1
|
Miller-Handley H, Harper G, Pham G, Turner LH, Shao TY, Russi AE, Erickson JJ, Ford ML, Araki K, Way SS. Immune suppression sustained allograft acceptance requires PD1 inhibition of CD8+ T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:192-198. [PMID: 40073258 PMCID: PMC11904129 DOI: 10.1093/jimmun/vkae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Accepted: 10/24/2024] [Indexed: 03/14/2025]
Abstract
Organ transplant recipients require continual immune-suppressive therapies to sustain allograft acceptance. Although medication nonadherence is a major cause of rejection, the mechanisms responsible for graft loss in this clinically relevant context among individuals with preceding graft acceptance remain uncertain. Here, we demonstrate that skin allograft acceptance in mice maintained with clinically relevant immune-suppressive therapies, tacrolimus and mycophenolate, sensitizes hypofunctional PD1hi graft-specific CD8+ T cells. Uninterrupted immune-suppressive therapy is required because drug discontinuation triggers allograft rejection, replicating the requirement for immune-suppressive therapy adherence in transplant recipients. Graft-specific CD8+ T cells in allograft-accepted mice show diminished effector differentiation and cytokine production, with reciprocally increased PD1 expression. Allograft acceptance-induced PD1 expression is essential, as PDL1 blockade reinvigorates graft-specific CD8+ T cell activation with ensuing allograft rejection despite continual immune-suppressive therapy. Thus, PD1 sustained CD8+ T cell inhibition is essential for allograft acceptance maintained by tacrolimus plus mycophenolate. This necessity for PD1 in sustaining allograft acceptance explains the high rates of rejection in transplant recipients with cancer administered immune checkpoint inhibitors targeting PD1/PDL1, highlighting shared immune suppression pathways exploited by tumor cells and current therapies for averting allograft rejection.
Collapse
Affiliation(s)
- Hilary Miller-Handley
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
- Department of Medicine, University of Cincinnati College of Medicine
| | - Gavin Harper
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Giang Pham
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Lucien H. Turner
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Tzu-Yu Shao
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Abigail E. Russi
- Division of Gastroenterology, Hepatology and Advanced Nutrition, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - John J. Erickson
- Division of Neonatology, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Mandy L. Ford
- Winship Cancer Institute, Emory University School of Medicine
| | - Koichi Araki
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| | - Sing Sing Way
- Division of Infectious Diseases, Center for Inflammation and Tolerance, Cincinnati Children’s Hospital, Department of Pediatrics, University of Cincinnati College of Medicine
| |
Collapse
|
2
|
Struckmeier AK, Gosau M, Smeets R. Cutaneous squamous cell carcinoma in solid organ transplant recipients: Current therapeutic and screening strategies. Transplant Rev (Orlando) 2024; 38:100882. [PMID: 39348772 DOI: 10.1016/j.trre.2024.100882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024]
Abstract
Solid organ transplant recipients (SOTRs) are particularly prone to developing malignancies, often manifesting multiple tumors and tumors with a heightened susceptibility to metastasis, resulting in much lower survival rates when compared to the general population. Among these, cutaneous squamous cell carcinoma (CSCC) respresent a major challenge in terms of morbidity and mortality following organ transplantation. The management of post-transplant CSCC requires expertise from various disciplines, including dermatology, maxillofacial surgery, transplant medicine, radiation oncology, and medical oncology. Furthermore, the unique behaviors and prevalence of tumors in SOTRs necessitate tailored pathways for screening and treatment, distinct from those designed for immunocompetent patients. Despite the proven efficacy of immune checkpoint inhibitors (ICIs) in several cancers, SOTRs have often been systematically excluded from clinical trials due to concerns about potential allograft rejection and loss. Consequently, most data on the safety and efficacy of ICIs in SOTRs are derived from case series and reports. Given the significant risks involved, alternative therapeutic options should be thoroughly discussed with patients before considering ICI therapy. This literature review aims to provide an overview of CSCC in SOTRs, with a specific emphasis on therapeutic and screening strategies, particularly highlighting immunotherapy.
Collapse
Affiliation(s)
- Ann-Kristin Struckmeier
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
3
|
Ji S, Liu H, Pachella L, Stephenson RD, Groisberg R, Weiss SA. Use of immune checkpoint inhibitors in solid organ transplant recipients with advanced cutaneous malignancies. FRONTIERS IN TRANSPLANTATION 2023; 2:1284740. [PMID: 38993910 PMCID: PMC11235332 DOI: 10.3389/frtra.2023.1284740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 10/13/2023] [Indexed: 07/13/2024]
Abstract
Background Immune checkpoint inhibitors (ICI) are standard of care therapy for patients with cutaneous malignancies, the most frequently diagnosed cancers in solid organ transplant (SOT) recipients. The activity and rate of allograft rejection in SOT recipients with advanced skin cancers treated with ICI is understudied. Methods We conducted a retrospective analysis of SOT recipients with advanced melanoma, cutaneous squamous cell carcinoma (cSCC), and merkel cell carcinoma (MCC) who were treated with ICI. Unpublished cases from our institution and published cases from the literature were aggregated. Demographics, type of immunosuppressive therapy, type of ICI(s) administered, prior systemic therapies, tumor response to ICI, and evidence of organ rejection and/or failure were recorded. Objective response rates (ORR) and rates of graft rejection and failure are reported. Results Ninety patients were identified; four patients from our institution and 86 unique patients from a literature review. ORR to first-line ICI for the entire cohort was 41.1% (37/90). ORR by tumor type was 31% (18/58), 64.3% (18/28), and 25.0% (1/4) for melanoma, cSCC, and MCC, respectively. The rate of graft rejection was 37.8% (34/90) with 61.8% (21/34) of these cases progressing to graft failure. Number of immunosuppressive agents (0, 1, 2, or 3) was inversely associated with rate of graft failure. Conclusions In this retrospective analysis, ICIs demonstrate clinical activity in SOT recipients with cutaneous malignancies; however, the rate of graft rejection is high. Treatment plans should be individualized through thorough interdisciplinary discussion. Immunosuppressive modifications may be considered prior to starting treatment, but when feasible, enrollment on clinical trials is preferred.
Collapse
Affiliation(s)
- Stephanie Ji
- Department of Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Hao Liu
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, New Brunswick, NJ, United States
| | - Laura Pachella
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Ryan D Stephenson
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Roman Groisberg
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| | - Sarah A Weiss
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, United States
| |
Collapse
|
4
|
Immune Checkpoint Inhibitor Myocarditis and Cellular Rejection in Orthotopic Heart Transplant Recipients. JACC CardioOncol 2022; 4:717-721. [PMID: 36636444 PMCID: PMC9830197 DOI: 10.1016/j.jaccao.2022.07.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/11/2022] [Indexed: 12/24/2022] Open
Key Words
- ACR, acute cellular rejection
- CNI, calcineurin inhibitor
- CTLA-4, cytotoxic T-lymphocyte-associated antigen-4
- EMB, endomyocardial biopsy
- ICI, immune checkpoint inhibitor
- ISHLT, International Society for Heart and Lung Transplantation
- LVEF, left ventricular ejection fraction
- NT-proBNP, N-terminal pro–B-type natriuretic peptide
- PD-1, programmed cell death protein-1
- PET, positron emission tomography
- SOT, solid organ transplant
- TTE, transthoracic echocardiogram
- heart failure
- immunotherapy
- mTOR, mammalian target of rapamycin
- myocarditis
Collapse
|
5
|
Miao X, Wu Z, Jiang Y, Liu H, Gong W. An efficient combination immunotherapy for antitumor immunity without accelerating cardiac allograft rejection. Immunology 2022; 169:157-166. [PMID: 36517459 DOI: 10.1111/imm.13618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy with immune checkpoint inhibitors (ICIs), including antibodies against programmed cell death protein-1 (PD-1) and its receptor programmed cell death ligand-1 (PD-L1), represents a promising systematic treatment for advanced human malignancies. Transplantation remains the ultimate therapy for end-stage organ diseases. However, the efficacy of ICI treatment in solid organ transplant (SOT) recipients remains controversial. We established a transgenic primary liver cancer mouse model and performed allogeneic heterotopic heart transplantation. Different treatments were performed and survival curves were calculated. Graft samples were collected, and immune cells and the cell surface expression of PD-L1 were analysed by flow cytometry. Inflammatory cytokine levels in the serum were measured by an inflammatory array. The specificity of the histochemical techniques was tested by staining sections. A combination immunotherapy comprising a BET protein inhibitor (JQ1) and an immune checkpoint inhibitor (anti-PD-L1 antibody) was administered to primary liver cancer model mice bearing cardiac allografts. Interestingly, the combination immunotherapy effectively suppressed the progression of primary liver cancer but did not accelerate allograft rejection. In accordance with our previous findings, BET protein inhibition enhances the expression of a putative membrane transporter (Rab8A), which upregulates the expression of PD-L1 on the plasma membrane in a transgenic primary liver cancer mouse model. This may be a crucial mechanism of tumour progression arrest. Our data showed that heart transplantation upregulated the expression of the proinflammatory factor IFN-γ and suggested that BET protein inhibition (with JQ1) decreased PD-L1 expression in heart tissues after cardiac transplantation. This phenomenon was accompanied by enhanced infiltration of inflammatory IFN-γ. Our study provides a novel and efficient therapeutic strategy for SOT recipients.
Collapse
Affiliation(s)
- Xiaolong Miao
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Zelai Wu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Yuancong Jiang
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Han Liu
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China
| | - Weihua Gong
- Department of Surgery, Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China
| |
Collapse
|
6
|
Immune checkpoint blockade for organ-transplant recipients with cancer: A review. Eur J Cancer 2022; 175:326-335. [DOI: 10.1016/j.ejca.2022.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022]
|
7
|
Moshirfar M, Basharat NF, Seitz TS, Ply BK, Ronquillo YC, Hoopes PC. Corneal Transplant Rejections in Patients Receiving Immune Checkpoint Inhibitors. J Clin Med 2022; 11:jcm11195647. [PMID: 36233514 PMCID: PMC9572806 DOI: 10.3390/jcm11195647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are antibodies that target and block immune checkpoints. These biologics were initially approved by the United States Food and Drug Administration (US FDA) in 2011 for the management of melanoma. Since then, the use of ICI therapy has increased, with many new medications on the market that treat approximately 50 types of cancers. Patients receiving this therapy are at an increased risk for transplant rejection, including corneal rejection. Ophthalmologists must be aware of individuals receiving ICI therapy as it may be a relative contraindication for patients with a history of corneal transplantation. Patients on ICIs may also experience ocular side effects, including uveitis, dry eye, and inflammation, while on checkpoint inhibitor therapy. This commentary discusses the current understanding of immune checkpoint inhibitors, their mechanism of action, their ocular side effects, and their role in corneal transplant rejection.
Collapse
Affiliation(s)
- Majid Moshirfar
- Hoopes Vision Research Center, Hoopes Vision, 11820 S. State St. #200, Draper, UT 84020, USA
- John A. Moran Eye Center, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Utah Lions Eye Bank, Murray, UT 84107, USA
- Correspondence: ; Tel.: +1-801-568-0200
| | - Noor F. Basharat
- University of Arizona College of Medicine—Phoenix, Phoenix, AZ 85004, USA
| | - Tanner S. Seitz
- Midwestern University Arizona College of Osteopathic Medicine, Glendale, AZ 85308, USA
| | - Briana K. Ply
- Hoopes Vision Research Center, Hoopes Vision, 11820 S. State St. #200, Draper, UT 84020, USA
| | - Yasmyne C. Ronquillo
- Hoopes Vision Research Center, Hoopes Vision, 11820 S. State St. #200, Draper, UT 84020, USA
| | - Phillip C. Hoopes
- Hoopes Vision Research Center, Hoopes Vision, 11820 S. State St. #200, Draper, UT 84020, USA
| |
Collapse
|
8
|
Hanania HL, Lewis DJ. Systematic Review of Programmed Cell Death-1 Inhibitor Therapy for Advanced-Stage Cutaneous Squamous Cell Carcinoma in Solid-organ Transplant Recipients. J DERMATOL TREAT 2022; 33:3119-3126. [PMID: 36018250 DOI: 10.1080/09546634.2022.2118516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2022]
Abstract
BACKGROUND Programmed cell death-1 (PD-1) inhibitors represent an effective treatment option for advanced cutaneous squamous cell carcinoma (cSCC). However, solid organ transplant (SOT) recipients with cSCC have traditionally been excluded from clinical trials. OBJECTIVE To assess the safety and efficacy of PD-1 inhibitors for stage III-IV cSCC in SOT recipients. MATERIALS & METHODS A systematic review was performed using the PubMed, EMBASE, and Scopus databases. RESULTS We identified 21 articles describing 33 SOT recipients (26 kidney, four liver, two lung, and one heart) with stage III-IV cSCC treated with PD-1 inhibitors. Eleven patients (33.3%) experienced allograft rejection. Of the 25 cases with iRECIST scores, twelve patients (48.0%) had a complete response (CR), eight (32.0%) showed a partial response (PR), three (12.0%) progressive disease, and two (8.0%) stable disease (SD). Including patients without available iRECIST scores, 21 patients (63.6%) showed tumor response. Eleven patients died, with six (50.0%) due to tumor progression and one (11.1%) due to allograft rejection after foregoing dialysis. CONCLUSION PD-1 inhibitors demonstrate efficacy for advanced cSCC and confer a risk of allograft rejection in SOT recipients, requiring careful assessment of risks and benefits. If anti-PD-1 therapy is pursued, use of mTOR inhibitors, prophylactic steroids, and donor-derived cell-free DNA monitoring may mitigate the risk of rejection.
Collapse
Affiliation(s)
- Hannah L Hanania
- Baylor College of Medicine, School of Medicine, Houston, TX, USA
| | - Daniel J Lewis
- Perelman School of Medicine at the University of Pennsylvania, Department of Dermatology, Philadelphia, PA, USA
| |
Collapse
|
9
|
Zelin E, Maronese CA, Dri A, Toffoli L, Di Meo N, Nazzaro G, Zalaudek I. Identifying Candidates for Immunotherapy among Patients with Non-Melanoma Skin Cancer: A Review of the Potential Predictors of Response. J Clin Med 2022; 11:3364. [PMID: 35743435 PMCID: PMC9225110 DOI: 10.3390/jcm11123364] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/02/2022] [Accepted: 06/09/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Non-melanoma skin cancer (NMSC) stands as an umbrella term for common cutaneous malignancies, including basal cell carcinoma (BCC) and cutaneous squamous cell carcinoma (cSCC), together with rarer cutaneous cancers, such as Merkel cell carcinoma (MCC) and other forms of adnexal cancers. The majority of NMSCs can be successfully treated with surgery or radiotherapy, but advanced and metastatic stages may require systemic approaches such as immunotherapy with immune checkpoint inhibitors (ICIs). SUMMARY Since immunotherapy is not effective in all patients and can potentially lead to severe adverse effects, an important clinical question is how to properly identify those who could be suitable candidates for this therapeutic choice. In this paper, we review the potential features and biomarkers used to predict the outcome of ICIs therapy for NMSCs. Moreover, we analyze the role of immunotherapy in special populations, such as the elderly, immunocompromised patients, organ transplant recipients, and subjects suffering from autoimmune conditions. KEY MESSAGES Many clinical, serum, histopathological, and genetic features have been investigated as potential predictors of response in NMSCs treated with ICIs. Although this field of research is very promising, definitive, cost-effective, and reproducible biomarkers are still lacking and further efforts are needed to validate the suggested predictors in larger cohorts.
Collapse
Affiliation(s)
- Enrico Zelin
- Dermatology Clinic, Maggiore Hospital, University of Trieste, 34125 Trieste, Italy; (E.Z.); (L.T.); (N.D.M.); (I.Z.)
| | - Carlo Alberto Maronese
- Dermatology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milan, Italy
| | - Arianna Dri
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy;
- Department of Medical Oncology, Azienda Sanitaria Friuli Centrale (ASUFC), 33100 Udine, Italy
| | - Ludovica Toffoli
- Dermatology Clinic, Maggiore Hospital, University of Trieste, 34125 Trieste, Italy; (E.Z.); (L.T.); (N.D.M.); (I.Z.)
| | - Nicola Di Meo
- Dermatology Clinic, Maggiore Hospital, University of Trieste, 34125 Trieste, Italy; (E.Z.); (L.T.); (N.D.M.); (I.Z.)
| | - Gianluca Nazzaro
- Dermatology Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Iris Zalaudek
- Dermatology Clinic, Maggiore Hospital, University of Trieste, 34125 Trieste, Italy; (E.Z.); (L.T.); (N.D.M.); (I.Z.)
| |
Collapse
|
10
|
Kawashima S, Joachim K, Abdelrahim M, Abudayyeh A, Jhaveri KD, Murakami N. Immune checkpoint inhibitors for solid organ transplant recipients: clinical updates. KOREAN JOURNAL OF TRANSPLANTATION 2022; 36:82-98. [PMID: 35919193 PMCID: PMC9296977 DOI: 10.4285/kjt.22.0013] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 11/22/2022] Open
Abstract
Transplant care continues to advance with increasing clinical experience and improvements in immunosuppressive therapy. As the population ages and long-term survival improves, transplant patient care has become more complex due to comorbidities, frailty, and the increased prevalence of cancer posttransplantation. Immune checkpoint inhibitors (ICIs) have become a standard treatment option for many cancers in non-transplant patients, but the use of ICIs in transplant patients is challenging due to the possibility of disrupting immune tolerance. However, over the past few years, ICIs have gradually started to be used in transplant patients as well. In this study, we review the current use of ICIs after all solid organ transplantation procedures (kidney, liver, heart, and lung). Increasing data suggest that the type and number of immunosuppressants may affect the risk of rejection after immunotherapy. Immunotherapy for cancer in transplant patients may be a feasible option for selected patients; however, prospective trials in specific organ transplant recipients are needed.
Collapse
Affiliation(s)
- Shun Kawashima
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Kole Joachim
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Maen Abdelrahim
- Section of GI Oncology, Department of Medical Oncology, Houston Methodist Cancer Center, Houston, TX, USA
| | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenar D. Jhaveri
- Division of Kidney Diseases and Hypertension, Donald and Barbara Zucker School of Medicine at Hofstra/ Northwell, Great Neck, NY, USA
- Department of Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, USA
| | - Naoka Murakami
- Transplantation Research Center, Division of Renal Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Nativi-Nicolau J, Stehlik J, Kelkhoff AJ, Khong B, Truax CM, Revelo MP, Gilbert EM, Drakos S, Wever-Pinzon O, Fang J, Catino A, Khong HT. Fatal Allograft Rejection and Cardiac Allograft Vasculopathy After Treatment With Pembrolizumab for Metastatic Melanoma in a Heart Transplant Recipient: A Case Report. Transplant Proc 2022; 54:193-196. [PMID: 35012763 DOI: 10.1016/j.transproceed.2021.09.069] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 09/14/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Checkpoint inhibitors decrease the progression of many cancers. However, the experience in immunosuppressed patients is limited, with reports of possible serious adverse events. We present a heart transplant recipient treated with pembrolizumab for metastatic melanoma who developed fatal rejection. The patient was a 29 year-old man who underwent heart transplantation at the age of 10 years for congenital heart disease. Seventeen years after transplant, he was diagnosed with scalp melanoma pT3a, N2a, M0, Stage IIIA, positive for BRAF V600E mutation treated with excision, which metastasized to his lungs and brain a year later. Dabrafenib and trametinib were started with transient response. Additional options and their risks were discussed, and pembrolizumab was started 4 months later due to the incomplete response to previous therapy. Five days after initiation the patient presented with moderate cellular rejection and possible antibody mediated rejection (ISHLT Grade 2R, pAMR 1H). Pembrolizumab was discontinued, and he was treated with steroids. Seven months later he presented in cardiogenic shock and severe coronary allograft vasculopathy. Biopsy was negative for cellular rejection, but suspicious for antibody mediated rejection (ISHLT Grade 0R, pAMR 1H), and he had a new serum alloantibody. Despite steroids and plasmapheresis he remained in refractory cardiogenic shock and died of cardiac arrest.
Collapse
Affiliation(s)
| | | | | | - Brian Khong
- Adventist Health White Memorial, Los Angeles, California
| | | | | | | | | | | | - James Fang
- University of Utah Health, Salt Lake City, Utah
| | - Anna Catino
- University of Utah Health, Salt Lake City, Utah
| | | |
Collapse
|
12
|
O'Connell KA, Schmults CD. Treatment of metastatic cutaneous squamous cell carcinoma in a solid organ transplant recipient with programmed death-1 checkpoint inhibitor therapy. J Eur Acad Dermatol Venereol 2021; 36 Suppl 1:45-48. [PMID: 34855241 DOI: 10.1111/jdv.17407] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/21/2021] [Indexed: 12/20/2022]
Abstract
Limited data exist on the use of immune checkpoint inhibitors (ICI) for the treatment of metastatic cutaneous squamous cell carcinoma (CSCC) in solid organ transplant recipients (SOTR). We report a case of a SOTR who developed metastatic disease following multiple surgeries, three cycles of adjuvant radiotherapy, and minimization of immunosuppression. He was subsequently treated with pembrolizumab and achieved a complete response. However, the patient developed ICI-induced allograft rejection requiring therapy discontinuation. The allograft was salvaged following IVIg and steroids. The patient developed recurrent disease which failed rechallenge with pembrolizumab but achieved a partial response following cemiplimab administration. This case illustrates the potential to treat metastatic CSCC in a SOTR with anti-programmed death-1 therapy and preserve graft function despite allograft rejection.
Collapse
Affiliation(s)
- K A O'Connell
- Brigham & Women's/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Eastern Virginia Medical School, School of Medicine, Norfolk, VA, USA
| | - C D Schmults
- Brigham & Women's/Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
13
|
The Correlation Between Immunohistochemistry Findings and Metastasis in Squamous Cell Carcinoma: A Review. Dermatol Surg 2021; 47:313-318. [PMID: 33165065 DOI: 10.1097/dss.0000000000002850] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND Cutaneous squamous cell carcinoma (SCC) is the second most common type of skin cancer. Only 2% to 5% of SCCs metastasize; however, those do carry a poor prognosis. Immunohistochemistry (IHC) is widely used by pathologists to characterize skin cancers and provide clinically useful information. OBJECTIVE To evaluate the potential prognostic associations between IHC findings and metastasis in SCC. METHODS Searches were conducted in MEDLINE via PubMed for articles published between 1999 and 2019. Search criteria included key words "immunohistochemistry" and "cutaneous squamous cell carcinoma." Six hundred and fifty-three articles were returned and screened, which ultimately left 31 for inclusion in our manuscript. RESULTS Thirty-one articles analyzed in this review included a discussion of the expression of a particular IHC marker and the associated risk of metastasis and/or clinical utility of IHC markers in SCC, especially metastatic SCC. Markers that had several or more studies supporting clinical utility were E-cadherin, podoplanin, CD8+ T cells, PD-L1, epidermal growth factor receptor, and Cyclin D1. CONCLUSION Immunohistochemistry profiling of SCC may be useful in select cases when providing a prognosis remains challenging and in identification of potential therapeutic targets for high-risk or metastatic tumors.
Collapse
|
14
|
Abstract
PURPOSE To report the first case of corneal graft rejection presumably associated with pembrolizumab immunotherapy. METHODS Case report and literature review. RESULTS An asymptomatic 85-year-old woman with a history of bilateral penetrating keratoplasty presented for a follow-up visit with bilateral diffuse keratic precipitates and subepithelial infiltrates. There were no anterior chamber cells. Bilateral subclinical corneal graft rejection was suspected. Three months previously, pembrolizumab immunotherapy was started for a metastatic urothelial cell tumor. Corneal graft rejection was managed with topical dexamethasone drops, which were tapered slowly. Pembrolizumab treatment was continued with careful ophthalmological follow-up. Unfortunately, recurrence of corneal graft rejection was observed 8 weeks after cessation of topical dexamethasone drops. After consulting the treating oncologist, pembrolizumab treatment was stopped to prevent recurrent corneal graft rejection. CONCLUSIONS We report the first case of corneal graft rejection presumably associated with pembrolizumab immunotherapy. Corneal graft rejection may be successfully managed with corticosteroid therapy. However, constant vigilance and follow-up are advised because of the risk of recurrence in case of continued pembrolizumab treatment. Given the subclinical presentation, baseline ophthalmological screening is advised in all corneal graft patients after initiating immune checkpoint inhibitor therapy.
Collapse
|
15
|
Miao K, Zhang L. Application of Immune Checkpoint Inhibitors in Solid Organ Transplantation Recipients: A Systematic Review. Interdiscip Sci 2021; 13:801-814. [PMID: 34152556 DOI: 10.1007/s12539-021-00437-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 04/29/2021] [Accepted: 05/06/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND Solid organ transplantation (SOT) is a treatment method for end-stage organ diseases and improve their life quality, while using long-term immunosuppressant drugs (ISD) is needed to suppress the function of the immune system. Immune checkpoint inhibitors (ICIs) are a class of anti-tumor drugs that kill tumors by activating the autoimmune system. The primary objective of our systematic review is to investigate the risk factors for organ rejection and the efficacy of ICIs in solid organ transplantation recipients (SOTRs). METHODS We searched four databases to find relevant articles up to January 2021. A total of 61 articles involving 106 SOTRs met the screening criteria and were included in our systematic review. The collected data were statistical described, and the risk factors were analyzed by logistic regression. RESULTS Forty-four patients (41.5%) developed host-versus-graft response (HVGR) after ICIs. mTOR inhibitors (pre-ICIs) (p = 0.069, OR = 0.377, 95% CI 0.132-1.078) and calcineurin inhibitors (post-ICIs) (p = 0.056, OR = 0.375, 95%CI 0.137-1.025) may help reduce the incidence of HVGR. Hormones (pre-ICIs) (p = 0.026, OR = 3.150, 95%CI 1.150-8.628) and anti-metabolites (pre-ICIs) (p = 0.022, OR = 3.214, 95%CI 1.185-8.720) may adversely affect the efficacy of ICIs. Only 35.6% of patients both responded well to ICIs treatment and did not develop HVGR. CONCLUSIONS Our systematic review summarizes the use of ICIs in SOTRs and evaluates the efficacy of ICIs and the risk factors that induce HVGR. Through risk factor analysis, we found that mTOR inhibitors and calcineurin inhibitors may help to reduce the occurrence of HVGR; hormones and anti-metabolic drugs may have adverse effects on the efficacy of ICIs. In addition, there is a contradictory relationship between the occurrence of HVGR and the efficacy of ICIs.
Collapse
Affiliation(s)
- Kang Miao
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 53 Dongdan North Avenue, Dongcheng District, Beijing, China
| | - Li Zhang
- Department of Pulmonary and Critical Care Medicine, Peking Union Medical Hospital, Chinese Academy of Medical Science and Peking Union Medical College, 53 Dongdan North Avenue, Dongcheng District, Beijing, China.
| |
Collapse
|
16
|
Hirotsu KE, Hua V, Tran AT, Morris L, Reddy SA, Kwong BY, Zaba LC. Complete remission from intralesional talimogene laherparepvec for regionally advanced Merkel cell carcinoma in an immunocompromised solid organ transplant patient. JAAD Case Rep 2021; 13:144-146. [PMID: 34195326 PMCID: PMC8226392 DOI: 10.1016/j.jdcr.2021.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Kelsey E Hirotsu
- Department of Dermatology, Stanford University School of Medicine, Stanford, California
| | - Vivian Hua
- Department of Dermatology, Stanford University School of Medicine, Stanford, California
| | - Anhthy T Tran
- Department of Dermatology, Stanford University School of Medicine, Stanford, California
| | - Laura Morris
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Sunil A Reddy
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Bernice Y Kwong
- Department of Dermatology, Stanford University School of Medicine, Stanford, California
| | - Lisa C Zaba
- Department of Dermatology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
17
|
Dang N, Waer M, Sprangers B, Lin Y. Intratumoral immunotherapy with anti-PD-1 and TLR9 agonist induces systemic antitumor immunity without accelerating rejection of cardiac allografts. Am J Transplant 2021; 21:60-72. [PMID: 32506732 DOI: 10.1111/ajt.16105] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/21/2020] [Accepted: 05/23/2020] [Indexed: 01/25/2023]
Abstract
Immune checkpoint inhibitors, such as programmed cell death 1 (PD-1) blockades, have revolutionized the field of cancer immunotherapy. However, there is a growing concern whether PD-1 inhibitors can be administered safely to transplant recipients with advanced cancer, as the T cells activated by checkpoint inhibitors may become reactive not only toward tumor antigens but also toward donor alloantigen, thereby resulting in allograft rejection. Here, immunotherapy with anti-PD-1/toll like receptor 9 agonist was administered to C57BL/6 mice bearing a cardiac allograft that were receiving maintenance immunosuppression or a PI4KIIIβ inhibitor-based tolerogenic regimen. Intratumoral (i.t.), but not systemic, immunotherapy promoted potent anti-tumor responses, but did not accelerate allograft rejection. This effect was associated with a pro-immunogenic effect induced by i.t. immunotherapy resulting in systemic cellular and humoral immune anti-tumor responses. Furthermore, when the tumor and cardiac allograft shared major histocompatibility complex (MHC) antigens, i.t. immunotherapy promoted immune responses directed against tumor and the cardiac allograft resulting in allograft rejection. The anti-tumor effect was compromised by maintenance immunosuppression with cyclosporin A, indicating that an optimal balance between enhanced anti-tumor immunity and decreased transplant immunoreactivity is critical. A clinically relevant approach could be to temporarily withdraw maintenance immunosuppression and/or replace it with a PI4KIIIβ inhibitor-based tolerance-inducing regimen to allow for effective immunotherapy to take place.
Collapse
Affiliation(s)
- Nana Dang
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Mark Waer
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Ben Sprangers
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium.,Department of Nephrology, University Hospitals Leuven, Leuven, Belgium
| | - Yuan Lin
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Delyon J, Zuber J, Dorent R, Poujol-Robert A, Peraldi MN, Anglicheau D, Lebbe C. Immune Checkpoint Inhibitors in Transplantation-A Case Series and Comprehensive Review of Current Knowledge. Transplantation 2021; 105:67-78. [PMID: 32355121 DOI: 10.1097/tp.0000000000003292] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Cancer is a leading cause of morbidity and deaths in solid organ transplant recipients. In immunocompetent patients, cancer prognosis has been dramatically improved with the development of immune checkpoint inhibitors (ICI), as programmed cell death protein 1/programmed death-ligand 1 and cytotoxic T lymphocyte-associated antigen 4 inhibitors, that increase antitumor immune responses. ICI has been developed outside of the scope of transplantation because of the theoretical risk of graft rejection, which has later been confirmed by the publication of several cases and small series. The use of ICI became unavoidable for treating advanced cancers including in organ transplant patients, but their management in this setting remains highly challenging, as to date no strategy to adapt the immunosuppression and to prevent graft rejection has been defined. In this article, we report a monocentric series of 5 solid organ transplant recipients treated with ICI and provide a comprehensive review of current knowledge of ICI management in the setting of solid organ transplantation. Strategies warranted to increase knowledge through collecting more exhaustive data are also discussed.
Collapse
Affiliation(s)
- Julie Delyon
- Department of Dermatology, AP-HP Hôpital Saint Louis, Université de Paris, INSERM U976, Team 1, HIPI, Paris, France
| | - Julien Zuber
- Department of Nephrology and Kidney Transplantation, University Hospital Center (CHU) Necker, Université de Paris, Paris, France
| | - Richard Dorent
- Department of Cardiac Surgery, AP-HP, Bichat-Claude Bernard University Hospital, Paris, France
| | - Armelle Poujol-Robert
- Department of Hepatology, AP-HP, Hôpital Saint-Antoine, UPMC University, Paris, France
| | - Marie-Noelle Peraldi
- Department of Nephrology, AP-HP Hôpital Saint Louis, Université de Paris, Paris, France
| | - Dany Anglicheau
- Department of Nephrology and Kidney Transplantation, University Hospital Center (CHU) Necker, Université de Paris, Paris, France
| | - Celeste Lebbe
- Department of Dermatology, AP-HP Hôpital Saint Louis, Université de Paris, INSERM U976, Team 1, HIPI, Paris, France
| |
Collapse
|
19
|
Malviya N, Tattersall IW, Leventhal J, Alloo A. Cutaneous immune-related adverse events to checkpoint inhibitors. Clin Dermatol 2020; 38:660-678. [PMID: 33341200 DOI: 10.1016/j.clindermatol.2020.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The development of immunotherapy has led to a paradigm shift in the treatment of both solid and hematologic malignancies. As immunomodulatory therapies are employed with increasing frequency, a greater number of immune-related adverse reactions are being reported, and the majority of these involve the skin. As a result, dermatologists are increasingly becoming involved in the management of these cutaneous adverse reactions-often providing critical recommendations regarding ongoing cancer treatment. Cutaneous immune-related adverse reactions can vary significantly from patient to patient, making early recognition and timely intervention imperative to mitigate associated morbidity and potential treatment interruption. Although there is considerable overlap in the cutaneous adverse events caused by these immune checkpoint inhibitors, specific eruptions are characteristically associated with particular checkpoint inhibitors. In addition, a patient's comorbidities or immune status can play a significant role in the presentation and management of such adverse reactions. This review characterizes and provides management guidelines for the various cutaneous toxicities associated with checkpoint inhibitor therapy, including CTLA-4 inhibitors, PD-1 inhibitors, and PD-L1 inhibitors. © 2020 Elsevier Inc. All rights reserved.
Collapse
Affiliation(s)
- Neeta Malviya
- Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York, USA
| | - Ian W Tattersall
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jonathan Leventhal
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Allireza Alloo
- Department of Dermatology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New Hyde Park, New York, USA.
| |
Collapse
|
20
|
Fisher J, Zeitouni N, Fan W, Samie FH. Immune checkpoint inhibitor therapy in solid organ transplant recipients: A patient-centered systematic review. J Am Acad Dermatol 2020; 82:1490-1500. [DOI: 10.1016/j.jaad.2019.07.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 06/12/2019] [Accepted: 07/03/2019] [Indexed: 02/06/2023]
|
21
|
Qiu J, Tang W, Du C. Immune Checkpoint Inhibitors in Patients with Recurrent Hepatocellular Carcinoma after Liver Transplantation: A Case Report and Literature Review. Curr Cancer Drug Targets 2020; 20:720-727. [PMID: 32433005 DOI: 10.2174/1568009620666200520084415] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 04/21/2020] [Accepted: 04/26/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Immune checkpoint modulators, such as the programmed death protein-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibitor, cytotoxic T-Lymphocyte-associated antigen 4 (CTLA-4) inhibitor have been investigated with encouraging results for hepatocellular carcinoma (HCC). However, the safety of this strategy in patients with previous liver transplantation (LT) is not well studied. OBJECTIVE To explore the safety and feasibility of immune checkpoints inhibitors in recurrent and metastatic HCC patients on a background of LT. METHODS A case of recurrent, refractory, metastatic HCC after LT, where PD-1 inhibitor was initiated, was described and related literature was reviewed. RESULTS There was complete remission in lung metastases and the partial radiological response of metastatic retroperitoneal lymph node to the drug with no liver graft rejection after 13 cycles of PD- 1 inhibitor injection. PD-1inhibitor, at least in this patient, was verified to play an important role in controlling tumor progression and prolonging patient survival. CONCLUSION This novel drug might be a useful method to allow doctors to guarantee a better chance for long-term survival in recurrent, metastatic HCC patients with the previous LT. However, it should be used with caution in allograft recipients due to the risk of acute graft rejection, further larger, prospective studies are needed to determine optimal immunomodulatory therapy to achieve optimal anti-tumor efficacy with transplant liver preservation.
Collapse
Affiliation(s)
- Jianguo Qiu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Wei Tang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Chengyou Du
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
22
|
Grabie N, Lichtman AH, Padera R. T cell checkpoint regulators in the heart. Cardiovasc Res 2020; 115:869-877. [PMID: 30721928 DOI: 10.1093/cvr/cvz025] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 01/07/2019] [Accepted: 02/04/2019] [Indexed: 12/27/2022] Open
Abstract
T lymphocyte-mediated immune responses in the heart are potentially dangerous because they can interfere with the electromechanical function. Furthermore, the myocardium has limited regenerative capacity to repair damage caused by effector T cells. Myocardial T cell responses are normally suppressed by multiple mechanisms of central and peripheral tolerance. T cell inhibitory molecules, so called immune checkpoints, limit the activation and effector function of heart antigen-reactive T cells that escape deletion during development in the thymus. Programmed cell protein death-1 (PD-1) and cytotoxic T-lymphocyte-associated protein-4 (CTLA-4) are checkpoint molecules homologous to the costimulatory receptor CD28, and they work to block activating signals from the T cell antigen receptor and CD28. Nonetheless, PD-1 and CTLA-4 function in different ways and at different steps in a T cell response to antigen. Studies in mice have established that genetic deficiencies of checkpoint molecules, including PD-1, PD-L1, CTLA-4, and lymphocyte activation gene-3, result in enhanced risk of autoimmune T cell-mediated myocarditis and increased pathogenicity of heart antigen-specific effector T cells. The PD-1/PD-L1 pathway appears to be particularly important in cardiac protection from T cells. PD-L1 is markedly up-regulated on myocardial cells by interferon-gamma secreted by T cells and PD-1 or PD-L1 deficiency synergizes with other defects in immune regulation in promoting myocarditis. Consistent with these studies, myocarditis has emerged as a serious adverse reaction of cancer therapies that target checkpoint molecules to enhance anti-tumour T cell responses. Histopathology and immunohistochemical analyses of myocardial tissue from immune checkpoint blockade (ICB)-treated patients echoes findings in checkpoint-deficient mice. Many questions about myocarditis in the setting of cancer immunotherapy still need to be answered, including the nature of the target antigens, genetic risk factors, and variations in the disease with combined therapies. Addressing these questions will require further immunological analyses of blood and heart tissue from patients treated with ICB.
Collapse
Affiliation(s)
- Nir Grabie
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, NRB Room 752N, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, NRB Room 752N, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Robert Padera
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, NRB Room 752N, 77 Avenue Louis Pasteur, Boston, MA, USA
| |
Collapse
|
23
|
Recent and Emerging Therapies for Cutaneous Squamous Cell Carcinomas of the Head and Neck. Curr Treat Options Oncol 2020; 21:37. [PMID: 32328817 DOI: 10.1007/s11864-020-00739-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OPINION STATEMENT Cutaneous squamous cell carcinoma (cSCC) of the head and neck is typically managed with Mohs Micrographic Surgery (MMS) for cosmetic reasons. MMS also improves oncologic outcomes for high-risk tumors. Patients with certain high-risk subsets of the disease also benefit from adjuvant radiation therapy. The PD-1 inhibitor, cemiplimab, was recently approved for treatment of locally advanced and metastatic cSCC unamenable to curative surgery or radiation therapy after the drug demonstrated encouraging, durable response rates. Cemiplimab and other systemic immunotherapies are now being evaluated in clinical trials in the neoadjuvant and adjuvant settings as well. Localized immunotherapies are also being studied, including oncolytic viruses such as talimogene laherparepvec, a modified herpes simplex virus previously approved for the treatment of advanced cutaneous melanoma. Most importantly, multidisciplinary care is crucial in optimizing outcomes for patients with high-risk cSCC of the head and neck.
Collapse
|
24
|
Bibee K, Swartz A, Sridharan S, Kurten CHL, Wessel CB, Skinner H, Zandberg DP. Cutaneous squamous cell carcinoma in the organ transplant recipient. Oral Oncol 2020; 103:104562. [PMID: 32065978 PMCID: PMC7217490 DOI: 10.1016/j.oraloncology.2019.104562] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/13/2019] [Accepted: 12/31/2019] [Indexed: 12/31/2022]
Abstract
One in twenty solid organ transplant recipients (SOTRs) will develop a highly morbid or fatal cutaneous carcinoma after transplantation. The majority of these cases develop on the head and neck and may require intervention on the part of dermatology, dermatologic surgery, otolaryngology, transplant medicine, radiation oncology, and medical oncology. In this review, we discuss the problem of cutaneous squamous cell carcinoma (cSCC) in SOTRs as well as the prognostic factors and management strategies to care for this population.
Collapse
Affiliation(s)
- Kristin Bibee
- Department of Dermatology, University of Pittsburgh, 3708 Fifth Ave #5, Pittsburgh, PA 15213, USA; Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA 15232, USA.
| | - Andrew Swartz
- Department of Medicine, University of Pittsburgh, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - Shaum Sridharan
- Department of Otolaryngology, University of Pittsburgh, 203 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Cornelius H L Kurten
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA 15232, USA; Department of Otorhinolaryngology, University Hospital Essen, University Duisburg-Essen, Hufelandstrabe 55, 45147 Essen, Germany
| | - Charles B Wessel
- Health Sciences Library, University of Pittsburgh, 200 Scaife Hall, 3550 Terrace St, Pittsburgh, PA 15261, USA
| | - Heath Skinner
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA 15232, USA; Department of Radiation Oncology, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
| | - Dan P Zandberg
- Hillman Cancer Center, University of Pittsburgh Medical Center, 5115 Centre Ave, Pittsburgh, PA 15232, USA; Department of Medicine, University of Pittsburgh, 3550 Terrace St, Pittsburgh, PA 15261, USA
| |
Collapse
|
25
|
Kumar V, Shinagare AB, Rennke HG, Ghai S, Lorch JH, Ott PA, Rahma OE. The Safety and Efficacy of Checkpoint Inhibitors in Transplant Recipients: A Case Series and Systematic Review of Literature. Oncologist 2020; 25:505-514. [PMID: 32043699 DOI: 10.1634/theoncologist.2019-0659] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 12/09/2019] [Indexed: 02/06/2023] Open
Abstract
Limited data exist on safety and efficacy of immune checkpoint inhibitors (ICIs) among organ transplant recipients. The objective of this study was to report a case series of two patients with renal transplant who received treatment with an ICI and to conduct a pooled analysis of published cases to describe the safety and efficacy of ICIs in organ transplant patients. A systematic search in the Google Scholar and PubMed databases was carried out to include all the published cases of organ transplant patients who received treatment with ICIs including programmed cell death protein 1 (PD-1), programmed death-ligand 1, or cytotoxic lymphocyte antigen-4 inhibitors since their inscription to January 31, 2019. In the present series of two cases with renal allografts who received pembrolizumab, one patient with squamous cell carcinoma of the skin experienced complete response (CR), whereas another patient with melanoma had a mixed response. Both patients experienced allograft rejection, but graft was salvaged. The pooled analysis of 64 patients published in literature showed that overall allograft rejection rate is 41% in organ transplant recipients following ICI therapy. The graft rejection rate was 44% (17/39) for renal, 39% (7/19) for liver, and 20% (1/5) for cardiac allografts. The highest risk was seen among patients who were treated with PD-1 inhibitors, 20/42 (48%)-13/24 (54%) on nivolumab and 7/18 (39%) on pembrolizumab. The risk was lowest with ipilimumab, 23% (3/13). The overall response rate (CR + partial response [PR]) was 20% with ipilimumab, 26% with nivolumab, and 53% with pembrolizumab, whereas disease control rate (CR + PR + stable disease) was 35% with ipilimumab, 37% with nivolumab, and 53% with pembrolizumab. None of the variables including age, gender, type of cancer, type of allograft, type of immunosuppression, time since transplantation to initiation of ICI, and prior history of rejection were significantly associated with the transplant rejection on univariate analysis. The efficacy of ICI among patients with organ transplant appears promising, warranting testing in prospective clinical trials. The risk of rejection and allograft loss is considerable; therefore, the risk and alternative form of therapies should be thoroughly discussed with the transplant patients prior to initiating ICI therapy. IMPLICATIONS FOR PRACTICE: Transplant recipients are at higher risk of developing cancers. Although immune checkpoint inhibitors have been shown to improve the outcome in more than one cancer type, transplant recipients were excluded from these trials. Most of the data on the safety and efficacy of immune checkpoint inhibitors in transplant patients are based upon case series and case reports. The pooled data from these reports suggest that anti-programmed death-ligand 1 inhibitors have reasonable safety and efficacy among organ transplant patients, which warrants testing in clinical trials.
Collapse
Affiliation(s)
- Vivek Kumar
- Department of Medical Oncology, Dana Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Atul B Shinagare
- Department of Radiology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Helmut G Rennke
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sandeep Ghai
- Transplant Nephrology, Boston Medical Center, Boston, Massachusetts, USA
| | - Jochen H Lorch
- Department of Medical Oncology, Dana Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Patrick A Ott
- Department of Medical Oncology, Dana Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Osama E Rahma
- Department of Medical Oncology, Dana Farber Cancer Institute and Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
26
|
Manohar S, Thongprayoon C, Cheungpasitporn W, Markovic SN, Herrmann SM. Systematic Review of the Safety of Immune Checkpoint Inhibitors Among Kidney Transplant Patients. Kidney Int Rep 2020; 5:149-158. [PMID: 32043028 PMCID: PMC7000848 DOI: 10.1016/j.ekir.2019.11.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 11/11/2019] [Accepted: 11/25/2019] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Kidney transplant (Ktx) recipients are excluded from clinical trials of immune checkpoint inhibitors. The aim of this systematic review was to assess the safety of immune checkpoint inhibitors among Ktx patients. METHODS A literature search was conducted using MEDLINE, EMBASE, and Cochrane Database from inception through April 2019. We included studies that reported outcomes of Ktx recipients who received immune checkpoint inhibitors for cancer treatment. Outcomes of interest were allograft rejection and/or allograft failure. RESULTS Twenty-seven articles with a total of 44 Ktx patients treated with immune checkpoint inhibitor were identified. Of 44 Ktx patients, 18 were reported to have acute rejection. Median time from immune checkpoint inhibitors to acute rejection diagnosis was 24 (interquartile range, 10-60) days. Reported types of acute allograft rejection were cellular rejection (33%), mixed cellular and antibody-mediated rejection (17%), and unspecified type (50%). Fifteen (83%) had allograft failure and 8 (44%) died. Three patients had a partial remission (17%), 1 patient achieved cancer response (6%), and 5 patients had stable disease (28%). CONCLUSION The findings of our study raise awareness of the increased risk for acute allograft rejection/failure following immune checkpoint inhibitors for cancer treatment among Ktx patients, in particular with programmed cell death 1 (PD-1) inhibitors. Future large-scale clinical studies are required to appraise the pathogenesis and plan optimal balanced therapy that helps sustain graft tolerance.
Collapse
Affiliation(s)
- Sandhya Manohar
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Charat Thongprayoon
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| | - Wisit Cheungpasitporn
- Division of Nephrology, Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | | | - Sandra M. Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
27
|
Hu B, Yang XB, Sang XT. Liver graft rejection following immune checkpoint inhibitors treatment: a review. Med Oncol 2019; 36:94. [DOI: 10.1007/s12032-019-1316-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 09/09/2019] [Indexed: 02/07/2023]
Abstract
Abstract
Immune checkpoint inhibitors (ICIs) have demonstrated remarkable efficacy in a variety of solid tumors; nonetheless, they have not been well investigated and are still recognized as a relative contraindication for patients with a liver transplantation (LT) history, since ICIs treatment might potentially lead to graft rejection. The program death-1 (PD-1) and the cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) pathways are implicated in the tolerance of transplanted organ, as well as blockade of the pathways, which contribute to eliminating tumors and may inadvertently lead to peripheral transplant rejection. Currently, no guidelines are available regarding the treatment for ICIs patients with a prior LT history. Therefore, this study was carried out to review the recent studies, attempting to introduce the ICIs-related graft rejection after LT from various aspects. We believed that ICIs could be given for the well-informed patients receiving LT and developed recurrence in a controlled setting. Typically, these patients should be treated according to a clinical care path or a prospective clinical trial, so as obtain a persistent anti-tumor immune response in the meantime of avoiding graft rejection, adjust the immunosuppression, reduce the possibility of graft loss following rejection, and have the opportunity to develop biomarkers for tumor response and transplant rejection.
Collapse
|
28
|
Diagnosis of immune checkpoint inhibitor-associated myocarditis: A systematic review. Int J Cardiol 2019; 296:113-121. [PMID: 31327516 DOI: 10.1016/j.ijcard.2019.07.025] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 06/14/2019] [Accepted: 07/08/2019] [Indexed: 01/10/2023]
Abstract
BACKGROUND Myocarditis is a rare but severe adverse event associated with immune checkpoint inhibitors, its diagnosis depending on a high index of suspicion and appropriate investigations. Our objective was to systematically review the diagnostic approaches to myocarditis associated with immune checkpoint inhibitors. METHODS The systematic review was conducted according to the PRISMA guidelines (PROSPERO Registration: CRD42018097247). We searched Medline and Embase for case reports, case series, and observational studies published in journal articles or presented as conference abstracts that describe patients who developed myocarditis after immune checkpoint inhibitor therapy. RESULTS After a review of 2326 citations, we included 88 cases (53 case reports/series published in journal articles and 35 cases in the observational study). Serum troponin was elevated in 98% of the case reports and 94% of participants in the observational study. ST changes including ST elevation were present in almost a third of case reports. Echocardiography revealed preserved left ventricular ejection fraction in 32% of case reports and 51% of cases in the observational study; however, preserved systolic function did not predict greater survival. Patients who suffered poorer prognosis tended to have major conduction defects or ventricular arrhythmias more frequently than patients who did not. Acute myocardial ischemia was ruled out in all cases (n = 31) when the diagnostic workup included coronary angiography. CONCLUSIONS Immune checkpoint inhibitor-associated myocarditis is characterized by elevation of cardiac troponin levels and non-specific electrocardiographic changes. Early coronary angiography may distinguish it from myocardial ischemia or myocardial infarction.
Collapse
|
29
|
Ros J, Matos I, Martin-Liberal J. Immunotherapy in organ-transplanted cancer patients: efficacy and risk of organ rejection. Ann Oncol 2019; 30:1173-1177. [PMID: 30977776 DOI: 10.1093/annonc/mdz129] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- J Ros
- Medical Oncology, Vall d´Hebron University Hospital, Barcelona.
| | - I Matos
- Medical Oncology, Vall d´Hebron University Hospital, Barcelona; Molecular Therapeutics Research Unit (UITM), Vall d'Hebron Institute of Oncology (VHIO), Barcelona
| | - J Martin-Liberal
- Molecular Therapeutics Research Unit (UITM), Vall d'Hebron Institute of Oncology (VHIO), Barcelona; Melanoma, Sarcoma and Genitourinary Tumors Unit, Early Drug Development Unit, Institut Català d'Oncologia (ICO) L'Hospitalet, Barcelona, Spain
| |
Collapse
|
30
|
Abstract
Immune checkpoint inhibitors present clinicians with both an exciting step forward in cancer treatment and the unknown possibilities of an unshackled immune system. The latter phenomena, known as immune-related adverse events (irAEs), are of particular interest because they may affect any organ system with autoimmune-like pathologies, such as hepatitis and colitis. Within the cardiovascular system, irAEs associated with immune checkpoint blockade exist as a broad clinical spectrum, with autoimmune myocarditis being the best-characterized entity at this time. In general, irAEs are often reversible with immunosuppression. However, irAEs that affect the cardiovascular system pose the possibility of a rapid and fatal clinical deterioration. The mortality attributed to immune checkpoint blockade-associated autoimmune myocarditis, as reported in the WHO database, exists from 36% to 67%, dependent on the therapeutic regimen. Yet, despite the potential severity such events pose, guidelines dictating the identification of immune checkpoint inhibition irAEs do not exist, providing a stark contrast with other anticancer medications with known cardiovascular effects. The lack of guidelines may be related to the perceived rarity of these events, yet a recent study of immune checkpoint inhibition-associated autoimmune myocarditis suggests that this clinical entity may be more prevalent than initially believed. Until more standardized information regarding these potentially serious events is available, the study of documented cases is instructive to improve identification of such phenomena, as well as the outcomes for patients who develop them.
Collapse
|
31
|
Bottomley MJ, Thomson J, Harwood C, Leigh I. The Role of the Immune System in Cutaneous Squamous Cell Carcinoma. Int J Mol Sci 2019; 20:E2009. [PMID: 31022866 PMCID: PMC6515307 DOI: 10.3390/ijms20082009] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/16/2019] [Accepted: 04/19/2019] [Indexed: 02/06/2023] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most common skin cancer. In immunosuppressed populations it is a source of considerable morbidity and mortality due to its enhanced recurrence and metastatic potential. In common with many malignancies, leucocyte populations are both protective against cancer development and also play a role in 'sculpting' the nascent tumor, leading to loss of immunogenicity and tumor progression. UV radiation and chronic viral carriage may represent unique risk factors for cSCC development, and the immune system plays a key role in modulating the response to both. In this review, we discuss the lessons learned from animal and ex vivo human studies of the role of individual leucocyte subpopulations in the development of cutaneous SCC. We then discuss the insights into cSCC immunity gleaned from studies in humans, particularly in populations receiving pharmacological immunosuppression such as transplant recipients. Similar insights in other malignancies have led to exciting and novel immune therapies, which are beginning to emerge into the cSCC clinical arena.
Collapse
Affiliation(s)
- Matthew J Bottomley
- Transplantation Research and Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| | - Jason Thomson
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| | - Catherine Harwood
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| | - Irene Leigh
- Centre for Cell Biology and Cutaneous Research, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK.
| |
Collapse
|
32
|
Abdel-Wahab N, Safa H, Abudayyeh A, Johnson DH, Trinh VA, Zobniw CM, Lin H, Wong MK, Abdelrahim M, Gaber AO, Suarez-Almazor ME, Diab A. Checkpoint inhibitor therapy for cancer in solid organ transplantation recipients: an institutional experience and a systematic review of the literature. J Immunother Cancer 2019; 7:106. [PMID: 30992053 PMCID: PMC6469201 DOI: 10.1186/s40425-019-0585-1] [Citation(s) in RCA: 198] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Accepted: 04/01/2019] [Indexed: 02/06/2023] Open
Abstract
Background Checkpoint inhibitors (CPIs) have revolutionized the treatment of cancer, but their use remains limited by off-target inflammatory and immune-related adverse events. Solid organ transplantation (SOT) recipients have been excluded from clinical trials owing to concerns about alloimmunity, organ rejection, and immunosuppressive therapy. Thus, we conducted a retrospective study and literature review to evaluate the safety of CPIs in patients with cancer and prior SOT. Methods Data were collected from the medical records of patients with cancer and prior SOT who received CPIs at The University of Texas MD Anderson Cancer Center from January 1, 2004, through March 31, 2018. Additionally, we systematically reviewed five databases through April 2018 to identify studies reporting CPIs to treat cancer in SOT recipients. We evaluated the safety of CPIs in terms of alloimmunity, immune-related adverse events, and mortality. We also evaluated tumor response to CPIs. Results Thirty-nine patients with allograft transplantation were identified. The median age was 63 years (range 14–79 years), 74% were male, 62% had metastatic melanoma, 77% received anti-PD-1 agents, and 59% had prior renal transplantation, 28% hepatic transplantation, and 13% cardiac transplantation. Median time to CPI initiation after SOT was 9 years (range 0.92–32 years). Allograft rejection occurred in 41% of patients (11/23 renal, 4/11 hepatic, and 1/5 cardiac transplantations), at similar rates for anti-CTLA-4 and anti-PD-1 therapy. The median time to rejection was 21 days (95% confidence interval 19.3–22.8 days). There were no associations between time since SOT and frequency, timing, or type of rejection. Overall, 31% of patients permanently discontinued CPIs because of allograft rejection. Graft loss occurred in 81%, and death was reported in 46%. Of the 12 patients with transplantation biopsies, nine (75%) had acute rejection, and five of these rejections were T cell-mediated. In melanoma patients, 36% responded to CPIs. Conclusions SOT recipients had a high allograft rejection rate that was observed shortly after CPI initiation, with high mortality rates. Further studies are needed to optimize the anticancer treatment approach in these patients. Electronic supplementary material The online version of this article (10.1186/s40425-019-0585-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Noha Abdel-Wahab
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.,Department of Rheumatology and Rehabilitation, Faculty of Medicine, Assiut University Hospitals, Assiut, Egypt
| | - Houssein Safa
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ala Abudayyeh
- Section of Nephrology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel H Johnson
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Van Anh Trinh
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chrystia M Zobniw
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Heather Lin
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael K Wong
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Maria E Suarez-Almazor
- Section of Rheumatology and Clinical Immunology, Department of General Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adi Diab
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
33
|
Bajwa R, Cheema A, Khan T, Amirpour A, Paul A, Chaughtai S, Patel S, Patel T, Bramson J, Gupta V, Levitt M, Asif A, Hossain MA. Adverse Effects of Immune Checkpoint Inhibitors (Programmed Death-1 Inhibitors and Cytotoxic T-Lymphocyte-Associated Protein-4 Inhibitors): Results of a Retrospective Study. J Clin Med Res 2019; 11:225-236. [PMID: 30937112 PMCID: PMC6436564 DOI: 10.14740/jocmr3750] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/13/2019] [Indexed: 12/11/2022] Open
Abstract
In recent years the use of immunomodulating therapy to treat various cancers has been on the rise. Three checkpoint inhibitors have been approved by the Food and Drug Administration (ipilimumab, pembrolizumab and nivolumab). The use of these drugs comes with serious adverse events related to excessive immune activation, collectively known as immune-related adverse events (irAEs). We conducted a system-based review of 139 case reports/case series that have described these adverse events between January 2016 and April 2018, found in the PubMed database. There was a broad spectrum of presentations, doses and checkpoint inhibitors used. The most common check point inhibitor observed in our literature review was nivolumab. The most common adverse effects encountered were colitis (14/139), hepatitis (11/139), adrenocorticotropic hormone insufficiency (12/139), hypothyroidism (7/139), type 1 diabetes (22/139), acute kidney injury (16/139) and myocarditis (10/139). The treatment most commonly consisted of cessation of the immune checkpoint inhibitor, initiation of steroids and supportive therapy. This approach provided a complete resolution in a majority of cases; however, there were many that developed long-term adverse events with deaths reported in a few cases. The endocrine system was the mostly commonly affected with the development of type 1 diabetes mellitus or diabetic ketoacidosis being the most frequently reported adverse events. While immunomodulating therapy is a significant advance in the management of various malignancies, it is capable of serious adverse effects. Because the majority of the cases developed pancreatic dysfunction within five cycles of therapy, in addition to the evaluation of other systems, pancreatic function should be closely monitored to minimize adverse impact on patients.
Collapse
Affiliation(s)
- Ravneet Bajwa
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Anmol Cheema
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Taimoor Khan
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Alireza Amirpour
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Anju Paul
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Saira Chaughtai
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Shrinil Patel
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Tejas Patel
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Joshua Bramson
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Varsha Gupta
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Michael Levitt
- Department of Hematology/Oncology, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Arif Asif
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Mohammad A Hossain
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| |
Collapse
|
34
|
Zehou O, Leibler C, Arnault JP, Sayegh J, Montaudié H, Rémy P, Glotz D, Cordonnier C, Martin L, Lebbé C. Ipilimumab for the treatment of advanced melanoma in six kidney transplant patients. Am J Transplant 2018; 18:3065-3071. [PMID: 30107088 DOI: 10.1111/ajt.15071] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 07/31/2018] [Accepted: 08/02/2018] [Indexed: 01/25/2023]
Abstract
Immune checkpoint inhibitors are new therapeutic options for metastatic melanoma, but few data are available in organ transplant recipient populations. Six French patients, three men and three women, mean age 66 years (range 44-74), all kidney transplant recipients, received ipilimumab (CTLA-4 inhibitor) for metastatic melanoma. At diagnosis of advanced melanoma, immunosuppressive therapy had been minimized in all but one. Adverse effects included one case of grade 1 diarrhea and one of grade 1 pruritus. One patient had acute T cell-mediated rejection confirmed by histology after the first injection of ipilimumab. After a median follow-up of 4.5 (3-20) months, one patient achieved partial response, one had stable disease, and four had disease progression. All the patients died, five from melanoma, one from another cause. In this series and in the literature, ipilimumab proved to be safe and possibly active. The acute rejection we encountered was probably related to both a rapid, drastic reduction of immunosuppression and the use of ipilimumab. Our safety data on ipilimumab contrast with the organ transplant rejections already reported with PD-1 inhibitors. We consider that immunosuppression should not be minimized, as the impact on metastatic disease control is probably small.
Collapse
Affiliation(s)
- Ouidad Zehou
- Department of Dermatology, Henri Mondor Hospital, APHP, Créteil, France
| | - Claire Leibler
- Department of Nephrology and Transplantation, Henri Mondor Hospital, APHP, Créteil, France
| | | | - Johnny Sayegh
- Department of Nephrology, Dialysis and Transplantation, CHU Angers, Angers, France
| | - Henri Montaudié
- Department of Dermatology, Hôpital Archet 2, and INSERM, U1065, Centre Méditerranéen de Médecine Moléculaire, Nice, France
| | - Philippe Rémy
- Department of Nephrology and Transplantation, Henri Mondor Hospital, APHP, Créteil, France
| | - Denis Glotz
- Department of Nephrology and Transplantation, Saint-Louis Hospital, APHP, Paris, France
| | | | | | - Céleste Lebbé
- APHP Dermatology department, INSERM U976, University Paris Diderot, Saint-Louis Hospital, Paris, France
| |
Collapse
|
35
|
DeLeon TT, Salomao MA, Aqel BA, Sonbol MB, Yokoda RT, Ali AH, Moss AA, Mathur AK, Chascsa DM, Rakela J, Bryce AH, Borad MJ. Pilot evaluation of PD-1 inhibition in metastatic cancer patients with a history of liver transplantation: the Mayo Clinic experience. J Gastrointest Oncol 2018; 9:1054-1062. [PMID: 30603124 DOI: 10.21037/jgo.2018.07.05] [Citation(s) in RCA: 114] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Background Patients with solid organ transplants (SOTs) have been excluded from programmed death protein-1 (PD-1)/programmed death ligand-1 (PD-L1) inhibitor clinical trials due to concern for allograft rejection. The use of immune checkpoint inhibitor therapy remains controversial in transplant patients. Methods A retrospective pilot evaluation was conducted to assess the safety and efficacy of PD-1 inhibitors in patients with liver transplantation (LT). The primary endpoint was the rate of allograft rejection. Secondary endpoints included overall response rate (ORR), progression free survival (PFS) and overall survival (OS). Translational objectives included evaluation of tumor PD-L1, tumor infiltrating lymphocytes (TILs) and allograft PD-L1 expression. Results Seven metastatic cancer patients with a history of LT who received PD-1 inhibitor therapy were included [hepatocellular carcinoma (HCC), n=5; melanoma, n=2]. Rejection was observed in 2 of 7 patients. When rejection occurs it appears to be an early event with a median time to rejection of 24 days in our cohort. One patient achieved a complete response (CR), 3 patients had progressive disease (PD) and 3 patients discontinued therapy prior to restaging assessments. Two of five patients with available tissue had PD-L1 expression in the allograft and both developed rejection. One of five evaluable patients had abundant TILs. Two of five evaluable patients had PD-L1 tumor staining. The single patient with both abundant TILs and PD-L1 staining obtained a response. The median OS and PFS were 1.1 (0.3-21.1) and 1.8 (0.7-21.1) months, respectively. Conclusions In this pilot evaluation both preliminary efficacy (1 of 4) and allograft rejection (2 of 7) were exhibited in evaluable patients. Larger, prospective trials are needed to elucidate optimal patient selection.
Collapse
Affiliation(s)
- Thomas T DeLeon
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Marcela A Salomao
- Division of Anatomic Pathology & Laboratory Medicine, Department of Pathology, Mayo Clinic, Scottsdale, AZ, USA
| | - Bashar A Aqel
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Mohamad B Sonbol
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Raquel T Yokoda
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Ahmad H Ali
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Adyr A Moss
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Mayo Clinic, Scottsdale, AZ, USA
| | - Amit K Mathur
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, Mayo Clinic, Scottsdale, AZ, USA
| | - David M Chascsa
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Jorge Rakela
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Alan H Bryce
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA
| | - Mitesh J Borad
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Scottsdale, AZ, USA.,Department of Molecular Medicine, Mayo Clinic, Rochester, MN, USA.,Mayo Clinic Cancer Center, Phoenix, AZ, USA
| |
Collapse
|
36
|
Regalla DKR, Williams GR, Paluri RK. Immune checkpoint inhibitors in the management of malignancies in transplant recipients. Postgrad Med J 2018; 94:704-708. [DOI: 10.1136/postgradmedj-2018-136081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/10/2018] [Accepted: 10/16/2018] [Indexed: 12/26/2022]
Abstract
Cancer immunotherapy, an area of active research, has thus far yielded several exciting breakthroughs in cancer treatment strategies. So far, immune checkpoint inhibitors have been the most promising method of cancer immunotherapy. CTLA-4, PD-1 and PD-L1 are the immune checkpoint molecules against which monoclonal antibodies act against and revolutionised the treatment of several malignancies. However, it is still unclear whether using these monoclonal antibodies in patients with malignancy and a history of transplant is as beneficial as in patients without a history of transplantation. The reason being, with the therapeutic benefit, also comes the inherent disadvantage of transplant rejection because of the activation of T-cells against donor antigens. So, transplant-related complications limit the usage of the checkpoint blockade therapy to treat malignancies. Here, we review the data published in this context and suggest optimal approaches to using the currently available repertoire of immunotherapies.
Collapse
|
37
|
Aguirre LE, Guzman ME, Lopes G, Hurley J. Immune Checkpoint Inhibitors and the Risk of Allograft Rejection: A Comprehensive Analysis on an Emerging Issue. Oncologist 2018; 24:394-401. [PMID: 30413665 DOI: 10.1634/theoncologist.2018-0195] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 09/06/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND It is well known that the state of immune tolerance induced by broad immunosuppression to prevent allograft rejection leads to an increased risk of the development of cancer. One of the most promising new areas of cancer treatment has been the development of immune checkpoint inhibitors that target the cytotoxic T-lymphocyte-associated antigen 4 and programmed cell death protein 1/programmed death-ligand 1 (PD-L1) pathways. As a logical consequence, growing interest in these agents translated into their implementation in patients with transplant-related malignancies. Because of overlapping and perhaps mutually exclusive mechanisms of action of transplant immunosuppression and cancer immunomodulation, it is critical to examine these interactions. MATERIALS AND METHODS We carried out a systematic search for review articles and case reports published between July 2014 and November 2017 using three engines: Usearch, PubMed, and Up-to-date. RESULTS Overall, there were 20 cases with 12 allograft rejections. The rejection rate associated with nivolumab was 73% (8/11) and with pembrolizumab it was 100% (2/2). The use of ipilimumab did not lead to rejection in any instance (0/4, 0%). Of the two patients treated with the sequential use of ipilimumab/nivolumab, one lost his allograft, yielding a rejection rate of 50%. The sequential use of ipilimumab/pembrolizumab led to a rejection rate of 100% (1/1, 100%). CONCLUSION The use of agents that act on the PD-L1 pathway are contraindicated in the face of solid organ allografts because of unacceptably high rates of irreversible allograft rejection. It appears that the use of ipilimumab may be tolerated as the mechanism is different from that of the PD-L1 agents. IMPLICATIONS FOR PRACTICE Transplant rejection is a complex process that puts stress on patients and their families and can lead to tragic results. Significant advancements in the field of immunosuppression have led to the engenderment of agents devised to extend the survival of transplant recipients. The advent of immunomodulators in cancer therapy has been paradigm-shifting; however, because of their mechanism of action, their use must be carefully considered in patients with allografts and concomitant cancer. It appears that ipilimumab can be administered safely in these patients but that agents acting on the programmed death-ligand 1 pathway are contraindicated because of high rates of irreversible rejection.
Collapse
Affiliation(s)
- Luis E Aguirre
- Department of Internal Medicine, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, Florida, USA
| | - Maria E Guzman
- Department of Internal Medicine, University of Miami Miller School of Medicine/Jackson Memorial Hospital, Miami, Florida, USA
| | - Gilberto Lopes
- Department of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Judith Hurley
- Department of Hematology/Oncology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
38
|
Kissick HT. Is It Possible to Develop Cancer Vaccines to Neoantigens, What Are the Major Challenges, and How Can These Be Overcome? Neoantigens as Vaccine Targets for Cancer. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a033704. [PMID: 29254981 DOI: 10.1101/cshperspect.a033704] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent work by several groups has undoubtedly shown that we can produce cancer vaccines targeting neoantigens. However, each vaccine is essentially a single-use, patient-specific product, making this approach resource-intensive. For this reason, it is important to ask whether this approach will be any more successful than what has been attempted during the last 30 years using vaccines targeting self-epitopes. Here, we discuss what might be expected from neoantigen vaccines based on our experience in chronic viral infections, and how this new approach may be applied to cancer immunotherapy.
Collapse
Affiliation(s)
- Haydn T Kissick
- Departments of Urology and Microbiology and Immunology, Emory University School of Medicine, Atlanta, Georgia 30322
| |
Collapse
|
39
|
Tio M, Rai R, Ezeoke OM, McQuade JL, Zimmer L, Khoo C, Park JJ, Spain L, Turajlic S, Ardolino L, Yip D, Goldinger SM, Cohen JV, Millward M, Atkinson V, Kane AY, Ascierto PA, Garbe C, Gutzmer R, Johnson DB, Rizvi HA, Joshua AM, Hellmann MD, Long GV, Menzies AM. Anti-PD-1/PD-L1 immunotherapy in patients with solid organ transplant, HIV or hepatitis B/C infection. Eur J Cancer 2018; 104:137-144. [PMID: 30347289 PMCID: PMC10176037 DOI: 10.1016/j.ejca.2018.09.017] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 09/20/2018] [Indexed: 02/06/2023]
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.
Collapse
Affiliation(s)
- Martin Tio
- Melanoma Institute Australia, Sydney, Australia.
| | - Rajat Rai
- Melanoma Institute Australia, Sydney, Australia
| | | | | | - Lisa Zimmer
- University of Duisburg-Essen Hospital, Heidelberg, Germany
| | - Chloe Khoo
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - John J Park
- Crown Princess Mary Cancer Centre, Sydney, Australia; Westmead Hospital, Sydney, Australia
| | - Lavinia Spain
- Skin and Renal Unit, The Royal Marsden NHS Foundation Trust, London, UK
| | - Samra Turajlic
- Skin and Renal Unit, The Royal Marsden NHS Foundation Trust, London, UK; Francis Crick Institute, London, UK
| | | | - Desmond Yip
- The Canberra Hospital, Canberra, Australia; ANU Medical School, Australian National University, Canberra, Australia
| | | | | | | | | | - Alisa Y Kane
- Liverpool Hospital, Sydney, Australia; Garvan Institute, Sydney, Australia
| | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | | | | | | | - Hira A Rizvi
- Memorial Sloan Kettering Cancer Center, New York, USA
| | - Anthony M Joshua
- Melanoma Institute Australia, Sydney, Australia; St Vincents Hospital, Sydney, Australia; University of New South Wales, Sydney, Australia
| | | | - Georgina V Long
- Melanoma Institute Australia, Sydney, Australia; The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia
| | - Alexander M Menzies
- Melanoma Institute Australia, Sydney, Australia; The University of Sydney, Sydney, Australia; Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
40
|
Grant MJ, DeVito N, Salama AKS. Checkpoint inhibitor use in two heart transplant patients with metastatic melanoma and review of high-risk populations. Melanoma Manag 2018; 5:MMT10. [PMID: 30459942 PMCID: PMC6240846 DOI: 10.2217/mmt-2018-0004] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 08/03/2018] [Indexed: 12/12/2022] Open
Abstract
Due to the unique side-effect profile of immune checkpoint inhibitors (ICIs), groups of patients deemed to be at high risk of complications were excluded from trials that proved the efficacy and safety of these agents in patients with various malignancies. Among these excluded patients were those with prior solid organ transplantation, chronic viral infections and pre-existing autoimmune diseases including paraneoplastic syndromes. We present follow-up on a patient from a previously published case report with an orthotopic heart transplantation who was treated with both cytotoxic T-lymphocyte antigen 4 and PD-1 inhibition safely, without organ rejection. Additionally, we describe the case of a patient with a cardiac allograft who also did not experience organ rejection after treatment with pembrolizumab. Through smaller trials, retrospective analyses, case series and individual case reports, we are accumulating initial data on how these agents are tolerated by the aforementioned groups. Our survey of the literature has found more evidence of organ transplant rejection in patients treated with PD-1 inhibitors than those treated with inhibitors of cytotoxic T-lymphocyte antigen 4. Patients with chronic viral infections, especially hepatitis C, seem to have little to no risk of treatment-related increase in serum RNA levels. The literature contains few documented cases of devastating exacerbations of pre-existing autoimmune disease during treatment with ICIs, and flares seem to be easily controlled by immunosuppression in the vast majority of cases. Last, several cases allude to a promising role for disease-specific antibodies and other serum biomarkers in identifying patients at high risk of developing certain immune-related adverse events, detecting subclinical immune-related adverse event onset, and monitoring treatment response to immunosuppressive therapy in patients treated with ICIs. Though these excluded populations have not been well studied in randomized placebo-controlled trials, we may be able to learn and derive hypotheses from the existing observational data in the literature.
Collapse
Affiliation(s)
- Michael J Grant
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA.,Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | - Nicholas DeVito
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| | - April K S Salama
- Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA.,Duke Cancer Institute, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
41
|
Akturk HK, Alkanani A, Zhao Z, Yu L, Michels AW. PD-1 Inhibitor Immune-Related Adverse Events in Patients With Preexisting Endocrine Autoimmunity. J Clin Endocrinol Metab 2018; 103:3589-3592. [PMID: 30124874 PMCID: PMC6179163 DOI: 10.1210/jc.2018-01430] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 08/09/2018] [Indexed: 01/01/2023]
Abstract
CONTEXT Immune checkpoint inhibitors, including monoclonal antibodies directed against programmed cell death protein 1 (PD-1) and its ligand, have emerged as beneficial cancer immunotherapies. These therapies are known to cause immune-related side effects; however, their role in patients with a preexisting autoimmune disease is not clear. CASE DESCRIPTION We describe two cases of anti-PD-1 immune-related adverse events. A 52-year-old male with longstanding type 1 diabetes (T1D), long-term stable kidney transplant, and hypothyroidism received two separate anti-PD-1 monoclonal antibodies for metastatic melanoma. The patient developed acute kidney graft rejection requiring hemodialysis and worsening of autoimmune hypothyroidism 3 weeks after starting treatment. He continued anti-PD-1 treatments and remained on hemodialysis and increased levothyroxine dosage. The second case is a 62-year-old male with no previous history of diabetes who received anti-PD-1 treatment and developed severe diabetic ketoacidosis (DKA) 5 days following the start of therapy. Further laboratory testing revealed high titer antibodies directed against glutamic acid decarboxylase. These antibodies, which were of the IgG isotype and involved in memory immune responses, were likely present before anti-PD-1 treatment. He also had human leukocyte antigen genes that confer T1D genetic risk. Despite normal pretreatment blood glucose levels and HbA1c, the patient requires permanent exogenous insulin treatment. CONCLUSION Patients with preexisting endocrine autoimmunity may have more frequent and severe immune-related side effects with anti-PD-1 treatment. Given the morbidity and mortality associated with solid organ transplant rejection and DKA, clinicians caring for patients receiving these state-of-the-art therapies need to be aware of the potential adverse events.
Collapse
Affiliation(s)
- Halis Kaan Akturk
- Barbara Davis Center for Diabetes, University of Colorado, School of Medicine, Aurora, Colorado
| | - Aimon Alkanani
- Barbara Davis Center for Diabetes, University of Colorado, School of Medicine, Aurora, Colorado
| | - Zhiyuan Zhao
- Barbara Davis Center for Diabetes, University of Colorado, School of Medicine, Aurora, Colorado
| | - Liping Yu
- Barbara Davis Center for Diabetes, University of Colorado, School of Medicine, Aurora, Colorado
| | - Aaron W Michels
- Barbara Davis Center for Diabetes, University of Colorado, School of Medicine, Aurora, Colorado
- Correspondence and Reprint Requests: Aaron W. Michels, MD, Barbara Davis Center for Diabetes, University of Colorado School of Medicine, 1775 Aurora Ct A-140, Aurora, Colorado 80045. E-mail:
| |
Collapse
|
42
|
Smedman TM, Line PD, Guren TK, Dueland S. Graft rejection after immune checkpoint inhibitor therapy in solid organ transplant recipients. Acta Oncol 2018; 57:1414-1418. [PMID: 29912605 DOI: 10.1080/0284186x.2018.1479069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/14/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Tor Magnus Smedman
- a Department of Oncology , Oslo University Hospital , Oslo , Norway
- b Institute of Clinical Medicine , University of Oslo , Oslo , Norway
| | - Pål-Dag Line
- b Institute of Clinical Medicine , University of Oslo , Oslo , Norway
- c Section for Transplantation Surgery, Department of Transplantation Medicine , Oslo University Hospital , Oslo , Norway
| | - Tormod K Guren
- a Department of Oncology , Oslo University Hospital , Oslo , Norway
| | - Svein Dueland
- a Department of Oncology , Oslo University Hospital , Oslo , Norway
| |
Collapse
|
43
|
Cardiac Complications Associated With Checkpoint Inhibition: A Systematic Review of the Literature in an Important Emerging Area. Can J Cardiol 2018; 34:1059-1068. [DOI: 10.1016/j.cjca.2018.03.012] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 01/22/2023] Open
|
44
|
Liver Allograft Failure After Nivolumab Treatment-A Case Report With Systematic Literature Research. Transplant Direct 2018; 4:e376. [PMID: 30255136 PMCID: PMC6092180 DOI: 10.1097/txd.0000000000000814] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 02/06/2023] Open
Abstract
Background Orthotopic liver transplantation (OLT) is a potential curative treatment in patients with hepatocellular carcinoma (HCC); however, treatment options for recurrent HCC after OLT are limited. Immune checkpoint inhibitors, such as nivolumab, an inhibitor of programmed cell death protein 1, have been successfully used for metastatic HCC but data on safety of nivolumab following solid organ transplantation are limited. Methods We report a 53-year-old woman with HCC who was treated with OLT. After 2 years, HCC recurred. Initial treatment with sorafenib was discontinued due to side effects and disease progression. Progressive HCC in the lung and lymph nodes was subsequently treated with nivolumab. One week after the first nivolumab dose, rapid progressive liver dysfunction was noted. Liver biopsy revealed severe cellular graft rejection prompting treatment with intravenous steroids and tacrolimus. Liver function continued to decline, leading to severe coagulopathy. The patient succumbed to intracranial hemorrhage. Results A systematic PubMed search revealed 29 cases treated with a checkpoint inhibitor following solid organ transplantation. Loss of graft was described in 4 (36%) of 11 cases with OLT and in 7 (54%) of 13 cases after kidney transplantation. However, cases with favorable outcome were also described. Eighteen cases with adverse events were identified upon searching the World Health Organization database VigiBase, including 2 cases with fatal outcome in liver transplant recipients due to graft loss. Conclusion Experience with checkpoint inhibitors in solid organ transplant recipients is limited. Published cases so far suggest severe risks for graft loss as high as 36% to 54%.
Collapse
|
45
|
Wong AS, Thian YL, Kapur J, Leong CN, Kee P, Lee CT, Lee MB. Pushing the limits of immune-related response: a case of "extreme pseudoprogression". Cancer Immunol Immunother 2018; 67:1105-1111. [PMID: 29728723 PMCID: PMC6006242 DOI: 10.1007/s00262-018-2167-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 04/30/2018] [Indexed: 12/14/2022]
Abstract
The advent of immune checkpoint targeted immunotherapy has seen a spectrum of immune-related phenomena in both tumor responses and toxicities. We describe a case of pseudoprogression that pushes the limits of immune-related response criteria and challenges the boundaries and definitions set by trial protocols. A middle-aged man with conventional clear cell renal cell carcinoma (RCC) had received multiple prior systemic treatments including vascular endothelial growth factor receptor tyrosine kinase inhibitors, as well as multiple surgeries and radiotherapy treatments. He was eventually started on nivolumab—the anti-programmed death receptor-1 monoclonal antibody approved for the treatment of advanced RCC. Clinical deterioration was observed soon after a 100 mg dose of nivolumab, with onset of acute renal failure and declining performance status. Radiologic progression was documented in multiple sites including worsening tumor infiltration of his residual kidney. The patient was on palliative treatment and visited by the home hospice team in an end-of-life situation. The patient unexpectedly improved and went on to achieve a durable tumor response. The case is illustrative of an extreme manifestation of pseudoprogression, and impels us to probe the assumptions and controversies surrounding this phenomenon.
Collapse
Affiliation(s)
- Alvin S Wong
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, NUHS Tower Block level 7, 1E Kent Ridge Road, Singapore, 119228, Singapore.
| | - Yee-Liang Thian
- Department of Diagnostic Imaging, National University Health System, Singapore, Singapore
| | - Jeevesh Kapur
- Department of Diagnostic Imaging, National University Health System, Singapore, Singapore
| | - Cheng-Nang Leong
- Department of Radiation Oncology, National University Cancer Institute, National University Health System, Singapore, Singapore
| | - Patrick Kee
- Hospice Care Association, Singapore, Singapore
| | - Chun-Tsu Lee
- Department of Hematology-Oncology, National University Cancer Institute, National University Health System, NUHS Tower Block level 7, 1E Kent Ridge Road, Singapore, 119228, Singapore
| | - Martin B Lee
- Department of Medicine, National University Health System, Singapore, Singapore
| |
Collapse
|
46
|
Postow MA, Sidlow R, Hellmann MD. Immune-Related Adverse Events Associated with Immune Checkpoint Blockade. N Engl J Med 2018; 378:158-168. [PMID: 29320654 DOI: 10.1056/nejmra1703481] [Citation(s) in RCA: 3045] [Impact Index Per Article: 435.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Michael A Postow
- From Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York
| | - Robert Sidlow
- From Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York
| | - Matthew D Hellmann
- From Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York
| |
Collapse
|
47
|
Chae YK, Galvez C, Anker JF, Iams WT, Bhave M. Cancer immunotherapy in a neglected population: The current use and future of T-cell-mediated checkpoint inhibitors in organ transplant patients. Cancer Treat Rev 2017; 63:116-121. [PMID: 29276997 DOI: 10.1016/j.ctrv.2017.12.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/04/2017] [Accepted: 12/06/2017] [Indexed: 12/22/2022]
Abstract
Although the indications for immune checkpoint inhibitors continue to grow, organ transplant recipients with advanced malignancies have been largely excluded from clinical trials testing the safety and efficacy of these therapies given their need for chronic immunosuppression and the risk of allograft rejection. With the rapid growth of transplant medicine and the increased risk of malignancy associated with chronic immunosuppression, it is critical that we systematically analyze the available data describing immune checkpoint blockade in the organ transplant population. Herein we provide a current and comprehensive review of cases in which immune checkpoint blockade was used on organ transplant recipients. Furthermore, we discuss the differences in efficacy and risk of allograft rejection between CTLA-4 and PD-1 inhibitors and make recommendations based on the limited available clinical data. We also discuss the future of immune checkpoint blockade in this subpopulation and explore the emerging data of promising combination therapies with mTOR, BRAF/MEK, and BTK/ITK inhibitors. Further clinical experience and larger clinical trials involving immune checkpoint inhibitors, whether as monotherapies or combinatorial therapies, will help develop regimens that optimize anti-tumor response and minimize the risk of allograft rejection in organ transplant patients.
Collapse
Affiliation(s)
- Young Kwang Chae
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA.
| | - Carlos Galvez
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jonathan F Anker
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Wade T Iams
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Manali Bhave
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
48
|
Yoest JM. Clinical features, predictive correlates, and pathophysiology of immune-related adverse events in immune checkpoint inhibitor treatments in cancer: a short review. Immunotargets Ther 2017; 6:73-82. [PMID: 29067284 PMCID: PMC5644546 DOI: 10.2147/itt.s126227] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Identification and characterization of T-cell regulatory mechanisms, or checkpoints, have led to a wave of drug development aimed at inhibiting these targets to "remove the brakes" of the immune system. This class of anticancer therapeutics, termed immune checkpoint inhibitors (ICIs), has harnessed the potential of the body's own immune system to recognize cancerous cells and selectively eliminate them, in some cases with alarming success. This new breakthrough, however, has not been without its drawbacks. Immune-related adverse events (irAEs) are adverse events encountered during treatment with ICIs that are thought to be mediated through the patient's immune system which can manifest with a variety of symptoms which often resemble autoimmunity. These events range widely in presentation and severity and are reported frequently. Here, we will discuss a large selection of case reports in order to inform the clinician, laboratorian, and researcher of the scope of organ systems affected, the severity of the conditions being encountered, and the responses of these events to treatment, as well as explore the use of ICIs in the setting of preexisting autoimmunity. We will also consider the ability to detect autoantibodies before and during irAEs as well as the correlations that irAEs have with clinical outcomes. Finally, we will conclude by exploring the possibility that two distinct pathways may be contributing to the phenomenon of irAEs within this class of drugs, and the role that this might play in future research and clinical practice.
Collapse
Affiliation(s)
- Jennifer M Yoest
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
49
|
|
50
|
Schvartsman G, Perez K, Flynn JE, Myers JN, Tawbi H. Safe and effective administration of T-VEC in a patient with heart transplantation and recurrent locally advanced melanoma. J Immunother Cancer 2017; 5:45. [PMID: 28642816 PMCID: PMC5477138 DOI: 10.1186/s40425-017-0250-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 05/15/2017] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Immunotherapy plays a key role in the treatment of metastatic melanoma. Patients with autoimmune conditions and/or on immunosuppressive therapy due to orthotropic transplants, however, are systematically excluded from clinical trials. Talimogene laherparepvec (T-VEC) is the first oncolytic virus to be approved by the FDA for cancer therapy. To our knowledge, this is the first report of T-VEC being administered in the setting of an organ transplant recipient. CASE PRESENTATION Here we present the case of a patient with recurrent locally advanced cutaneous melanoma receiving salvage T-VEC therapy in the setting of orthotropic heart transplantation. After 5 cycles of therapy, no evidence of graft rejection has been observed to date, and the patient achieved a complete remission, and is currently off therapy. CONCLUSION This case advocates for further investigation on the safety and efficacy of immunotherapeutic approaches, such as T-VEC, in solid organ transplant recipients.
Collapse
Affiliation(s)
- Gustavo Schvartsman
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd, Unit 463, Houston, TX 77030 USA
| | - Kristen Perez
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd., Houston, TX 77030 USA
| | - Jill E. Flynn
- Department of Head & Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, USA
| | - Jeffrey N. Myers
- Department of Head & Neck Surgery, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, USA
| | - Hussein Tawbi
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd., Houston, TX 77030 USA
- Department of Melanoma Medical Oncology, Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, 1400 Holcombe Blvd., Houston, TX 77030 USA
| |
Collapse
|