1
|
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
|
2
|
Joo V, Abdelhamid K, Noto A, Latifyan S, Martina F, Daoudlarian D, De Micheli R, Pruijm M, Peters S, Hullin R, Gaide O, Pantaleo G, Obeid M. Primary prophylaxis with mTOR inhibitor enhances T cell effector function and prevents heart transplant rejection during talimogene laherparepvec therapy of squamous cell carcinoma. Nat Commun 2024; 15:3664. [PMID: 38693123 PMCID: PMC11063183 DOI: 10.1038/s41467-024-47965-3] [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: 08/03/2023] [Accepted: 04/15/2024] [Indexed: 05/03/2024] Open
Abstract
The application of mammalian target of rapamycin inhibition (mTORi) as primary prophylactic therapy to optimize T cell effector function while preserving allograft tolerance remains challenging. Here, we present a comprehensive two-step therapeutic approach in a male patient with metastatic cutaneous squamous cell carcinoma and heart transplantation followed with concomitant longitudinal analysis of systemic immunologic changes. In the first step, calcineurin inhibitor/ mycophenolic acid is replaced by the mTORi everolimus to achieve an improved effector T cell status with increased cytotoxic activity (perforin, granzyme), enhanced proliferation (Ki67) and upregulated activation markers (CD38, CD69). In the second step, talimogene laherparepvec (T-VEC) injection further enhances effector function by switching CD4 and CD8 cells from central memory to effector memory profiles, enhancing Th1 responses, and boosting cytotoxic and proliferative activities. In addition, cytokine release (IL-6, IL-18, sCD25, CCL-2, CCL-4) is enhanced and the frequency of circulating regulatory T cells is increased. Notably, no histologic signs of allograft rejection are observed in consecutive end-myocardial biopsies. These findings provide valuable insights into the dynamics of T cell activation and differentiation and suggest that timely initiation of mTORi-based primary prophylaxis may provide a dual benefit of revitalizing T cell function while maintaining allograft tolerance.
Collapse
Affiliation(s)
- Victor Joo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Karim Abdelhamid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Alessandra Noto
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Sofiya Latifyan
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Federica Martina
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Douglas Daoudlarian
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Rita De Micheli
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Menno Pruijm
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Nephrology Division, Rue du Bugnon 17, CH-1011, Lausanne, Switzerland
| | - Solange Peters
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Oncology Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Roger Hullin
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Cardiology, Cardiovascular Department, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Olivier Gaide
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Dermatology Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Giuseppe Pantaleo
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland
| | - Michel Obeid
- Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne, Department of Medicine, Immunology and Allergy Division, Rue du Bugnon 46, CH-1011, Lausanne, Switzerland.
| |
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
|
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]
|
6
|
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
|
7
|
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
|
8
|
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
|
9
|
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
|
10
|
Hu W, Wang G, Wang Y, Riese MJ, You M. Uncoupling Therapeutic Efficacy from Immune-Related Adverse Events in Immune Checkpoint Blockade. iScience 2020; 23:101580. [PMID: 33083746 PMCID: PMC7554032 DOI: 10.1016/j.isci.2020.101580] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Immunotherapy with monoclonal antibodies targeting immune checkpoint molecules, including programmed death-1 (PD-1), PD ligand-1 (PD-L1), and cytotoxic T-lymphocyte-associated antigen (CTLA)-4, has become prominent in the treatment of many types of cancer. However, a significant number of patients treated with immune checkpoint inhibitors (ICIs) develop immune-related adverse events (irAEs). irAEs can affect any organ system, and although most are clinically manageable, irAEs can result in mortality or long-term morbidity. Factors that can predict irAEs remain elusive. Understanding the etiology of ICI-induced irAEs and ways to limit these adverse events are needed. In this review, we provide basic science and clinical insights on the mechanisms responsible for ICI efficacy and ICI-induced irAEs. We further provide insights into approaches that may uncouple irAEs from the ability of ICIs to kill tumor cells.
Collapse
Affiliation(s)
- Weilei Hu
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Guosheng Wang
- Department of Biomedical Engineering, Binghamton University—SUNY, 4400 Vestal Pkwy E, Binghamton, NY 13902, USA
| | - Yian Wang
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Matthew J. Riese
- Department of Medicine, Division of Hematology/Oncology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
- Blood Research Institute, Versiti Inc, Milwaukee, WI 53226, USA
| | - Ming You
- Center for Disease Prevention Research and Department of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| |
Collapse
|
11
|
Goebell PJ, Ivanyi P, Bedke J, Bergmann L, Berthold D, Boegemann M, Busch J, Doehn C, Krege S, Retz M, Amsberg GV, Grimm MO, Gruenwald V. Consensus paper: current state of first- and second-line therapy in advanced clear-cell renal cell carcinoma. Future Oncol 2020; 16:2307-2328. [PMID: 32964728 DOI: 10.2217/fon-2020-0403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The therapy of advanced (clear-cell) renal cell carcinoma (RCC) has recently experienced tremendous changes. Several new treatments have been developed, with PD-1 immune-checkpoint inhibition being the backbone of therapy. Diverse immunotherapy combinations change current first-line standards. These changes also require new approaches in subsequent lines of therapy. In an expert panel, we discussed the new treatment options and how they change clinical practice. While first-line immunotherapies introduce a new level of response rates, data on second-line therapies remains poor. This scenario poses a challenge for clinicians as guideline recommendations are based on historical patient cohorts and agents may lack the appropriate label for their in guidelines recommended use. Here, we summarize relevant clinical data and consider appropriate treatment strategies.
Collapse
Affiliation(s)
- Peter J Goebell
- Division of Urology, University Hospital Erlangen, D-91054, Erlangen, Germany
| | - Philipp Ivanyi
- Department of Hematology, Hemostaseology, Oncology & Stem Cell Transplantation, Hannover Medical School, D-30625, Hannover, Germany
| | - Jens Bedke
- Department of Urology, Eberhard Karls University, D-72076, Tuebingen, Germany
| | - Lothar Bergmann
- University Hospital Frankfurt, Medical Clinic II, D-60590, Frankfurt, Germany
| | - Dominik Berthold
- Centre Hospitalier Universitaire Vaudois, CH-1011, Lausanne, Switzerland
| | - Martin Boegemann
- Department of Urology, University Hospital Münster, D-48149, Münster, Germany
| | - Jonas Busch
- Department of Urology, Charité-University Medicine, D-10117, Berlin, Germany
| | | | - Susanne Krege
- Department of Urology, Pediatric Urology & Urologic Oncology, Kliniken Essen-Mitte, D-45136, Essen, Germany
| | - Margitta Retz
- Department of Urology, Technical University of Munich, Rechts der Isar Medical Center, D-81675, Munich, Germany
| | - Gunhild von Amsberg
- Department of Oncology & Hematology, University Cancer Center Hamburg & Martini-Clinic, University Medical Center Hamburg-Eppendorf, D-20246, Hamburg, Germany
| | - Marc-Oliver Grimm
- Department of Urology, Jena University Hospital, D-07747, Jena, Germany
| | - Viktor Gruenwald
- Interdisciplinary GU Oncology, Clinic for Medical Oncology & Clinic for Urology, University Hospital Essen, D-45147, Essen, Germany
| |
Collapse
|
12
|
Owoyemi I, Vaughan LE, Costello CM, Thongprayoon C, Markovic SN, Herrmann J, Otley CC, Taner T, Mangold AR, Leung N, Herrmann SM, Kukla A. Clinical outcomes of solid organ transplant recipients with metastatic cancers who are treated with immune checkpoint inhibitors: A single-center analysis. Cancer 2020; 126:4780-4787. [PMID: 32786022 DOI: 10.1002/cncr.33134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 06/07/2020] [Accepted: 07/13/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy, but to the authors' knowledge, limited data exist regarding the safety and efficacy of these agents in transplant recipients. Herein, the authors have reported their experience with 17 patients who were treated with ICIs for metastatic malignancies after undergoing solid organ transplantation. METHODS Data were abstracted for solid organ transplant recipients who received ICIs for the treatment of malignancy between January 1, 2016, and September 30, 2019. The authors identified 7 kidney, 8 liver, and 2 heart transplant recipients. Outcomes of interest were adverse drug reactions, cancer progression, and patient survival. RESULTS The most common malignancies treated with ICIs were metastatic squamous cell carcinoma (5 patients; 29%) and hepatocellular carcinoma (5 patients; 29%), which were noted exclusively among liver transplant recipients. The median duration on ICIs was 1.7 months (interquartile range, 0.4-7.6 months). Five patients (29%) developed adverse reactions, including 4 patients (24%) with immune-related adverse events(irAEs), 3 patients (18%) with acute allograft rejections, 1 patient (6%) with autoimmune colitis, and 1 patient (6%) with ICI-induced cardiotoxicity (the patient was a heart transplant recipient). The cumulative incidence of cancer progression was 50% and 69%, respectively, at 6 months and 12 months. Eleven patients (65%) died over the median follow-up period of 4.6 months (interquartile range, 1.5-13.2 months) from the time of ICI initiation, with cancer progression being the most common cause of death. CONCLUSIONS ICIs can be used as individualized therapy in selected patients who have undergone solid organ transplantation but more studies are needed to determine how best to use these agents to improve outcomes further.
Collapse
Affiliation(s)
- Itunu Owoyemi
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Lisa E Vaughan
- Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | | | | | | | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| | - Clark C Otley
- Department of Dermatology, Mayo Clinic, Rochester, Minnesota
| | - Timucin Taner
- Department of Surgery, Mayo Clinic, Rochester, Minnesota.,William J. von Liebig Transplant Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota.,Department of Immunology, Mayo Clinic, Rochester, Minnesota
| | | | - Nelson Leung
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Aleksandra Kukla
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota.,William J. von Liebig Transplant Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
13
|
Richter MD, Hughes GC, Chung SH, Ezeanuna M, Singh N, Thompson JA. Immunologic adverse events from immune checkpoint therapy. Best Pract Res Clin Rheumatol 2020; 34:101511. [DOI: 10.1016/j.berh.2020.101511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
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
|
15
|
Immune checkpoint inhibitors in heart or lung transplantation: Early results from a registry initiative. J Heart Lung Transplant 2020; 39:604-606. [DOI: 10.1016/j.healun.2020.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 01/15/2023] Open
|
16
|
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]
|
17
|
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
|
18
|
Haanen J, Ernstoff MS, Wang Y, Menzies AM, Puzanov I, Grivas P, Larkin J, Peters S, Thompson JA, Obeid M. Autoimmune diseases and immune-checkpoint inhibitors for cancer therapy: review of the literature and personalized risk-based prevention strategy. Ann Oncol 2020; 31:724-744. [PMID: 32194150 DOI: 10.1016/j.annonc.2020.03.285] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 03/03/2020] [Accepted: 03/07/2020] [Indexed: 02/08/2023] Open
Abstract
Patients with cancer and with preexisting active autoimmune diseases (ADs) have been excluded from immunotherapy clinical trials because of concerns for high susceptibility to the development of severe adverse events resulting from exacerbation of their preexisting ADs. However, a growing body of evidence indicates that immune-checkpoint inhibitors (ICIs) may be safe and effective in this patient population. However, baseline corticosteroids and other nonselective immunosuppressants appear to negatively impact drug efficacy, whereas retrospective and case report data suggest that use of specific immunosuppressants may not have the same consequences. Therefore, we propose here a two-step strategy. First, to lower the risk of compromising ICI efficacy before their initiation, nonselective immunosuppressants could be replaced by specific selective immunosuppressant drugs following a short rotation phase. Subsequently, combining ICI with the selective immunosuppressant could prevent exacerbation of the AD. For the most common active ADs encountered in the context of cancer, we propose specific algorithms to optimize ICI therapy. These preventive strategies go beyond current practices and recommendations, and should be practiced in ICI-specialized clinics, as these require multidisciplinary teams with extensive knowledge in the field of clinical immunology and oncology. In addition, we challenge the exclusion from ICI therapy for patients with cancer and active ADs and propose the implementation of an international registry to study such novel strategies in a prospective fashion.
Collapse
Affiliation(s)
- J Haanen
- Netherlands Cancer Institute, Division of Medical Oncology, Amsterdam, The Netherlands
| | - M S Ernstoff
- Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Y Wang
- Department of Gastroenterology, Hepatology & Nutrition, University of Texas MD Anderson Cancer Center, Houston, USA
| | - A M Menzies
- Melanoma Institute Australia, The University of Sydney, Sydney, Australia; Royal North Shore and Mater Hospitals, Sydney, Australia
| | - I Puzanov
- Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - P Grivas
- University of Washington, Seattle Cancer Care Alliance, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - J Larkin
- Royal Marsden NHS Foundation Trust, London, UK
| | - S Peters
- Oncology Department, Centre Hospitalier Universitaire Vaudois (CHUV) and Lausanne University, Lausanne, Switzerland
| | - J A Thompson
- University of Washington, Seattle Cancer Care Alliance, Fred Hutchinson Cancer Research Center, Seattle, USA; National Cancer Institute/NIH, Bethesda, USA
| | - M Obeid
- Department of Medicine, Service of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland; Vaccine and Immunotherapy Center, Centre Hospitalier Universitaire Vaudois (CHUV), Centre d'Immunothérapie et de Vaccinologie, Lausanne, Switzerland.
| |
Collapse
|
19
|
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
|
20
|
Stevenson ML, Carucci J, Colegio OR. Skin cancer in transplant recipients: Scientific retreat of the international immunosuppression and transplant skin cancer collaborative and skin care in organ transplant patients—Europe. Clin Transplant 2019; 33:e13736. [DOI: 10.1111/ctr.13736] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 09/30/2019] [Accepted: 10/04/2019] [Indexed: 01/02/2023]
Affiliation(s)
- Mary L. Stevenson
- The Ronald O. Perelman Department of Dermatology NYU Langone Health New York New York
| | - John Carucci
- The Ronald O. Perelman Department of Dermatology NYU Langone Health New York New York
| | | |
Collapse
|