Abstract
Background:
Anti-programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) immunotherapy is now routinely used to treat several cancers. Clinical trials have excluded several populations, including patients with solid organ transplant, HIV infection and hepatitis B/C infection. We examined the safety outcomes of these populations treated with anti-PD-1/PD-L1 treatment in a multicentre retrospective study.
Methods:
Patients from 16 centres with advanced cancer and solid organ transplant, HIV infection or hepatitis B/C infection were included. Demographic, tumour, treatment, toxicity and outcome data were recorded.
Results:
Forty-six patients were included for analysis, with a median age of 60 years, and the majority of patients diagnosed with melanoma (72%). Among six patients with solid organ transplants, two graft rejections occurred, with one resulting in death, whereas two patients achieved partial responses. There were four responses in 12 patients with HIV infection. In 14 patients with hepatitis B, there were three responses, and similarly, there were three responses in 14 patients with hepatitis C. There was no unexpected toxicity in any viral infection group or an increase in viral load.
Conclusion:
Patients with HIV or hepatitis B/C infections treated with anti-PD-1/PD-L1 immunotherapy may respond to treatment without increased toxicity. Given the risk of graft rejection in solid organ transplant patients and also the potential for response, the role of anti-PD-1/PD-L1 immunotherapy needs to be carefully considered.
Keywords: PD-1, Immunotherapy, Cancer, HIV, Hepatitis B, Hepatitis C, Organ transplant
1. Introduction
The use of anti-programmed cell death protein 1/programmed death ligand 1 (PD-1/PD-L1) checkpoint inhibitors in the treatment of cancer has substantially broadened in scope over the past few years. Originally shown to have high response rates and improve survival in advanced melanoma [1,2], subsequent trials across multiple cancer types have also shown benefit, such that anti-PD-1/PD-L1 inhibitors are now standard therapy for many cancer patients [3–8].
The PD-1/PD-L1 axis is known to play a pivotal role in immune tolerance, with PD-1 expressed on activated T cells and PD-L1 expressed on a large range of normal tissue [9]. Mice lacking expression of PD-1 have a higher risk of developing autoimmune cardiomyopathy and a lupus-like syndrome [10,11], while there is emerging evidence that certain PD-1 polymorphisms in humans may increase the risk of autoimmune conditions [12]. The PD-1/PD-L1 axis also plays a role in chronic infection and organ transplant tolerance, with PD-1 upregulation associated with T cell exhaustion phenotypes in chronic HIV and hepatitis B and C [13–15] and multiple animal models indicating that an intact PD-1/PD-L1 axis is required for transplant tolerance [16–18].
The major trials of anti-PD-1/PD-L1 inhibitors have excluded a number of specific patient populations, including those with autoimmune conditions, solid organ transplants and chronic viral infections such as HIV and hepatitis B/C, yet these patients are at greater risk of developing cancer [19–21]. Recent evidence indicates that selected patients with pre-existing autoimmune conditions can be safely and effectively treated with anti-PD-1 therapy, with mild and reversible flares of their underlying condition [22–24].
Approximately 30,000 patients receive solid organ transplants each year in the United States of America alone [25], whereas 1.2 million have HIV [26], 730,000 have hepatitis B [27] and 3.5 million have hepatitis C [28]. Many of these patients will be diagnosed with cancers that may respond to PD-1/PD-L1–based immunotherapy. Whether these patients can be safely and effectively treated with these therapies is unclear, with current evidence largely based on individual case reports, case series or small early-phase trials [29–31]. We therefore sought to explore the outcomes with anti-PD-1/PD-L1–based immunotherapy in these populations in a multicentre retrospective study.
2. Methods
The study was performed with institutional ethical review board approval. Patients with advanced cancer and concurrent solid organ transplant, HIV, hepatitis B or hepatitis C infection who were treated with anti-PD-1–or PD-L1–based therapy between July 2014 and March 2017 were retrospectively identified from 16 centres.
Data collected included demographics, cancer details (subtype, stage and performance status), underlying disease factors (condition, viral load, CD4 count and organ function), concurrent immunosuppressive or antiviral therapy at treatment commencement and immunotherapy treatment.
Conventional immune-related adverse events (irAEs) were assessed by the Common Terminology Criteria for Adverse Events (CTCAE), and details of any change in underlying condition, treatment required and outcome were collected. Response to treatment was assessed by Response Evaluation Criteria in Solid Tumours criteria v1.1 [32]. Overall survival (OS) was calculated using the Kaplan–Meier estimate.
3. Results
3.1. Patients and treatment
Forty-six patients were included for analysis; 37 (80%) were male, with a median age of 60 years (Table 1). A wide spectrum of cancers were included, the majority being melanoma (N = 33, 72%). Most patients (N = 41, 89%) had American Joint Committee on Cancer stage IV disease, and almost all patients (N = 43, 94%) had Eastern Cooperative Oncology Group performance status 0 or 1. Most patients (N = 35, 76%) received anti-PD-1/PD-L1 monotherapy, whereas seven and four patients (15% and 9%) received sequential and concurrent PD-1 and cytoxic T-lymphocyte antigen-4 (CTLA-4) therapy (ipilimumab), respectively. Six patients had a solid organ transplant, 12 patients had HIV, 14 patients had hepatitis B and 14 patients had hepatitis C.
Table 1.
Baseline characteristics | Number | Percentage |
---|---|---|
Median age | 60 | – |
Male | 37 | 80% |
ECOG performance status | ||
0 | 20 | 44% |
1 | 23 | 50% |
2 | 1 | 2% |
Unknown | 2 | 4% |
Cancer subtype | ||
Melanoma | 33 | 72% |
Urothelial carcinoma | 4 | 9% |
Renal cell carcinoma | 2 | 4% |
Mesothelioma | 2 | 4% |
Hepatocellular carcinoma | 2 | 4% |
Non-small cell lung cancer | 1 | 2% |
Gastric cancer | 1 | 2% |
Glioblastoma | 1 | 2% |
AJCC stage | ||
I | 1 | 2% |
II | 1 | 2% |
III | 3 | 7% |
IV | 41 | 89% |
Underlying condition | ||
Solid organ transplant | 6 | 13% |
HIV | 12 | 6% |
Hepatitis B | 14 | 30% |
Hepatitis C | 14 | 30% |
Immunotherapy | ||
Pembrolizumab | 21 | 46% |
Nivolumab | 12 | 26% |
Atezolizumab | 2 | 4% |
Sequential PD-1 then CTLA-4 | 7 | 15% |
Concurrent PD-1 and CTLA-4 | 4 | 9% |
Total | 46 |
ECOG: Eastern Cooperative Oncology Group; AJCC: American Joint Committee on Cancer.
The median OS of the cohort was 19.4 months (95% confidence interval, 13.2–25.6). Twelve patients (26%) had an objective response, and a total of 32 patients (70%) had disease control (complete/partial response or stable disease). irAEs occurred in 17 patients (37%), with five irAEs grade 3 or higher (11%).
3.2. Solid organ transplants
Six patients with a history of solid organ transplant were included, five with renal transplants and one with a liver transplant (Table 2). All had advanced melanoma; five were treated with anti-PD-1 monotherapy, and one was treated with sequential pembrolizumab and then ipilimumab. All patients were on immunosuppression at the start of immunotherapy.
Table 2.
Organ | Cancer | Age | Gender | Immunotherapy | Immunosuppression | Toxicity | Response |
---|---|---|---|---|---|---|---|
Kidney | Melanoma | 65 | M | Sequential pembrolizumab + ipilimumab | Prednisone, mycophenolate and everolimus | G2 pneumonitis | PD |
Kidney | Melanoma | 70 | M | Pembrolizumab | Tacrolimus and prednisone | – | PD |
Kidney | Melanoma | 75 | M | Pembrolizumab | Prednisone | – | PR |
Kidney | Melanoma | 65 | M | Pembrolizumab | Prednisone, mycophenolate and tacrolimus | – | PD |
Kidney | Melanoma | 48 | M | Nivolumab | Prednisone and tacrolimus | G4 acute rejection of kidney transplant resulting in graft failure, requiring dialysis | PR |
Liver | Melanoma | 63 | F | Pembrolizumab | Cyclosporine | G5 acute rejection of liver transplant. Death within 18 days of the first cycle |
NE |
PR: partial response; PD: disease progression; irAE: immune-related adverse event; NE; not evaluable.
Two patients (33%) developed graft rejection. One patient had acute rejection of a kidney transplant after one cycle of nivolumab, with subsequent graft failure despite IV methylprednisolone, and the sole liver transplant patient had acute rejection after one cycle of pembrolizumab, resulting in death after 18 days despite IV methylprednisolone, increased cyclosporine dosing and addition of tacrolimus and mycophenolate. A third patient had grade 2 pneumonitis, which responded to steroids. Two patients (33%) had partial responses, whereas the remainder had disease progression (N = 3) or death from toxicity (N = 1).
3.3. Patients with HIV
Twelve patients had concurrent HIV. Nine (75%) had melanoma, and three (25%) were co-infected with hepatitis B and/or C (Table 3). Seven patients (58%) were treated with anti-PD-1/PD-L1 monotherapy, with five patients (42%) treated with sequential or concurrent PD-1 and CTLA-4 immunotherapy. Eleven patients (92%) were on anti-retroviral therapy (ART) at commencement (one unknown). Seven patients (58%) had viral loads recorded before commencement, with four undetectable (25%) and a median of CD4 count of 492 in the patients with evaluable counts. Seven (58%) had viral loads recorded during treatment.
Table 3.
Cancer | Intercurrent infection | Immunotherapy | ART | CD4 count | Viral load | Response | IrAE |
---|---|---|---|---|---|---|---|
Melanoma | – | Concurrent ipilimumab + nivolumab | Rilpivirine, emtricitabine and tenofovir | 285-> 350-> 525 |
<20 -> <20 |
PR | – |
Melanoma | Hep B | Concurrent ipilimumab + nivolumab | Raltegravir, emtricitabine and tenofovir | PR | G3 colitis | ||
Melanoma | Hep C | Sequential pembrolizumab + ipilimumab | Emtricitabine, tenofovir and dolutegravir | 206- >269- >239- >347 |
160,624 -> <20 -> 30 |
SD | G2 right facial palsy |
Melanoma | – | Sequential pembrolizumab + ipilimumab | Efavirenz, emtricitabine and tenofovir | – | – | PD | – |
Melanoma | – | Sequential nivolumab + ipilimumab | Nevirapine, abacavir and lamivudine | – | – | PD | – |
Melanoma | – | Pembrolizumab | Unknown | – | <20 | SD | G2 nephritis |
Melanoma | – | Pembrolizumab | Raltegravir, tenofovir and emtricitabine | 613-> 700 |
<20-> <20 |
CR | – |
Melanoma | – | Pembrolizumab | Abacavir, darunavir and ritonavir | – | <40 -> <40 |
PD | – |
Melanoma | – | Pembrolizumab | Ritonavir, emtricitabine and tenofovir | – | – | SD | – |
Urothelial carcinoma | – | Atezolizumab | Emtricitabine and tenofovir | 492 | – | PD | – |
HCC | Hep B + C | Pembrolizumab | Emtricitabine, tenofovir and dolutegravir | 550- >550- >484 |
19,440-> <20 -> <20 |
PR | G2 hypothyroidism |
RCC | – | Nivolumab | Atazanavir, ritonavir, tenofovir and emtricitabine | <20 | PD | G1 diarrhoea |
ART: anti-retroviral therapy; CR: complete response; PR: partial response; SD: stable disease; PD: disease progression; irAE: immune-related adverse event.
Five patients (42%) developed conventional irAEs, with the only grade 3 event (colitis) occurring in a patient treated with concurrent ipilimumab and nivolumab. No HIV viral loads increased by one log or more during therapy, and there were two patients with HIV viral loads that decreased by one log or more during therapy; however, both were on ART. There were no events of immune reconstitution inflammatory syndrome. Four patients (33%) had a complete or partial response to therapy.
3.4. Patients with hepatitis B and C
Fourteen patients had hepatitis B; ten patients (71%) had melanoma and thirteen patients (93%) were treated with anti-PD-1/PD-L1 monotherapy (Table 4). Eight patients (57%) had viral loads measured at the start of treatment, four patients (29%) had an undetectable viral load at baseline and eight patients (57%) were on antiviral therapy. Five patients (36%) developed grade 1–2 conventional irAEs, with no higher grade events. No patients developed hepatitis, and no patients had a viral load increase of more than one log during therapy. Two patients had a viral load decrease of more than one log, with both patients being on concurrent anti-viral therapy. There were no viral load increases of one log or more. Three patients (21%) had a complete or partial response to therapy.
Table 4.
Cancer | Viral hepatitis | Immunotherapy | Anti-viral therapy | Viral load (IU/ml) | Response | IrAE |
---|---|---|---|---|---|---|
Melanoma | Hepatitis B | Sequential pembrolizumab + ipilimumab | Nil | <20-> 20-> <20-> <20 |
SD | – |
Melanoma | Hepatitis B | Pembrolizumab | Tenofovir | <20-> 20-> <20-> <20 |
SD | – |
Melanoma | Hepatitis B | Pembrolizumab | Tenofovir | Unknown | PR | G2 hypothyroidism |
Melanoma | Hepatitis B | Pembrolizumab | Entecavir | Unknown | PD | – |
Melanoma | Hepatitis B | Pembrolizumab | Nil | Unknown | PD | – |
Melanoma | Hepatitis B | Pembrolizumab | Nil | <20-> <20-> <20-> <20 |
CR | G1 rash |
Melanoma | Hepatitis B | Pembrolizumab | Nil | Unknown | PD | G2 rash |
Melanoma | Hepatitis B | Nivolumab | Entecavir | 279-> 20-> <20-> 43 |
SD | G2 pneumonitis |
Melanoma | Hepatitis B | Nivolumab | Nil | 300-> 170 | PR | G1 vitiligo |
Mesothelioma | Hepatitis B | Pembrolizumab | Entecavir | 467-> <20-> <20-> <20 |
SD | – |
Hepatocellular carcinoma | Hepatitis B | Nivolumab | Tenofovir | 39-> <20-> <20-> <20 |
SD | – |
Glioblastoma | Hepatitis B | Pembrolizumab | Entecavir | <20-> 20-> <20-> <20 |
SD | – |
Gastric carcinoma | Hepatitis B | Pembrolizumab | Nil | Unknown | SD | – |
Urothelial carcinoma | Hepatitis B | Nivolumab | Entecavir | Unknown | SD | – |
Melanoma | Hepatitis C | Concurrent nivolumab + ipilimumab | Sofosbuvir and ribavirin | 6,550,000- > undetectable |
CR | G4 colitis |
Melanoma | Hepatitis C | Concurrent nivolumab + ipilimumab | Ledipasvir and sofosbuvir | 9,360,000 | PR | G3 autoimmune hepatitis |
Melanoma | Hepatitis C | Sequential nivolumab + ipilimumab | Ombitasvir, paritaprevir, ritonavir and dasabuvir | 1,218,319-> 920,912 |
PD | – |
Melanoma | Hepatitis C | Sequential pembrolizumab + ipilimumab | Interferon | 58,000 | CR | – |
Melanoma | Hepatitis C | Pembrolizumab | Nil | Unknown | SD | G3 adrenal insufficiency |
Melanoma | Hepatitis C | Pembrolizumab | Nil | Unknown | SD | – |
Melanoma | Hepatitis C | Pembrolizumab | Ribavirin and pegylated interferon | Undetectable | PD | – |
Melanoma | Hepatitis C | Nivolumab | Nil | Undetectable | SD | – |
Melanoma | Hepatitis C | Nivolumab | Nil | Unknown | SD | – |
Urothelial carcinoma | Hepatitis C | Nivolumab | Ribavirin and pegylated interferon | 81,159 | SD | G1 rash |
Urothelial carcinoma | Hepatitis C | Atezolizumab | Ledipasvir and sofosbuvir | Undetectable | SD | – |
Mesothelioma | Hepatitis C | Pembrolizumab | Unknown | 903,029 | SD | – |
Renal cell carcinoma | Hepatitis C | Nivolumab | Ledipasvir and sofosbuvir | 379,836- > Undetectable |
SD | – |
Non-small cell lung cancer | Hepatitis C | Nivolumab | Ledipasvir and sofosbuvir | 3,952,419- > undetectable |
SD | – |
CR: complete response; PR: partial response; SD: stable disease; PD: disease progression; irAE: immune-related adverse event.
Fourteen patients had hepatitis C, nine (64%) had melanoma and ten (71%) were treated with anti-PD-1/PD-L1 monotherapy (Table 4). Eleven patients (79%) had viral loads measured at the start of treatment. Three patients (21%) had an undetectable viral load at baseline, and nine patients (64%) were on anti-viral therapy. Five patients (36%) had conventional irAEs, three (21%) of which were grade 3 or higher. Of note, one patient had grade 3 autoimmune hepatitis on concurrent ipilimumab and nivolumab; however, this patient’s viral load was unavailable beyond the baseline measurement, and therefore the role of hepatitis C in this irAE is uncertain. Three patients (21%) had a viral load decrease of more than one log, with all three patients on concurrent anti-viral therapy. There were no viral load increases of one log or more. Three patients (21%) had a complete or partial response to therapy.
4. Discussion
The use of anti-PD-1/PD-L1 therapies for patients with cancer is rapidly increasing, but the safety and efficacy in populations with solid organ transplants or with chronic viral infections are largely unknown. In this series of patients with chronic viral infections—HIV or hepatitis B or C—we found that anti-PD-1/PD-L1 therapies can be safety used, with responses in up to a third of patients. The rates of both toxicity and efficacy appeared relatively similar to the rates observed in patients in clinical trials without such infections [33,34]. There were no increases in viral loads to suggest that anti-PD-1/PD-L1 therapy interferes with control of infection; on the contrary, a total of five patients with hepatitis B or C and two patients with HIV had a decrease in viral loads while treated with anti-PD-1/PD-L1 therapy. Patients with solid organ transplantation, however, appear at a significant risk of early graft rejection and loss yet may also respond without toxicity.
Evidence from animal models with HIV and simian immunodeficiency virus (SIV) indicate that PD-1 blockade may contribute to improved viral load, antiviral immunity and survival [35,36], although a transient early increase in viral load before a reduction in viral load has been described in one SIV model [37] and a similar phenomenon has been reported in a single case report [38]. Similar to the HIV literature, there are supporting data from an animal model that PD-1/PD-L1 blockade can contribute to viral clearance of hepatitis B [39], and a small randomised trial of nivolumab in patients with hepatitis C but without malignancy showed promising reductions in viral load; however, only five of 42 patients treated with nivolumab met the primary end-point of a reduction in viral load of more than 0.5 log reduction [40]. However, two cases of reactivation of hepatitis B and one case of reactivation of hepatitis C have been reported [41–43]. In total, our data, coupled with similar experiences in the literature [29,30,44–46], support the use of anti-PD-1/PD-L1 therapy in patients with hepatitis B/C or HIV.
By contrast, previous case reports and data from this series suggest that there is a significant risk of graft rejection and death with anti-PD-1/PD-L1 therapy in patients with solid organ transplantation. Previous case reports have included 13 kidney [29,47–57], 14 liver [41,58–62] and two heart transplant recipients receiving anti-PD-1/PD-L1ebased immunotherapy [29], [63]. In total, eight of thirteen reported kidney transplant cases have resulted in rejection, one of two heart transplant cases resulted in rejection and four of fourteen liver transplant cases resulted in rejection. Adding these cases together with our series suggests a rate of rejection of nine out of eighteen (50%) in kidney transplant recipients and a rate of rejection of five out of fourteen (36%) in liver transplant recipients.
While responses occurred both in our cohort as well as in transplant recipients previously reported in the literature, the benefit of using anti-PD-1/PD-L1 therapy in this setting needs to be carefully weighed against the significant risk of transplant rejection and death. Although there is evidence suggesting that CTLA-4—based immunotherapy is associated with a lower risk of rejection and therefore perhaps could be considered in the first instance, the activity of CTLA-4 immunotherapy is low [29], [64]. In the circumstance where transplant rejection and graft failure are not acceptable outcomes, such as cardiac, lung or liver transplants, or renal transplant recipients who are not prepared to undergo dialysis, alternative regimens should be prioritised.
Our study had a number of limitations. There were limited longitudinal data on viral load during treatment for the HIV and hepatitis B and C cohorts, and similarly, there were limited data on anti-viral changes during treatment. Data on the degree of matching between transplant and recipient were unavailable, as well as immunosuppressant dosing and regimen changes during treatment. Given the retrospective observational design of this study, selection bias is also a potential limitation, however less so than in the case reports published so far. Further prospective studies are needed.
5. Conclusions
Patients with HIV or hepatitis B/C treated with anti-PD-1/PD-L1 therapy for malignancy can respond to immunotherapy with no apparent increase in toxicity compared with uninfected patients. In patients with solid organ transplants, graft rejection with graft failure and death can occur; alternatives should be sought where possible, and in the absence of alternatives, the potential benefits of anti-PD-1/PD-L1 therapy need to be carefully weighed against the mortality risk of untreated malignancy. These results provide not only important clinical information but also biological insight into the fundamental role and diverse function of the PD-1/PD-L1 pathway in cancer, infection and tolerance. Further research in these populations is warranted.
Funding source
None.
Footnotes
Conflict of interest statement
Chloe Khoo received travel and conference support from Roche. Lisa Zimmer served a consultant for Roche, Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Sanofi and Pierre Fabre; received honoraria from Roche, Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Merck KGaA and Pierre Fabre and support for scientific meetings from Bristol-Myers Squibb, Merck Sharp & Dohme, Novartis, Amgen and Pierre Fabre. Michael Millward was an advisory board member for Bristol-Myers Squibb, Roche, AstraZeneca and Merck Sharp & Dohme. Paolo A. Ascierto served as a consultant for Bristol-Myers Squibb, Roche-Genentech, Merck Sharp & Dohme, Novartis, Amgen, Array, Merck Serono, Pierre Fabre, Incyte, NewLink Genetics, Genmab, Medimmune, AstraZeneca, Syndax, Sun Pharma, Sanofy, Idera and Ultimovacs and received research funding from Bristol-Myers Squibb, Roche-Genentech and Array. Douglas B. Johnson was an advisory board member for Array, Bristol-Myers Squibb, Incyte and Merck and received research funding from Bristol-Myers Squibb and Incyte. Victoria Atkinson was an advisory board member for Bristol-Myers Squibb, Merck Sharp & Dohme, Pierre Fabre and Novartis and received travel support from Bristol-Myers Squibb and speakers fee from Merck Sharp & Dohme, Novartis and Bristol-Myers Squibb. Simone M. Goldinger was an advisory board member and received travel grant support from Roche, Bristol-Myers Squibb, Merck Sharp & Dohme and Novartis. Claus Garbe received grant support from Bristol-Myers Squibb, Novartis and Roche and personal fees from Amgen, Bristol-Myers Squibb, Novartis, Roche, Merck Sharp & Dohme and Philogen. Ralf Gutzmer received lecture fees from Amgen, Boehringer Ingelheim, AstraZeneca, Roche, Bristol-Myers Squibb, Novartis, Pierre Fabre, Merck-EMD and Almirall-Hermal; honoraria from Amgen, Leo Pharma, Inctye, Roche, Bristol-Myers Squibb, Novartis, Regeneron, 4SC, Pierre Fabre, Merck-EMD and Almirall-Hermal; patient fees from Amgen, Roche, Bristol-Myers Squibb, Novartis, Regenerom, 4SC, Philogen and Array; grant support from Novartis, Pfizer and Johnson & Johnson and support for scientific meetings from Roche, Bristol-Myers Squibb, Pierre Fabre. Matthew Hellman was a consultant for Bristol-Myers Squibb, Merck, Genentech, Roche, Astra-Zeneca, MedImmune, Novartis and Janssen and received research funding from Bristol-Myers Squibb. Georgina V. Long was a consultant for Array, Bristol-Myers Squibb, Novartis, Roche, Amgen, Pierre Fabre, Merck Sharp & Dohme and Incyte. Alexander M. Menzies was an advisory board member for Merck Sharp & Dohme, Chugai, Novartis and Pierre Fabre and received honoraria from Bristol-Myers Squibb and Novartis. All other authors report no conflicts of interest to disclose.
Writing assistance
None.
References
- [1].Robert C, Long GV, Brady B, et al. Nivolumab in previously untreated melanoma without BRAF mutation. N Engl J Med 2015;372(4):320–30. [DOI] [PubMed] [Google Scholar]
- [2].Robert C, Schachter J, Long GV, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med 2015;372(26):2521–32. [DOI] [PubMed] [Google Scholar]
- [3].Borghaei M, Paz-Ares L, Horn L, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med 2015;373(17):1627–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Bellmunt J, de Wit R, Vaughn DJ, et al. Pembrolizumab as second-line therapy for advanced urothelial carcinoma. N Engl J Med 2017;376(11):1015–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Lemery S, Keegan P, Pazdur R. First FDA approval agnostic of cancer site — when a biomarker defines the indication. N Engl J Med 2017;377(15):1409–12. [DOI] [PubMed] [Google Scholar]
- [6].Weber J, Mandala M, Del Vecchio M, et al. Adjuvant nivolumab versus ipilimumab in resected stage III or IV melanoma. N Engl J Med 2017;377(19):1824–35. [DOI] [PubMed] [Google Scholar]
- [7].Antonia SJ, Villegas A, Daniel D, et al. Durvalumab after chemoradiotherapy in stage III non-small-cell lung cancer. N Engl J Med 2017;377(20):1919–29. [DOI] [PubMed] [Google Scholar]
- [8].Kang YK, Boku N, Satoh T, et al. Nivolumab in patients with advanced gastric or gastro-oesophageal junction cancer refractory to, or intolerant of, at least two previous chemotherapy regimens (ONO-4538-12, ATTRACTION-2): a randomized, double-blind, placebo-controlled, phase 3 trial. Lancet 2017;390(10111):2461–71. [DOI] [PubMed] [Google Scholar]
- [9].Sun C, Mezzadra R, Schumacher TN. Regulation and function of the PD-L1 checkpoint. Immunity 2018;48(3):434–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [10].Nishimura H, Okazaki T, Tanaka Y, et al. Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001;291(5502):319–22. [DOI] [PubMed] [Google Scholar]
- [11].Nishimura H, Nose M, Hiai H, et al. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity 1999;11: 141–51. [DOI] [PubMed] [Google Scholar]
- [12].Gianchechi E, Delfino DV, Fierabracci A. Recent insights into the role of the PD-1/PD-L1 pathway in immunological tolerance and autoimmunity. Autoimmun Rev 2013;12(11):1091–10013. [DOI] [PubMed] [Google Scholar]
- [13].Trautmann L, Janbazian L, Chomont N, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nat Med 2006;12(10):1198–202. [DOI] [PubMed] [Google Scholar]
- [14].Urbani S, Amadei B, Tola D, et al. PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. J Virol 2006;80(22):11398–403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [15].Peng G, Li S, Wu W, et al. PD-1 upregulation is associated with HBV-specific T cell dysfunction in chronic hepatitis B patients. Mol Immunol 2008;45(4):963–70. [DOI] [PubMed] [Google Scholar]
- [16].Yang J, Popoola J, Khandwala S, et al. Critical role of donor tissue expression of programmed death ligand-1 in regulating cardiac allograft rejection and vasculopathy. Circulation 2008;117(5):660–9. [DOI] [PubMed] [Google Scholar]
- [17].Wang L, Han R, Hancock WW. Programmed cell death (PD-1) and its ligand PD-L1 are required for allograft tolerance. Eur J Immunol 2007;37(10):2983–90. [DOI] [PubMed] [Google Scholar]
- [18].Tanaka K, Albin MJ, Yuan X, et al. PDL1 is required for peripheral transplantation tolerance and protection from chronic allograft rejection. J Immunol 2007;179(8):5204–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Engels EA, Biggar RJ, Hall HI, et al. Cancer risk in people infected with human immunodeficiency virus in the United States. Int J Cancer 2008;123(1):187–94. [DOI] [PubMed] [Google Scholar]
- [20].Engels EA, Pfeiffer RM, Fraumeni JF, et al. Spectrum of cancer risk among US solid organ transplant recipients. JAMA 2011;306(17):1891–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Perz JF, Armstrong GL, Farrington LA, et al. The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. J Hepatol 2006;45(4):529–38. [DOI] [PubMed] [Google Scholar]
- [22].Menzies AM, Johnson DB, Ramanujam S, et al. Anti-Pd-1 therapy in patients with advanced melanoma and preexisting autoimmune disorders or major toxicity with ipilimumab. Ann Oncol 2017;28(2):368–76. [DOI] [PubMed] [Google Scholar]
- [23].Gutzmer R, Koop A, Meier F, et al. Programmed cell death protein-1 (PD-1) inhibitor therapy in patients with advanced melanoma and preexisting autoimmunity or ipilimumab-triggered autoimmunity. Eur J Cancer 2017;75:24–32. [DOI] [PubMed] [Google Scholar]
- [24].Leonardi GC, Gainor JF, Altan M, et al. Safety of programmed death-1 pathway inhibitors among patients with non-small-cell lung cancer and preexisting autoimmune disorders. J Clin Oncol 2018;36(19):1905–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Organ procurement and transplantation network. h ttps://optn.transplant.hrsa.gov/data/view-data-reports/national-data/#. [Accessed 11 May 2018].
- [26].Bonacci RA, Holtgrave DR. Simplified estimates of HIV and incidence and transmission rates for the USA, 2008–12. AIDS 2016;30(2):332–3. [DOI] [PubMed] [Google Scholar]
- [27].Wasley A, Kruszon-Moran D, Kuhnert W, et al. The prevalence of hepatitis B virus infection in the United States in the era of vaccination. J Infect Dis 2010;202(2):192–201. [DOI] [PubMed] [Google Scholar]
- [28].Edlin BR, Eckhardt BJ, Shu MA, et al. Toward a more accurate estimate of the prevalence of hepatitis C in the United States. Hepatology 2015;62(5):1353–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Kittai AS, Oldham H, Cetnar J, et al. Immune checkpoint inhibitors in organ transplant patients. J Immunother 2017;40(7):277–81. [DOI] [PubMed] [Google Scholar]
- [30].Uldrick TS, Goncalves PH, Fling SP, et al. Interim safety analysis of Cancer Immunotherapy Trials Network-12 (CITN-12): a phase 1 study of pembrolizumab in patients with HIV and relapsed, refractory, or disseminated malignancies. J Immunother Cancer 2017;5(Supple 3):O40. [Google Scholar]
- [31].El-Khoueiry AB, Sangro B, Yau T, et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017;389(10088):2492–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [32].Eisenhauer EA, Therasse P, Bogaerts J, et al. New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). Eur J Cancer 2009;45(2):228–47. [DOI] [PubMed] [Google Scholar]
- [33].Schachter J, Ribas A, Long GV, et al. Pembrolizumab versus ipilimumab for advanced melanoma: final overall survival results of a multicenter, randomized, open-label phase 3 study (KEY-NOTE-006). Lancet 2017;390(10105):1853–62. [DOI] [PubMed] [Google Scholar]
- [34].Wolchok JD, Chiarion-Sileni V, Gonzalez R, et al. Overall survival with combined nivolumab and ipilimumab in advanced melanoma. N Engl J Med 2017;377(14):1345–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [35].Velu V, Titanji K, Zhu B, et al. Enhancing SIV-specific immunity in vivo by PD-1 blockade. Nature 2009;458(7235):206–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [36].Seung E, Dudek TE, Allen TM, et al. PD-1 blockade in chronically HIV-1-infected humanized mice suppresses viral loads. PLoS One 2013;8(10), e77780. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [37].Finnefrock AC, Tang A, Li F, et al. PD-1 blockade in rhesus macaques: impact on chronic infection and prophylactic vaccination. J Immunol 2009;182(2):980–7. [DOI] [PubMed] [Google Scholar]
- [38].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):517–8. [DOI] [PubMed] [Google Scholar]
- [39].Tzeng HT, Tsai HF, Liao HJ, et al. PD-1 blockage reverses immune dysfunction and hepatitis B viral persistence in a mouse animal model. PLoS One 2012;7(6), e39179. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [40].Gardiner D, Lalezari J, Lawitz E, et al. A randomized, double-blind, placebo-controlled assessment of BMS-936558, a fully human monoclonal antibody to programmed death-1 (PD-1), in patients with chronic hepatitis C virus infection. PLoS One 2013;8(5), e63818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [41].Schvartsman G, Perez K, Sood G, et al. Immune checkpoint inhibitor therapy in a liver transplant recipient with melanoma. Ann Intern Med 2017;167(5):361–2. [DOI] [PubMed] [Google Scholar]
- [42].Koksal AS, Toka B, Eminler AT, et al. HBV-related acute hepatitis due to immune checkpoint inhibitors in a patient with malignant melanoma. Ann Oncol 2017;28(12):3103–4. [DOI] [PubMed] [Google Scholar]
- [43].Lake AC. Hepatitis B reactivation in a long-term nonprogressor due to nivolumab therapy. AIDS 2017;31(15):2115–8. [DOI] [PubMed] [Google Scholar]
- [44].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):1065–6. [DOI] [PubMed] [Google Scholar]
- [45].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):3104–6. [DOI] [PubMed] [Google Scholar]
- [46].Wen X, Wang Y, Ding Y, et al. Safety of immune checkpoint inhibitors in Chinese patients with melanoma. Melanoma Res 2016;26(3):284–9. [DOI] [PubMed] [Google Scholar]
- [47].Kwatra V, Karanth NV, Priyadarshana K, et al. Pembrolizumab for metastatic melanoma in a renal allograft recipient with subsequent graft rejection and treatment response failure: a case report. J Med Case Rep 2017;11(1):73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [48].Lipson EJ, Bagnasco SM, Moore J Jr, et al. Tumor regression and allograft rejection after administration of Anti-PD-1. N Engl J Med 2016;374(9):896–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [49].Cl Boils, Aljadir DN Cantafio AW. Use of the PD-1 pathway inhibitor nivolumab in a renal transplant patient with malignancy. Am J Transplant 2016;16(8):2496–7. [DOI] [PubMed] [Google Scholar]
- [50].Ong M, Ibrahim AM, Bourassa-Blanchette S, et al. Antitumor activity of nivolumab on hemodialysis after renal allograft rejection. J Immunother Cancer 2016;4:64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [51].Herz S, Hofer T, Papapanagiotou M, et al. Checkpoint inhibitors in chronic kidney failure and an organ transplant recipient. Eur J Cancer 2016;67:66–72. [DOI] [PubMed] [Google Scholar]
- [52].Alhamad T, Venkatachalam K, Linette GP, et al. Checkpoint inhibitors in kidney transplant recipients and the potential risk of rejection. Am J Transplant 2016;16(4):1332–3. [DOI] [PubMed] [Google Scholar]
- [53].Winkler JK, Gutzmer R, Bender C, et al. Safe administration of an Anti-PD-1 antibody to kidney-transplant patients: 2 clinical cases and review of the literature. J Immunother 2017;40(9):341–4. [DOI] [PubMed] [Google Scholar]
- [54].Deltcombe C, Garandeau C, Renaudin K, et al. Severe allograft rejection and autoimmune hemolytic anemia after Anti-PD-1 therapy in a kidney transplanted patient. Transplantation 2017;101(9), e291. [DOI] [PubMed] [Google Scholar]
- [55].Miller DM, Faulkner-Jones BE, Stone JR, et al. Complete pathological response of metastatic cutaneous squamous cell carcinoma and allograft rejection after treatment with combination immune checkpoint blockade. JAAD Case Rep 2017;3(5):412–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [56].Barnett R, Barta VS, Jhaveri KD. Preserved renal-allograft function and the PD-1 pathway inhibitor nivolumab. N Engl J Med 2017;376(2):191–2. [DOI] [PubMed] [Google Scholar]
- [57].Tamain M, Garrouste C, Aguilera D, et al. Mixed acute kidney allograft rejection after an antiprogrammed cell death protein 1 antibody treatment for lung epidermoid carcinoma. Transpl Int 2016;29(11):1247–8. [DOI] [PubMed] [Google Scholar]
- [58].Friend BD, Venick RS, McDiarmid SV, et al. Fatal orthotopic liver transplant organ rejection induced by a checkpoint inhibitor in two patients with refractory, metastatic hepatocellular carcinoma. Pediatr Blood Cancer 2017;64(12), e26682. [DOI] [PubMed] [Google Scholar]
- [59].De Toni EN, Gerbes AL. Tapering of immunosuppression and sustained treatment with nivolumab in a liver transplant recipient. Gastroenterology 2017;152(6):1631–3. [DOI] [PubMed] [Google Scholar]
- [60].Kuo JC, Lilly LB, Hogg D. Immune checkpoint inhibitor therapy in a liver transplant recipient with a rare subtype of melanoma: a case report and literature review. Melanoma Res 2018;28(1):61–4. [DOI] [PubMed] [Google Scholar]
- [61].Varakis A, Lewis DW, Nugent FW. Preserved liver transplant after PD-1 pathway inhibitor for hepatocellular carcinoma. Am J Gastroenterol 2017;112(12):1895–6. [DOI] [PubMed] [Google Scholar]
- [62].Deleon TT, Salomao MA, Aqel BA, et al. Pilot evaluation of PD-1 inhibition in metastatic cancer patients with a history of liver transplantation: the Mayo Clinic experience. J Gastrointest Oncol 2018:1–9 [published ahead of print]. [DOI] [PMC free article] [PubMed]
- [63].Owonikoko TK, Kumar M, Yang S, et al. Cardiac allograft rejection as a complication of PD-1 checkpoint blockade for cancer immunotherapy: a case report. Cancer Immunol Immun-other 2017;66(1):45–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [64].Chae YK, Galvez C, Anker JF, et al. Cancer immunotherapy in a neglected population: the current use and future of T-cell-mediated checkpoint inhibitors in organ transplant patients. Cancer Treat Rev 2018;63:116–21. [DOI] [PubMed] [Google Scholar]