol Pract. 2020;16(6):319–325. doi: 10.1200/JOP.19.00754 [DOI] [PubMed] [Google Scholar]
- 48.Castelli V, Lombardi A, Palomba E, et al. Immune checkpoint inhibitors in people living with HIV/AIDS: facts and controversies. Cells. 2021;10(9):10. doi: 10.3390/cells10092227 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Cook MR, Kim C. Safety and efficacy of immune checkpoint inhibitor therapy in patients with HIV infection and advanced-stage cancer: a systematic review. JAMA Oncol. 2019;5(7):1049–1054. doi: 10.1001/jamaoncol.2018.6737 [DOI] [PubMed] [Google Scholar]
- 50.Wu M, Zheng X, Zhang Y, et al. Camrelizumab for cancers in patients living with HIV: one-single center experience. AIDS Res Ther. 2023;20(1):23. doi: 10.1186/s12981-023-00518-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Spano JP, Veyri M, Gobert A, et al. Immunotherapy for cancer in people living with HIV: safety with an efficacy signal from the series in real life experience. AIDS. 2019;33(11):F13–F19. doi: 10.1097/QAD.0000000000002298 [DOI] [PubMed] [Google Scholar]
- 52.Eun Y, Kim IY, Sun JM, et al. Risk factors for immune-related adverse events associated with anti-PD-1 pembrolizumab. Sci Rep. 2019;9(1):14039. doi: 10.1038/s41598-019-50574-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Proietti I, Skroza N, Michelini S, et al. Efficacy of nivolumab in HIV patient with melanoma brain metastases. AIDS. 2020;34(9):1433–1435. doi: 10.1097/QAD.0000000000002554 [DOI] [PubMed] [Google Scholar]
- 54.Lurain K, Ramaswami R, Mangusan R, et al. Use of pembrolizumab with or without pomalidomide in HIV-associated non-Hodgkin's lymphoma. J Immunother Cancer. 2021;9(2):6–7. doi: 10.1136/jitc-2020-002097 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Burke MM, Kluger HM, Golden M, et al. Case Report: response to ipilimumab in a patient with HIV with metastatic melanoma. J Clin Oncol. 2011;29(32):e792–794. doi: 10.1200/JCO.2011.36.9199 [DOI] [PubMed] [Google Scholar]
- 56.Heppt MV, Schlaak M, Eigentler TK, et al. Checkpoint blockade for metastatic melanoma and Merkel cell carcinoma in HIV-positive patients. Ann Oncol. 2017;28(12):3104–3106. doi: 10.1093/annonc/mdx538 [DOI] [PubMed] [Google Scholar]
- 57.McCullar B, Alloway T, Martin M. Durable complete response to nivolumab in a patient with HIV and metastatic non-small cell lung cancer. J Thorac Dis. 2017;9(6):E540–E542. doi: 10.21037/jtd.2017.05.32 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Fukumura D, Kloepper J, Amoozgar Z, et al. Enhancing cancer immunotherapy using antiangiogenics: opportunities and challenges. Nat Rev Clin Oncol. 2018;15(5):325–340. doi: 10.1038/nrclinonc.2018.29 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Yang G, Fang Y, Zhou M, et al. Case report: the effective response to pembrolizumab in combination with bevacizumab in the treatment of a recurrent glioblastoma with multiple extracranial metastases. Front Oncol. 2022;12:948933. doi: 10.3389/fonc.2022.948933 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Arevalo OD, Soto C, Rabiei P, et al. Assessment of glioblastoma response in the era of bevacizumab: longstanding and emergent challenges in the imaging evaluation of pseudoresponse. Front Neurol. 2019;10:460. doi: 10.3389/fneur.2019.00460 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Mylvaganam G, Yanez AG, Maus M, Walker BD. Toward T cell-mediated control or elimination of HIV reservoirs: lessons from cancer immunology. Front Immunol. 2019;10:2109. doi: 10.3389/fimmu.2019.02109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cloughesy TF, Mochizuki AY, Orpilla JR, et al. Neoadjuvant anti-PD-1 immunotherapy promotes a survival benefit with intratumoral and systemic immune responses in recurrent glioblastoma. Nat Med. 2019;25(3):477–486. doi: 10.1038/s41591-018-0337-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Garzon-Muvdi T, Bailey DD, Pernik MN, Pan E. Basis for immunotherapy for treatment of meningiomas. Front Neurol. 2020;11:945. doi: 10.3389/fneur.2020.00945 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Galstyan A, Markman JL, Shatalova ES, et al. Blood-brain barrier permeable nano immunoconjugates induce local immune responses for glioma therapy. Nat Commun. 2019;10(1):3850. doi: 10.1038/s41467-019-11719-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Sharabi AB, Lim M, DeWeese TL, Drake CG. Radiation and checkpoint blockade immunotherapy: radiosensitisation and potential mechanisms of synergy. Lancet Oncol. 2015;16(13):e498–509. doi: 10.1016/S1470-2045(15)00007-8 [DOI] [PubMed] [Google Scholar]
- 66.Ferrara R, Signorelli D, Proto C, et al. Novel patterns of progression upon immunotherapy in other thoracic malignancies and uncommon populations. Transl Lung Cancer Res. 2021;10(6):2955–2969. doi: 10.21037/tlcr-20-636 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Okada H, Weller M, Huang R, et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 2015;16(15):e534–e542. doi: 10.1016/S1470-2045(15)00088-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Evans VA, van der Sluis RM, Solomon A, et al. Programmed cell death-1 contributes to the establishment and maintenance of HIV-1 latency. AIDS. 2018;32(11):1491–1497. doi: 10.1097/QAD.0000000000001849 [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Data Availability Statement
Availability of data and material: The datasets from this study are available from the corresponding author upon reasonable request.
Funding: No funding was received for this manuscript.