1
|
Arata R, Tanimine N, Seidakhmetov A, Ide K, Tanaka Y, Ohdan H. Discrimination of Anti-Donor Response in Allogeneic Transplantation Using an Alloreactive T-Cell Detection Assay. Transpl Int 2025; 38:13879. [PMID: 39936124 PMCID: PMC11810569 DOI: 10.3389/ti.2025.13879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 01/08/2025] [Indexed: 02/13/2025]
Abstract
Understanding donor-reactive T-cell behavior post-transplantation is challenging owing to the rarity and diversity of these cells. Here, we aimed to evaluate the relevance of an assay for rapidly detecting alloreactive T cells in a mouse transplantation model. After 18 h of one-way mixed lymphocyte reaction (MLR) culture with pre-activated donor-derived stimulators, CD4+ and CD8+ donor-reactive T cells were identified by CD154 and CD137 expression, respectively. Using full MHC mismatched mouse skin transplant models, we observed an increased donor-reactive T-cell proportion by direct presentation with elevated interferon gamma and granzyme B production 7 days post-transplantation, before graft rejection. Immunosuppression with CTLA-4 IgG and anti-CD154 antibody varied depending on donor-recipient strain combinations. On day 7, donor-reactive CD8+ T-cell proportions were lower in the tolerance model (BALB/c to C3H/HeJ) than in the rejection model (BALB/c to C57BL/6); conventional proliferation readout after 4 days of MLR could not distinguish these responses. Overall, although the conventional readout for evaluating T-cell proliferation following an MLR quantifies the precursor frequency of alloreactive T cells, the assay reported herein assesses T-cell activation markers after a short-term MLR to characterize immediate immune status. These findings offer a promising tool to elucidate immune responses post-transplantation.
Collapse
Affiliation(s)
| | - Naoki Tanimine
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | | | | | | | | |
Collapse
|
2
|
Herrmann SM, Abudayyeh A, Gupta S, Gudsoorkar P, Klomjit N, Motwani SS, Karam S, Costa E Silva VT, Khalid SB, Anand S, Kala J, Leaf DE, Murakami N, Rashidi A, Wanchoo R, Kitchlu A. Diagnosis and management of immune checkpoint inhibitor-associated nephrotoxicity: a position statement from the American Society of Onco-nephrology. Kidney Int 2025; 107:21-32. [PMID: 39455026 DOI: 10.1016/j.kint.2024.09.017] [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: 06/07/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/28/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer and are now the backbone of therapy for several malignancies. However, ICIs can cause a spectrum of kidney immune-related adverse events including acute kidney injury (AKI), most commonly manifesting as acute interstitial nephritis (AIN), although glomerular disease and electrolyte disturbances have also been reported. In this position statement by the American Society of Onco-nephrology (ASON), we summarize the incidence and risk factors for ICI-AKI, pathophysiological mechanisms, and clinicopathologic features of ICI-AKI. We also discuss novel diagnostic approaches and promising biomarkers for ICI-AKI. From expert panel consensus, we provide clinical practice points for the initial assessment and diagnosis of ICI-AKI, management and immunosuppressive therapy, and consideration for rechallenge with ICI following AKI episodes. In addition, we explore ICI use in special populations, such as kidney transplant recipients, and propose key areas of focus for future research and clinical investigation.
Collapse
Affiliation(s)
- Sandra M Herrmann
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA.
| | - Ala Abudayyeh
- Section of Nephrology, The University of Texas, MD Anderson Cancer Center, Houston, Texas, USA
| | - Shruti Gupta
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA; Department of Medical Oncology, Dana-Farber Cancer Institute. Harvard Medical School, Boston, Massachusetts, USA
| | | | - Nattawat Klomjit
- Department of Medicine, Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota, USA
| | - Shveta S Motwani
- Department of Medical Oncology, Dana-Farber Cancer Institute. Harvard Medical School, Boston, Massachusetts, USA
| | - Sabine Karam
- Department of Medicine, Division of Nephrology and Hypertension, University of Minnesota, Minneapolis, Minnesota, USA; Department of Internal Medicine, Division of Nephrology and Hypertension, American University of Beirut, Beirut, Lebanon
| | - Verônica T Costa E Silva
- Serviço de Nefrologia, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Laboratório de Investigação Médica (LIM) 16, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sheikh B Khalid
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Shuchi Anand
- Department of Medicine (Nephrology), Stanford University, Stanford, California, USA
| | - Jaya Kala
- Division of Renal Diseases and Hypertension, University of Texas Health Science Center at Houston-McGovern Medical School, Houston, Texas, USA
| | - David E Leaf
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Naoka Murakami
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA; Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Arash Rashidi
- Division of Nephrology and Hypertension, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
| | - Rimda Wanchoo
- Division of Kidney Diseases and Hypertension, Glomerular Center at Northwell Health, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, New York, USA
| | - Abhijat Kitchlu
- Department of Medicine, Division of Nephrology, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
3
|
Chintalacheruvu LM, Chilluru VK. Clinical Implications of Skin Cancer in Kidney Transplant Recipients in the Era of Immune Checkpoint Inhibitors. J Clin Med Res 2024; 16:571-577. [PMID: 39759492 PMCID: PMC11699868 DOI: 10.14740/jocmr6088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Long-term survival has improved in kidney transplant recipients (KTRs) due to effective surgical techniques and anti-rejection therapies. Chronic immunosuppression associated with it has led to several types of skin cancers leading to substantial morbidity and mortality. Structured patient education including sun protective behaviors, regular dermatological surveillance, nicotinamide, long-chain omega-3 polyunsaturated fatty acids (PUFAs), early switch to mammalian target of rapamycin inhibitors (mTORis), combining them with low-dose calcineurin inhibitors (CNIs), can decrease the cancer risk. Checkpoint inhibitors (CPIs) are the major backbone of the treatment of advanced skin cancers. Unfortunately, these agents can increase the risk of graft rejection. Prospective studies done so far looking at combining steroids with CPI in treatment of skin cancer in KTRs have shown mixed results. Adoption of the weight-based approach of CPI has shown to decrease the amount of drug exposure with acceptable outcomes in the general population, which is something that can be studied in KTRs with skin cancer. Also, it is reasonable to consider surveillance allograft biopsies in KTRs receiving CPIs to detect early subclinical rejection. More studies are needed to develop guidelines to safely treat this population with minimal graft rejection. We conducted a comprehensive literature review from PubMed on skin cancer in kidney transplant patients, focusing on incidence, risk factors, protective behaviors, financial and treatment implications, especially with regards to CPIs therapy. We also discussed potential newer treatment options that will decrease skin cancer risk, as well as graft rejection.
Collapse
Affiliation(s)
- Lakshmi Manogna Chintalacheruvu
- Hematology/Oncology, Southern Illinois Health Cancer Institute, Carterville, IL, USA
- Both authors contributed equally to this manuscript
| | - Vamsi Krishna Chilluru
- Nephrology, Southern Illinois Health, Carbondale, IL, USA
- Both authors contributed equally to this manuscript
| |
Collapse
|
4
|
Nicosia M, Valujskikh A. Recognizing Complexity of CD8 T Cells in Transplantation. Transplantation 2024; 108:2186-2196. [PMID: 38637929 PMCID: PMC11489323 DOI: 10.1097/tp.0000000000005001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
The major role of CD8 + T cells in clinical and experimental transplantation is well documented and acknowledged. Nevertheless, the precise impact of CD8 + T cells on graft tissue injury is not completely understood, thus impeding the development of specific treatment strategies. The goal of this overview is to consider the biology and functions of CD8 + T cells in the context of experimental and clinical allotransplantation, with special emphasis on how this cell subset is affected by currently available and emerging therapies.
Collapse
Affiliation(s)
- Michael Nicosia
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Anna Valujskikh
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| |
Collapse
|
5
|
Barbir EB, Abdulmoneim S, Dudek AZ, Kukla A. Immune Checkpoint Inhibitor Therapy for Kidney Transplant Recipients - A Review of Potential Complications and Management Strategies. Transpl Int 2024; 37:13322. [PMID: 39479217 PMCID: PMC11521864 DOI: 10.3389/ti.2024.13322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy has enabled a paradigm shift in Oncology, with the treatment of metastatic cancer in certain tumor types becoming akin to the treatment of chronic disease. Kidney transplant recipients (KTR) are at increased risk of developing cancer compared to the general population. Historically, KTR were excluded from ICI clinical trials due to concern for allograft rejection and decreased anti-tumor efficacy. While early post-marketing data revealed an allograft rejection risk of 40%-50%, 2 recent small prospective trials have demonstrated lower rates of rejection of 0%-12%, suggesting that maintenance immunosuppression modification prior to ICI start modulates rejection risk. Moreover, objective response rates induced by ICI for the treatment of advanced or metastatic skin cancer, the most common malignancy in KTR, have been comparable to those achieved by immune intact patients. Non-invasive biomarkers may have a role in risk-stratifying patients before starting ICI, and monitoring for rejection, though allograft biopsy is required to confirm diagnosis. This clinically focused review summarizes current knowledge on complications of ICI use in KTR, including their mechanism, risk mitigation strategies, non-invasive biomarker use, approaches to treatment of rejection, and suggestions for future directions in research.
Collapse
Affiliation(s)
- Elena Bianca Barbir
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| | | | - Arkadiusz Z. Dudek
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, United States
| | - Aleksandra Kukla
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
6
|
Khaddour K, Murakami N, Ruiz ES, Silk AW. Cutaneous Squamous Cell Carcinoma in Patients with Solid-Organ-Transplant-Associated Immunosuppression. Cancers (Basel) 2024; 16:3083. [PMID: 39272941 PMCID: PMC11394667 DOI: 10.3390/cancers16173083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/26/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
The management of advanced cutaneous squamous cell carcinoma (CSCC) has been revolutionized by the introduction of immunotherapy. Yet, successful treatment with immunotherapy relies on an adequate antitumor immune response. Patients who are solid-organ transplant recipients (SOTRs) have a higher incidence of CSCC compared to the general population. This review discusses the current knowledge of epidemiology, pathophysiology, and management of patients with CSCC who are immunocompromised because of their chronic exposure to immunosuppressive medications to prevent allograft rejection. First, we discuss the prognostic impact of immunosuppression in patients with CSCC. Next, we review the risk of CSCC development in immunosuppressed patients due to SOT. In addition, we provide an overview of the biological immune disruption present in transplanted immunosuppressed CSCC patients. We discuss the available evidence on the use of immunotherapy and provide a framework for the management approach with SOTRs with CSCC. Finally, we discuss potential novel approaches that are being investigated for the management of immunosuppressed patients with CSCC.
Collapse
Affiliation(s)
- Karam Khaddour
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| | - Naoka Murakami
- Harvard Medical School, Boston, MA 02115, USA
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Emily S Ruiz
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
- Department of Dermatology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ann W Silk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
7
|
Wang X, Zhang J, Zhong P, Wei X. Exhaustion of T cells after renal transplantation. Front Immunol 2024; 15:1418238. [PMID: 39165360 PMCID: PMC11333218 DOI: 10.3389/fimmu.2024.1418238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024] Open
Abstract
Renal transplantation is a life-saving treatment for patients with end-stage renal disease. However, the challenge of transplant rejection and the complications associated with immunosuppressants necessitates a deeper understanding of the underlying immune mechanisms. T cell exhaustion, a state characterized by impaired effector functions and sustained expression of inhibitory receptors, plays a dual role in renal transplantation. While moderate T cell exhaustion can aid in graft acceptance by regulating alloreactive T cell responses, excessive exhaustion may impair the recipient's ability to control viral infections and tumors, posing significant health risks. Moreover, drugs targeting T cell exhaustion to promote graft tolerance and using immune checkpoint inhibitors for cancer treatment in transplant recipients are areas deserving of further attention and research. This review aims to provide a comprehensive understanding of the changes in T cell exhaustion levels after renal transplantation and their implications for graft survival and patient outcomes. We discuss the molecular mechanisms underlying T cell exhaustion, the role of specific exhaustion markers, the potential impact of immunosuppressive therapies, and the pharmaceutical intervention on T cell exhaustion levels. Additionally, we demonstrate the potential to modulate T cell exhaustion favorably, enhancing graft survival. Future research should focus on the distinctions of T cell exhaustion across different immune states and subsets, as well as the interactions between exhausted T cells and other immune cells. Understanding these dynamics is crucial for optimizing transplant outcomes and ensuring long-term graft survival while maintaining immune competence.
Collapse
Affiliation(s)
- Xiujia Wang
- Department of 1st Urology Surgery, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jinghui Zhang
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Pingshan Zhong
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Xiuwang Wei
- Department of 1st Urology Surgery, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| |
Collapse
|
8
|
Hullegie-Peelen DM, Tejeda-Mora H, Dieterich M, Heidt S, Bindels EMJ, Hoogduijn MJ, Hesselink DA, Baan CC. Tissue-resident memory T cells in human kidney transplants have alloreactive potential. Am J Transplant 2024; 24:1406-1413. [PMID: 38447886 DOI: 10.1016/j.ajt.2024.02.030] [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: 01/15/2024] [Revised: 02/28/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
The extent to which tissue-resident memory T (TRM) cells in transplanted organs possess alloreactivity is uncertain. This study investigates the alloreactive potential of TRM cells in kidney explants from 4 patients who experienced severe acute rejection leading to graft loss. Alloreactive T cell receptor (TCR) clones were identified in pretransplant blood samples through mixed lymphocyte reactions, followed by single-cell RNA and TCR sequencing of the proliferated recipient T cells. Subsequently, these TCR clones were traced in the TRM cells of kidney explants, which were also subjected to single-cell RNA and TCR sequencing. The proportion of recipient-derived TRM cells expressing an alloreactive TCR in the 4 kidney explants varied from 0% to 9%. Notably, these alloreactive TCRs were predominantly found among CD4+ and CD8+ TRM cells with an effector phenotype. Intriguingly, these clones were present not only in recipient-derived TRM cells but also in donor-derived TRM cells, constituting up to 4% of the donor population, suggesting the presence of self-reactive TRM cells. Overall, our study demonstrates that T cells with alloreactive potential present in the peripheral blood prior to transplantation can infiltrate the kidney transplant and adopt a TRM phenotype.
Collapse
Affiliation(s)
- Daphne M Hullegie-Peelen
- Erasmus Medical Center Transplant Institute, Department of Internal Medicine, Nephrology and Transplantation, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Hector Tejeda-Mora
- Erasmus Medical Center Transplant Institute, Department of Internal Medicine, Nephrology and Transplantation, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Marjolein Dieterich
- Erasmus Medical Center Transplant Institute, Department of Internal Medicine, Nephrology and Transplantation, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Sebastiaan Heidt
- Department of Immunology, Leiden University Medical Center, Leiden, the Netherlands
| | - Eric M J Bindels
- Department of Haematology, University Medical Center, Rotterdam, the Netherlands
| | - Martin J Hoogduijn
- Erasmus Medical Center Transplant Institute, Department of Internal Medicine, Nephrology and Transplantation, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Erasmus Medical Center Transplant Institute, Department of Internal Medicine, Nephrology and Transplantation, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Carla C Baan
- Erasmus Medical Center Transplant Institute, Department of Internal Medicine, Nephrology and Transplantation, University Medical Center Rotterdam, Rotterdam, the Netherlands
| |
Collapse
|
9
|
Gao Y, Dong K, Gao Y, Jin X, Yang J, Yan G, Liu Q. Unified cross-modality integration and analysis of T cell receptors and T cell transcriptomes by low-resource-aware representation learning. CELL GENOMICS 2024; 4:100553. [PMID: 38688285 PMCID: PMC11099349 DOI: 10.1016/j.xgen.2024.100553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 03/09/2024] [Accepted: 04/06/2024] [Indexed: 05/02/2024]
Abstract
Single-cell RNA sequencing (scRNA-seq) and T cell receptor sequencing (TCR-seq) are pivotal for investigating T cell heterogeneity. Integrating these modalities, which is expected to uncover profound insights in immunology that might otherwise go unnoticed with a single modality, faces computational challenges due to the low-resource characteristics of the multimodal data. Herein, we present UniTCR, a novel low-resource-aware multimodal representation learning framework designed for the unified cross-modality integration, enabling comprehensive T cell analysis. By designing a dual-modality contrastive learning module and a single-modality preservation module to effectively embed each modality into a common latent space, UniTCR demonstrates versatility in connecting TCR sequences with T cell transcriptomes across various tasks, including single-modality analysis, modality gap analysis, epitope-TCR binding prediction, and TCR profile cross-modality generation, in a low-resource-aware way. Extensive evaluations conducted on multiple scRNA-seq/TCR-seq paired datasets showed the superior performance of UniTCR, exhibiting the ability of exploring the complexity of immune system.
Collapse
Affiliation(s)
- Yicheng Gao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Kejing Dong
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yuli Gao
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Xuan Jin
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Jingya Yang
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai 201804, China
| | - Gang Yan
- Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai 201804, China.
| | - Qi Liu
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration (Tongji University), Ministry of Education, Tongji Hospital, School of Medicine, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Shanghai Research Institute for Intelligent Autonomous Systems, Shanghai 201804, China; Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou 311121, China.
| |
Collapse
|
10
|
Van Meerhaeghe T, Murakami N, Le Moine A, Brouard S, Sprangers B, Degauque N. Fine-tuning tumor- and allo-immunity: advances in the use of immune checkpoint inhibitors in kidney transplant recipients. Clin Kidney J 2024; 17:sfae061. [PMID: 38606169 PMCID: PMC11008728 DOI: 10.1093/ckj/sfae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Indexed: 04/13/2024] Open
Abstract
Cancer is a common complication after kidney transplantation. Kidney transplant recipients (KTR) have a 2- to 4-fold higher risk of developing cancer compared to the general population and post-transplant malignancy is the third most common cause of death in KTR. Moreover, it is well known that certain cancer types are overrepresented after transplantation, especially non-melanoma skin cancer. Immune checkpoint inhibitors (ICI) have revolutionized the treatment of cancer, with remarkable survival benefit in a subgroup of patients. ICI are monoclonal antibodies that block the binding of specific co-inhibitory signaling molecules. Cytotoxic T lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1), and its ligand programmed cell death ligand 1 (PD-L1) are the main targets of ICI. Solid organ transplant recipients (SOTR) have been excluded from clinical trials owing to concerns about tumor response, allo-immunity, and risk of transplant rejection. Indeed, graft rejection has been estimated as high as 48% and represents an emerging problem. The underlying mechanisms of organ rejection in the context of treatment with ICI are poorly understood. The search for restricted antitumoral responses without graft rejection is of paramount importance. This review summarizes the current knowledge of the use of ICI in KTR, the potential mechanisms involved in kidney graft rejection during ICI treatment, potential biomarkers of rejection, and how to deal with rejection in clinical practice.
Collapse
Affiliation(s)
- Tess Van Meerhaeghe
- Departement of Nephrology, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
| | - Naoka Murakami
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, USA
- Harvard Medical School, Boston, USA
| | - Alain Le Moine
- Departement of Nephrology, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Sophie Brouard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
| | - Ben Sprangers
- Biomedical Research Institute, Department of Immunology and Infection, UHasselt, Diepenbeek, Belgium
- Department of Nephrology, Ziekenhuis Oost Limburg, Genk, Belgium
| | - Nicolas Degauque
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology (CR2TI), UMR 1064, Nantes, France
| |
Collapse
|
11
|
Hanna GJ, Dharanesswaran H, Giobbie-Hurder A, Harran JJ, Liao Z, Pai L, Tchekmedyian V, Ruiz ES, Waldman AH, Schmults CD, Riella LV, Lizotte P, Paweletz CP, Chandraker AK, Murakami N, Silk AW. Cemiplimab for Kidney Transplant Recipients With Advanced Cutaneous Squamous Cell Carcinoma. J Clin Oncol 2024; 42:1021-1030. [PMID: 38252908 PMCID: PMC10950183 DOI: 10.1200/jco.23.01498] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 08/24/2023] [Accepted: 10/03/2023] [Indexed: 01/24/2024] Open
Abstract
PURPOSE Cemiplimab is approved for treating locally advanced or metastatic cutaneous squamous cell carcinoma (CSCC). Solid organ transplant recipients have been excluded from immunotherapy trials, given concern for allograft rejection despite their increased risk of skin cancers. Chronic immunosuppression is necessary to prevent organ rejection but may attenuate antitumor response with PD-1 inhibitors. METHODS We report a phase I study of cemiplimab for kidney transplant recipients (KTRs) with advanced CSCC. After cross-taper to a mammalian target of rapamycin (mTOR) inhibitor and pulsed dose corticosteroids (prednisone 40 mg once daily, the day before and on days 1-3 of each cycle, followed by 20 mg once daily on days 4-6, then 10 mg once daily until the day before each subsequent cycle), patients received cemiplimab 350 mg intravenously once every 3 weeks for up to 2 years and were assessed for response every 8 weeks. The primary end point was the rate of kidney rejection, with key secondary end points including rate and duration of response, and survival. RESULTS Twelve patients were treated. No kidney rejection or loss was observed. A response to cemiplimab was observed in five of 11 evaluable patients (46%; 90% CI, 22 to 73), including two with durable responses beyond a year. Median follow-up was 6.8 months (range, 0.7-29.8). Treatment-related grade 3 or greater adverse events occurred in five patients (42%), including diarrhea, infection, and metabolic disturbances. One patient died of angioedema and anaphylaxis attributed to mTOR inhibitor cross-taper. CONCLUSION mTOR inhibitor and corticosteroids represent a favorable immunosuppressive regimen for KTRs with advanced CSCC receiving immunotherapy. This combination resulted in durable antitumor responses with no kidney rejection events (funded by Regeneron Pharmaceuticals [ClinicalTrials.gov identifier: NCT04339062]).
Collapse
Affiliation(s)
- Glenn J. Hanna
- Center for Head and Neck Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - John J. Harran
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Zixi Liao
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Lori Pai
- Department of Hematology/Oncology, Tufts Medical Center, Boston, MA
| | | | - Emily S. Ruiz
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | | | - Leonardo V. Riella
- Department of Medicine, Renal Division, Massachusetts General Hospital, Boston, MA
| | - Patrick Lizotte
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA
| | - Cloud P. Paweletz
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Naoka Murakami
- Division of Renal Medicine, Brigham and Women's Hospital, Boston, MA
| | - Ann W. Silk
- Center for Cutaneous Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
12
|
Li S, Zou D. Harnessing mixed chimerism and programmed cell death protein 1/programmed death ligand 1 signaling for transplant tolerance. Am J Transplant 2024; 24:147-148. [PMID: 37757911 DOI: 10.1016/j.ajt.2023.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Affiliation(s)
- Shuang Li
- Department of Cardiovascular Science, Houston Methodist Research Institute, Houston, Texas, USA
| | - Dawei Zou
- Immunobiology and Transplant Science Center, Department of Surgery, Houston Methodist Research Institute, Houston Methodist Hospital, Houston, Texas, USA.
| |
Collapse
|
13
|
Venkataraman K, Salehi T, Carroll RP. Immune Checkpoint Inhibitors in Recipients of Renal Allografts. Semin Nephrol 2024; 44:151500. [PMID: 38548484 DOI: 10.1016/j.semnephrol.2024.151500] [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] [Indexed: 06/30/2024]
Abstract
Kidney transplant recipients are at increased risk of malignancy as a result of immunosuppression and are increasingly exposed to checkpoint inhibitors (CPIs). However, CPI therapy can precipitate allograft rejection. This review aims to summarize the current literature describing the epidemiology, immunological mechanisms, diagnosis, and treatment of CPI-associated allograft rejection.Initial studies of CPIs suggested allograft rejection post commencement of CPIs occured commonly (40-60%), occurring between 2 and 6 weeks after CPI initiation, with a cancer response rate approaching 50%. More recent studies with predefined, structured immunosuppressive regimens have seen rejection rates of 0-12.5%, with rejection occurring later. Allograft biopsy remains the mainstay of diagnosis; however, noninvasive tools are emerging, including donor-derived cell-free DNA, urinary chemokine assessment, and defining alloreactive T-cell clones prior to or during CPI therapy.
Collapse
Affiliation(s)
- Karthik Venkataraman
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Tania Salehi
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia
| | - Robert P Carroll
- Central and Northern Adelaide Renal and Transplantation Service, Royal Adelaide Hospital, Adelaide, Australia; Australian Red Cross Lifeblood Service, Department of Health Sciences, University of South Australia, Adelaide, Australia.
| |
Collapse
|
14
|
Arai Y, Goto Y, Sazuka T, Fujimoto A, Sato H, Imamura Y, Sakamoto S, Ota M, Ikeda JI, Ichikawa T. A case of bladder cancer after bilateral lung transplantation following bone marrow transplantation. IJU Case Rep 2023; 6:471-474. [PMID: 37928309 PMCID: PMC10622205 DOI: 10.1002/iju5.12651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/20/2023] [Indexed: 11/07/2023] Open
Abstract
Introduction The incidence of bladder cancer following transplantation is high; however, no previous studies have reported the development of bladder cancer following bone marrow and bilateral lung transplantations. Case presentation A 42-year-old man who was followed for bilateral lung transplantation due to chronic graft-versus-host disease following bone marrow transplantation complained of gross hematuria. Transurethral resection of the bladder tumor was performed for cT1N0M0 bladder cancer. On the following night, he experienced severe respiratory failure and was intubated. He was discharged on postoperative day 32 with the introduction of home oxygen therapy. The pathological diagnosis was invasive urothelial carcinoma, high-grade, pT1, with urothelial carcinoma in situ. Further treatment could not be performed because of his poor performance status and immunosuppressive state. Conclusion Vigorous screening for bladder cancer coexisting with other malignancies should be performed for transplant recipients for the early diagnosis and prompt treatment of a relatively aggressive bladder cancer.
Collapse
Affiliation(s)
- Yutaro Arai
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Yusuke Goto
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Tomokazu Sazuka
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Ayumi Fujimoto
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Hiroaki Sato
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Yusuke Imamura
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Shinichi Sakamoto
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| | - Masayuki Ota
- Department of Diagnostic Pathology Chiba University Graduate School of Medicine Chiba Japan
| | - Jun-Ichiro Ikeda
- Department of Diagnostic Pathology Chiba University Graduate School of Medicine Chiba Japan
| | - Tomohiko Ichikawa
- Department of Urology Chiba University Graduate School of Medicine Chiba Japan
| |
Collapse
|
15
|
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
|
16
|
Allocco J, Alegre ML. Biological Time Travel: Tracking the Journey of Alloreactive T Cells Through Longitudinal Biobanking. Transplantation 2023; 107:1862-1864. [PMID: 37606904 PMCID: PMC10926360 DOI: 10.1097/tp.0000000000004684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/23/2023]
Abstract
A significant risk for transplant recipients is the development of tumors. In general, some but not all malignancies are more frequent in transplant hosts due to chronic immunosuppression caused by a compromised immune surveillance. Of additional relevance, checkpoint blockade therapies (CBT) to treat malignancies can also drive transplant rejection. In a recent study published in Nature Communications , Dunlap et al. reported a case study of a patient who experienced kidney allograft rejection following CBT for melanoma. The foresight of longitudinally preserving donor splenocytes, blood samples, and graft biopsies in addition to tumor and metastatic lymph nodes enabled paired single-cell RNA-seq (scRNA-seq) and TCR-sequencing (TCR-seq) and subsequent tracking of alloreactive T cells before and after CBT. This revealed an enrichment of alloreactive TCRs in the kidney transplant post-CBT but not the tumor. In addition, this approach helped identify an alloreactive CD8+ T cell subset with a unique transcriptional profile. This study illustrates possible advances in personalized medicine and highlights a transcriptional signature that may serve as a prospective biomarker of rejection.
Collapse
Affiliation(s)
- Jennifer Allocco
- Department of Medicine, University of Chicago, Chicago, IL 60637
| | | |
Collapse
|