1
|
McCallion O, Cross AR, Brook MO, Hennessy C, Ferreira R, Trzupek D, Mulley WR, Kumar S, Soares M, Roberts IS, Friend PJ, Lombardi G, Wood KJ, Harden PN, Hester J, Issa F. Regulatory T cell therapy is associated with distinct immune regulatory lymphocytic infiltrates in kidney transplants. MED 2025; 6:100561. [PMID: 39731908 DOI: 10.1016/j.medj.2024.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/17/2024] [Accepted: 11/25/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND Adoptive transfer of autologous regulatory T cells (Tregs) is a promising therapeutic strategy aimed at enabling immunosuppression minimization following kidney transplantation. In our phase 1 clinical trial of Treg therapy in living donor renal transplantation, the ONE Study (ClinicalTrials.gov: NCT02129881), we observed focal lymphocytic infiltrates in protocol kidney transplant biopsies that are not regularly seen in biopsies of patients receiving standard immunosuppression. METHODS We present 7 years of follow-up data on patients treated with adoptive Treg therapy early post-transplantation who exhibited focal lymphocytic infiltrates on a 9-month protocol biopsy. We phenotyped their adoptively transferred and peripherally circulating Treg compartments using CITE-seq and investigated the focal lymphocytic infiltrates with spatial proteomic and transcriptomic technologies. FINDINGS Graft survival rates were not significantly different between Treg-treated patients and the control reference group. None of the Treg-treated patients experienced clinical rejection episodes or developed de novo donor-specific antibodies, and three of ten successfully reduced their immunosuppression to tacrolimus monotherapy. All Treg-treated patients who underwent a protocol biopsy 9 months post-transplantation exhibited focal lymphocytic infiltrates. Spatial profiling analysis revealed prominent CD20+ B cell and regulatory (IKZF2, IL10, PD-L1, TIGIT) signatures within cell-therapy-associated immune infiltrates, distinct from the pro-inflammatory myeloid signature associated with rejection biopsies. CONCLUSIONS We demonstrate for the first time that immune cell infiltrates in transplanted kidneys can occur following adoptive Treg therapy in humans, potentially facilitating within-graft T:B cell interactions that promote local immune regulation. FUNDING This work was funded by the 7th EU Framework Programme, grant/award no. 260687, and the National Institute for Health Research (NIHR).
Collapse
Affiliation(s)
- Oliver McCallion
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Amy R Cross
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Matthew O Brook
- Department of Renal Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LH, UK
| | - Conor Hennessy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Ricardo Ferreira
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - Dominik Trzupek
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - William R Mulley
- Department of Nephrology, Monash Medical Centre & Department of Medicine, Monash University, Clayton, VIC 3168, Australia
| | - Sandeep Kumar
- Advanced Therapy Manufacturing (GMP) Unit, Guy's & St Thomas' NHS Foundation Trust and King's College London, London SE1 9RT, UK
| | - Maria Soares
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Ian S Roberts
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 9DU, UK
| | - Peter J Friend
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, King's College London, London SE1 9RT, UK
| | - Kathryn J Wood
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Paul N Harden
- Department of Renal Medicine, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LH, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 9DU, UK.
| |
Collapse
|
2
|
Musalem P, Sáez-Vera C. Forty years of kidney transplantation: Insights into malignancies at a single center in Latin America. Transpl Immunol 2025; 90:102216. [PMID: 40120985 DOI: 10.1016/j.trim.2025.102216] [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: 10/15/2024] [Revised: 03/03/2025] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Kidney transplantation is the optimal therapy for end-stage kidney disease (ESKD), but lifelong immunosuppression increases malignancy risk, a major cause of mortality in transplant recipients. This study evaluates post-transplant malignancies in a kidney transplant cohort. METHODS We conducted a retrospective analysis of 375 kidney transplant recipients at Hospital Las Higueras, Talcahuano (January 1981-July 2024). Demographics, clinical characteristics, and malignancy data were extracted from medical records. RESULTS Of 375 patients, 33 (8.8 %) developed malignancies, with 27 % experiencing multiple cancers. While the mean age at transplantation was 51.5 years, the mean age of those developing a malignancy was 60.4 years. Non-melanoma skin cancers were most common (55.1 %), followed by solid organ cancers (26.5 %), chronic kidney disease (CKD)-related cancers (8.2 %), and hematologic malignancies (8.2 %). The mean time to malignancy onset was 106 months post-transplant. Cancer-related mortality was 30 %. CONCLUSION The high incidence of malignancies, particularly skin cancers, highlights the need for regular clinical and dermatologic surveillance in transplant recipients. Optimizing immunosuppression to balance rejection prevention and cancer risk, along with comprehensive cancer screening, is essential for improving long-term outcomes.
Collapse
Affiliation(s)
- Pilar Musalem
- Nephrology, Dialysis and Transplantation Service, Hospital Las Higueras, Talcahuano, Chile.; Departamento de Medicina Interna, Facultad de Medicina, Universidad de Concepción, Concepción, Chile..
| | - Carolina Sáez-Vera
- Nephrology, Dialysis and Transplantation Service, Hospital Las Higueras, Talcahuano, Chile.; Departamento de Medicina Interna, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
3
|
Avalos-de Leon CG, Thomson AW. Regulatory Immune Cell-derived Exosomes: Modes of Action and Therapeutic Potential in Transplantation. Transplantation 2025:00007890-990000000-00994. [PMID: 39865513 DOI: 10.1097/tp.0000000000005309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Reduced dependence on antirejection agents, improved long-term allograft survival, and induction of operational tolerance remain major unmet needs in organ transplantation due to the limitations of current immunosuppressive therapies. To address this challenge, investigators are exploring the therapeutic potential of adoptively transferred host- or donor-derived regulatory immune cells. Extracellular vesicles of endosomal origin (exosomes) secreted by these cells seem to be important contributors to their immunoregulatory properties. Twenty years ago, it was first reported that donor-derived exosomes could extend the survival of transplanted organs in rodents. Recent studies have revealed that regulatory immune cells, such as regulatory myeloid cells (dendritic cells, macrophages, or myeloid-derived suppressor cells), regulatory T cells, or mesenchymal stem/stromal cells can suppress graft rejection via exosomes that express a cargo of immunosuppressive molecules. These include cell surface molecules that interact with adaptive immune cell receptors, immunoregulatory enzymes, and micro- and long noncoding RNAs that can regulate inflammatory gene expression via posttranscriptional changes and promote tolerance through promotion of regulatory T cells. This overview analyzes the diverse molecules and mechanisms that enable regulatory immune cell-derived exosomes to modulate alloimmunity and promote experimental transplant tolerance. We also discuss the potential benefits and limitations of their application as therapeutic entities in organ transplantation.
Collapse
Affiliation(s)
- Cindy G Avalos-de Leon
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
| | - Angus W Thomson
- Department of Surgery, Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh PA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh PA
| |
Collapse
|
4
|
Zeng M, Lin A, Jiang A, Qiu Z, Zhang H, Chen S, Xu M, Liu Z, Cheng Q, Zhang J, Luo P. Decoding the mechanisms behind second primary cancers. J Transl Med 2025; 23:115. [PMID: 39856672 PMCID: PMC11762917 DOI: 10.1186/s12967-025-06151-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025] Open
Abstract
Second Primary Cancers (SPCs) are defined as cancers that develop either simultaneously or metachronously in the same individual who has been diagnosed with and survived one primary cancer. SPCs exhibit a high incidence rate and represent the primary cause of mortality among survivors of first primary cancers. There is growing concern about the dangers of SPCs. This review summarizes recent studies on the mechanisms of SPCs, including the roles of genomic changes after first primary cancer (FPC) treatments, stromal cell phenotypic and metabolic changes, hormone levels and receptor expression, immunosuppression, aberrant gene methylation, EGFR signaling, and cell-free DNA in SPC development. This comprehensive analysis contributes to elucidating current research trends in SPC mechanisms and enhances our understanding of the underlying pathophysiology. Furthermore, potential applications of intratumoral microbes, single-cell multi-omics, and metabolomics in investigating SPC mechanisms are also discussed, providing new ideas for follow-up studies.
Collapse
Affiliation(s)
- Meiyuan Zeng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhengang Qiu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hongman Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Shifu Chen
- HaploX Biotechnology, Shenzhen, China
- Faculty of Data Science, City University of Macau, Macau, China
| | | | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China.
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
5
|
Kreuz M, Cardoso JCO, Sobreira LER, Cavalcanti Souza ME, Campos LE, Kelly FA, de Moraes FCA. Azathioprine and risk of non-melanoma skin cancers in organ transplant recipients: a systematic review and update meta-analysis. Clin Transl Oncol 2025:10.1007/s12094-024-03839-0. [PMID: 39825996 DOI: 10.1007/s12094-024-03839-0] [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: 08/21/2024] [Accepted: 12/24/2024] [Indexed: 01/20/2025]
Abstract
BACKGROUND Immunosuppression might increase the risk of skin cancer in organ transplant recipients (OTRs), with azathioprine (AZA), exerting a fundamental role in the carcinogenesis of those tumors. This systematic review and meta-analysis aims to address the risk of developing malignant skin neoplasms in OTRs undergoing immunosuppression with AZA. METHODS PubMed, Cochrane and Embase were searched for studies with OTRs who have a treatment regimen involving Azathioprine therapy after transplantation and that analyzed the emergence of skin neoplasia. We performed the meta-analysis using RStudio v4.4.2 software. RESULTS A total of 17 studies comprising a total of 12,708 patients were included, of whom 3567 (28,06%) had a treatment regimen involving AZA therapy after transplantation. The majority of individuals were male 7298 (56,52%) and the median age of patients ranged from 41.5 to 63.2 years. The overall summary estimate showed a significantly increased risk of all types of skin cancer in relation to AZA exposure (OR 1.55; 95% CI 1.07-2.25; p = 0.018; I2 = 82%). These results show that the overall result is statistically significant, which means that the observed effect is unlikely to be caused by chance. CONCLUSION This study highlights the increased risk of developing skin cancer, particularly squamous cell carcinoma (SCC), in OTRs receiving immunosuppressive therapy with AZA, which allows for rigorous screening and appropriate preventive and therapeutic interventions.
Collapse
Affiliation(s)
- Michele Kreuz
- Lutheran University of Brazil, 133 Montenegro Street, ap 1201, Novo Hamburgo, Canoas, Rio Grande do Sul, 92425-900, Brazil.
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil.
| | - Jorge Cavalcanti Orestes Cardoso
- University of Pernambuco, Recife, Pernambuco, 50100-130, Brazil
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil
| | - Luis Eduardo Rodrigues Sobreira
- Federal University of Pará, Altamira, Pará, 68371-040, Brazil
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil
| | - Maria Eduarda Cavalcanti Souza
- University of Pernambuco, Recife, Pernambuco, 50100-130, Brazil
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil
| | - Lara Eduardo Campos
- School of Medicine and Surgery of the Federal University of the State of Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, 20271-062, Brazil
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil
| | - Francinny Alves Kelly
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil
- Dante Pazzanese Institute of Cardiology, São Paulo, São Paulo, 04012-909, Brazil
| | - Francisco Cezar Aquino de Moraes
- Anhembi University Morumbi, São José dos Campos, São Paulo, 12235-181, Brazil
- Federal University of Pará, Belém, 66073-005, Brazil
| |
Collapse
|
6
|
Watanabe H, Kadomatsu Y, Hakiri S, Yoshioka H, Hiramitsu T, Futamura K, Okada M, Goto N, Narumi S, Watarai Y, Chen-Yoshikawa TF. Lung cancer after kidney transplantation: a 50-year experience at a single institution. Surg Today 2024; 54:1124-1130. [PMID: 38546862 DOI: 10.1007/s00595-024-02819-9] [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: 11/17/2023] [Accepted: 01/22/2024] [Indexed: 09/21/2024]
Abstract
PURPOSE To investigate the clinical characteristics of lung cancer that develops after kidney transplantation. METHODS The clinical data of patients with lung cancer diagnosed after kidney transplantation were collected retrospectively. The medical records were extracted from our database. All patients underwent routine chest examination after kidney transplantation. RESULTS In total, 17 lung tumors were detected in 15 (0.6%) of 2593 patients who underwent kidney transplantation at our institution. Eleven lung tumors were completely resected from a collective 10 patients (surgical group). The remaining five patients did not receive surgical treatment (nonsurgical group). The surgical group underwent wedge resection (n = 5), segmentectomy (n = 1), lobectomy (n = 3), and bilobectomy (n = 1). The pathological stages were 0 (n = 1), IA1 (n = 2), IA2 (n = 4), IA3 (n = 2), and IB (n = 1). The surgical group had a significantly better prognosis than the nonsurgical group. There were no perioperative complications related to kidney transplantation in either group. CONCLUSIONS Routine chest examination would be useful for the early diagnosis and treatment of lung cancer after kidney transplantation. Moreover, surgical resection for early-stage lung cancer was associated with a better prognosis for kidney transplantation patients.
Collapse
Affiliation(s)
- Hiroki Watanabe
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan.
| | - Yuka Kadomatsu
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| | - Shuhei Hakiri
- Department of Thoracic Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, 2-9 Myoken-Cho, Syowa-Ku, Nagoya, 466-8650, Japan
| | - Hiromu Yoshioka
- Department of Thoracic Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, 2-9 Myoken-Cho, Syowa-Ku, Nagoya, 466-8650, Japan
| | - Takahisa Hiramitsu
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Kenta Futamura
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Manabu Okada
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Norihiko Goto
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Shunji Narumi
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Yoshihiko Watarai
- Department of Transplant and Endocrine Surgery, Japanese Red Cross Aichi Medical Center Nagoya Daini Hospital, Nagoya, Japan
| | - Toyofumi Fengshi Chen-Yoshikawa
- Department of Thoracic Surgery, Nagoya University Graduate School of Medicine, 65 Tsurumai-Cho, Showa-Ku, Nagoya, 466-8550, Japan
| |
Collapse
|
7
|
Purnomo AF, Nurkolis F, Syahputra RA, Moon S, Lee D, Taslim NA, Park MN, Daryanto B, Seputra KP, Satyagraha P, Lutfiana NC, Wisnu Tirtayasa PM, Kim B. Elucidating the nexus between onco-immunology and kidney transplantation: An insight from precision medicine perspective. Heliyon 2024; 10:e33751. [PMID: 39040404 PMCID: PMC11261886 DOI: 10.1016/j.heliyon.2024.e33751] [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: 04/04/2024] [Revised: 06/12/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024] Open
Abstract
The interplay of onco-immunology and kidney transplantation heralds a transformative era in medical science. This integration, while promising, presents significant challenges. Chief among these is the dichotomy of immunosuppression-boosting immunity against malignancies while suppressing it for graft survival. Additionally, limited clinical data on novel therapies, genetic variations influencing responses, economic concerns, and the narrow therapeutic window for post-transplant malignancies necessitate strategic addressal. Conversely, opportunities abound, including personalized immune monitoring, targeted therapies, minimized immunosuppression, and improved patient quality of life. Emphasizing collaborative research and interdisciplinary cooperation, the merging of these fields offers the potential for enhanced graft survival and reduced post-transplant malignancy risks. As we harness modern technology and promote patient-centric care, the vision for the future of kidney transplantation becomes increasingly hopeful, paving the way for more personalized and effective treatments. The article aims to elucidate the critical challenge of balancing immunosuppression to simultaneously combat malignancies and ensure graft survival. It addresses the scarcity of clinical data on novel therapies, the impact of genetic variations on treatment responses, and the economic and therapeutic concerns in managing post-transplant malignancies. Furthermore, it explores the opportunities precision medicine offers, such as personalized immune monitoring, targeted therapies, and reduced immunosuppression, which could significantly improve patient outcomes. Highlighting the importance of collaborative research and interdisciplinary efforts, the article seeks to demonstrate the potential for enhanced graft survival and reduced post-transplant malignancy risks. By leveraging modern technology and prioritizing patient-centric care, it envisions a future where kidney transplantation is more personalized and effective, offering hope for advancements in this field.
Collapse
Affiliation(s)
- Athaya Febriantyo Purnomo
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, United Kingdom
- Department of Urology, Faculty of Medicine Universitas Brawijaya–Saiful Anwar General Hospital, Malang, 65142, Indonesia
| | - Fahrul Nurkolis
- Department of Biological Sciences, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, 55281, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Seungjoon Moon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Chansol Hospital of Korean Medicine, 290, Buheung-ro, Bupyeong-gu, Incheon, South Korea, 21390, Republic of Korea
| | - Dain Lee
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Nurpudji Astuti Taslim
- Division of Clinical Nutrition, Department of Nutrition, Faculty of Medicine, Hasanuddin University, Makassar, 90245, Indonesia
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Besut Daryanto
- Department of Urology, Faculty of Medicine Universitas Brawijaya–Saiful Anwar General Hospital, Malang, 65142, Indonesia
| | - Kurnia Penta Seputra
- Department of Urology, Faculty of Medicine Universitas Brawijaya–Saiful Anwar General Hospital, Malang, 65142, Indonesia
| | - Paksi Satyagraha
- Department of Urology, Faculty of Medicine Universitas Brawijaya–Saiful Anwar General Hospital, Malang, 65142, Indonesia
| | - Nurul Cholifah Lutfiana
- Department of Biochemistry and Biomedicine, Faculty of Medicine, Universitas Muhammadiyah Surabaya, Surabaya, Indonesia
| | - Pande Made Wisnu Tirtayasa
- Department of Urology, Faculty of Medicine, Universitas Udayana, Universitas Udayana Teaching Hospital, Bali, 80361, Indonesia
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemun-gu, Seoul, 02447, Republic of Korea
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
8
|
Singh Z, Holt SK, Gore JL, Nyame YA, Wright JL, Schade GR. Chronic Glucocorticoid Use and Risk for Advanced Prostate Cancer at Presentation: A SEER-Medicare Cohort Study. Clin Genitourin Cancer 2024; 22:68-73.e2. [PMID: 37806926 DOI: 10.1016/j.clgc.2023.08.007] [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: 05/13/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Examine the relationship between exposure to systemic glucocorticoids (steroids) and advanced prostate cancer (PCa) at presentation. Prior work suggested that steroid use may be associated with increased PCa risk. MATERIALS AND METHODS We queried the linked SEER-Medicare database (2004-2015) to identify PSA screened patients diagnosed with PCa. Criteria for screening included a PSA lab test or DRE exam in both the 12 month and 13 to 36 month periods prior to diagnosis of PCa. Steroid exposure was determined using Medicare Part D and groups were divided based on duration of use in the 3 years prior to diagnosis: controls with no exposure, <30 days, 30 days - 1 year, 1 to 2 years, and >2+ years. Advanced PCa was defined as systemic metastases or regional lymph node metastasis at presentation. Risk estimates for advanced PCa at presentation for steroid exposure groups vs. controls were assessed with univariable and multivariable logistic regression models. RESULTS We identified 22,920 PSA screened patients diagnosed with PCa of which 29% used glucocorticoids in the exposure period. The mean (SD) duration for glucocorticoid use (in days) among all steroid users was 76.7 days (192.1). On univariable and multivariable analyses, > 2 years of steroid exposure was associated with significantly increased risk for advanced PCa (OR 2.06, 95% CI 1.35-3.14 and OR 1.74, 95% CI 1.12-2.69, respectively). CONCLUSION In this population-based PSA-screened cohort, prolonged steroid use was associated with increased risk of advanced PCa at diagnosis. With the widespread use of glucocorticoids, it is important to consider the role steroids may play in PCa pathogenesis.
Collapse
Affiliation(s)
- Zorawar Singh
- Department of Urology, University of Washington Medical Center, Seattle, WA; Smith Institute for Urology at Northwell Health of the Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, New York, NY
| | - Sarah K Holt
- Department of Urology, University of Washington Medical Center, Seattle, WA
| | - John L Gore
- Department of Urology, University of Washington Medical Center, Seattle, WA
| | - Yaw A Nyame
- Department of Urology, University of Washington Medical Center, Seattle, WA
| | - Jonathan L Wright
- Department of Urology, University of Washington Medical Center, Seattle, WA
| | - George R Schade
- Department of Urology, University of Washington Medical Center, Seattle, WA.
| |
Collapse
|
9
|
Wei C, Sun Y, Zeng F, Chen X, Ma L, Liu X, Qi X, Shi W, Gao H. Exosomal miR-181d-5p Derived from Rapamycin-Conditioned MDSC Alleviated Allograft Rejection by Targeting KLF6. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2304922. [PMID: 37870185 PMCID: PMC10700181 DOI: 10.1002/advs.202304922] [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: 07/19/2023] [Revised: 09/18/2023] [Indexed: 10/24/2023]
Abstract
Immune rejection and side effects of long-term administration of immunosuppressants are the two major obstacles to allograft acceptance and tolerance. The immunosuppressive extracellular vesicles (EVs)-based approach has been proven to be effective in treating autoimmune/inflammatory disorders. Herein, the anti-rejection advantage of exosomes (Rapa-Exo) from rapamycin-conditioned myeloid-derived suppressor cells (MDSCs) over exosomes (Exo-Nor) from the untreated MDSCs is shown. The exosomal small RNA sequencing and loss-of-function assays reveal that the anti-rejection effect of Rapa-Exo functionally relies on miR-181d-5p. Through target prediction and double-luciferase reporter assay, Kruppel-like factor (KLF) 6 is identified as a direct target of miR-181d-5p. Finally, KLF6 knockdown markedly resolves inflammation and prolongs the survival of corneal allografts. Taken together, these findings support that Rapa-Exo executes an anti-rejection effect, highlighting the immunosuppressive EVs-based treatment as a promising approach in organ transplantation.
Collapse
Affiliation(s)
- Chao Wei
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Yaru Sun
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Fanxing Zeng
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Xiunian Chen
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Li Ma
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Xiaoxue Liu
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
| | - Xiaolin Qi
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan250117China
- School of OphthalmologyShandong First Medical University & Shandong Academy of Medical ScienceJinan250117China
| | - Weiyun Shi
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan250117China
- School of OphthalmologyShandong First Medical University & Shandong Academy of Medical ScienceJinan250117China
| | - Hua Gao
- State Key Laboratory Cultivation BaseShandong Provincial Key Laboratory of OphthalmologyEye Institute of Shandong First Medical UniversityQingdao266071China
- Eye Hospital of Shandong First Medical University (Shandong Eye Hospital)Jinan250117China
- School of OphthalmologyShandong First Medical University & Shandong Academy of Medical ScienceJinan250117China
| |
Collapse
|
10
|
Li D, Liu F, Chen Y, Li P, Liu Y, Pang Y. Ipsilateral synchronous papillary renal neoplasm with reverse polarity and urothelial carcinoma in a renal transplant recipient: a rare case report with molecular analysis and literature review. Diagn Pathol 2023; 18:120. [PMID: 37924117 PMCID: PMC10623754 DOI: 10.1186/s13000-023-01405-w] [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: 01/31/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Renal transplant recipients (RTRs) have a 3- to 5-fold higher risk of developing malignant tumors than the general population, with new malignant tumors after transplantation considered to be the leading cause of death in RTRs. In pathological practice, it is rare for neoplasms with different histology to be located in the same organ. We report the first case of a synchronous papillary renal neoplasm with reverse polarity (PRNRP) and urothelial carcinoma (UC) in the ipsilateral kidney in an RTR. Molecular detection was conducted by next-generation sequencing. CASE PRESENTATION A 68-year-old female suffered from uremia 19 years ago and underwent renal transplantation (RT) after receiving dialysis for 6 months. Hematuria occurred one month ago and an enhanced CT showed that there were two abnormal density foci in the middle and lower parts of the autologous left kidney. A laparoscopic left nephrectomy and ureterectomy were performed. Gross examination revealed a mass (I) in the left renal parenchyma, 2*1.8*1.5 cm in size, that protruded from the renal capsule, and a cauliflower-like mass (II), 5*2.5*2 cm in size, adjacent to the mass (I). Microscopic findings revealed these lesions were PRNRP and UC, respectively. PCR analysis revealed a KRAS gene mutation (G12D in exon 2) in the PRNRP, while NGS analysis revealed FGFR3 (S249C in exon 7) and KDM6A (Q271Ter in exon 10 and A782Lfs in exon 17) mutations in the UC. CONCLUSIONS We report here for the first time an extraordinarily rare case of synchronous renal tumors of a PRNRP and UC in the ipsilateral kidney of an RTR. We identified simultaneous KRAS, FGFR3, and KDM6A mutations in two different renal masses in the ipsilateral kidney. Pathologic assessment with comparative molecular analysis of mutational profiles facilitates tumor studies after RT and may be of great value in clinical management strategies.
Collapse
Affiliation(s)
- Daosheng Li
- Department of Pathology, the Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Fenfen Liu
- Department of Urology, the Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Yiqian Chen
- Department of Rehabilitation, the Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Ping Li
- Department of Pathology, the Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Yuyu Liu
- Department of Hematology, the Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China
| | - Yu Pang
- Department of Pathology, the Affiliated Tai'an City Central Hospital of Qingdao University, Tai'an, 271000, China.
| |
Collapse
|
11
|
Yohannan B, Sridhar A, Kaur H, DeGolovine A, Maithel N. Screening for renal cell carcinoma in renal transplant recipients: a single-centre retrospective study. BMJ Open 2023; 13:e071658. [PMID: 37699639 PMCID: PMC10503370 DOI: 10.1136/bmjopen-2023-071658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 08/24/2023] [Indexed: 09/14/2023] Open
Abstract
OBJECTIVES The primary objective of our study was to evaluate the effectiveness of renal cell carcinoma (RCC) screening in renal transplant (RT) recipients. DESIGN Single-centre retrospective study. SETTING AND PARTICIPANTS 1998 RT recipients who underwent RT at Memorial Hermann Hospital (MHH) Texas Medical Center (TMC) between 1 January 1999 and 31 December 2019 were included and we identified 16 patients (0.8%) with RCC. An additional four patients with RCC who underwent RT elsewhere but received follow-up at MHH TMC were also included. Subject races included white (20%), black (50%), Hispanic (20%) and Asian (10%). OUTCOME MEASURES The RCC stage at diagnosis and outcomes were compared between patients who were screening versus those who were not. RESULTS We identified a total of 20 patients with post-RT RCC, 75% of whom were men. The median age at diagnosis was 56 years. RCC histologies included clear cell (75%), papillary (20%) and chromophobe (5%). Patients with post-RT RCC who had screening (n=12) underwent ultrasound or CT annually or every 2 years, whereas eight patients had no screening. All 12 patients who had screening had early-stage disease at diagnosis (stage I (n=11) or stage II (n=1)) and were cured by nephrectomy (n=10) or cryotherapy (n=2). In patients who had no screening, three (37.5%) had stage IV RCC at diagnosis and all of whom died of metastatic disease. There was a statistically significant difference in RCC-specific survival in patients who were screened (p=0.01) compared with those who were not screened. CONCLUSION All RT recipients who had RCC diagnosed based on screening had early-stage disease and there were no RCC-related deaths. Screening is an effective intervention in RT recipients to reduce RCC-related mortality.
Collapse
Affiliation(s)
- Binoy Yohannan
- Department of Hematology and Oncology, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Arthi Sridhar
- Department of Hematology and Oncology, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Harmanpreet Kaur
- Department of Internal Medicine, UT Southwestern Medical School, Dallas, Texas, USA
| | - Aleksandra DeGolovine
- Department of Renal Disease and Hypertension, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| | - Neha Maithel
- Department of Hematology and Oncology, The University of Texas Health Sciences Center at Houston, Houston, Texas, USA
| |
Collapse
|
12
|
Mayrdorfer M, Liefeldt L, Osmanodja B, Naik MG, Schmidt D, Duettmann W, Hammett C, Schrezenmeier E, Friedersdorff F, Wu K, Halleck F, Budde K. A single centre in-depth analysis of death with a functioning kidney graft and reasons for overall graft failure. Nephrol Dial Transplant 2023; 38:1857-1866. [PMID: 36477607 PMCID: PMC10387383 DOI: 10.1093/ndt/gfac327] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND High numbers of unknown classifications and inconsistent methodologies in previous studies make the interpretation of causes leading to graft loss difficult. In addition, data on a holistic view looking at both death with a functioning graft (DWFG) and death-censored graft failure (DCGF) are sparse. METHODS In this single-centre study we included 1477 adult kidney transplants performed between 1997 and 2017, of which all 286 DWFGs until the end of observation were analysed and causes for death assigned. Additionally, the results were compared with the causes of 303 DCGFs of the same cohort to evaluate the impact of causes for overall graft loss. RESULTS The most frequent causes for DWFG were cardiovascular disease (CVD) in 30.8%, malignancy in 28.3% and infections in 21%. Only 9.4% of reasons for DWFG were unknown. Sudden death occurred in 40% (35/88) of patients classified as DWFG due to CVD. Overall graft loss was related to the effect of immunosuppression in 36.2% [infection 20.9% (123/589), malignancy 15.3% (90/589)] and CVD in 22.4% (132/589). In 27.4% (161/589), graft failure was associated with underimmunosuppression (rejection). For infections (60 DWFG, 63 DCGF) and CVD (88 DWFG, 44 DCGF), a considerable overlap was observed between DWFG and DCGF. For patients >70 years of age at transplantation, medical events accounted for 78% of overall graft losses and only 6.5% were associated with rejection. CONCLUSIONS DWFG and DCGF share more causes for graft loss than previously reported and sudden death plays an underestimated role in death with a functioning graft.
Collapse
Affiliation(s)
- Manuel Mayrdorfer
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Division of Nephrology and Dialysis, Department of Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, Austria
| | - Lutz Liefeldt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Bilgin Osmanodja
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Marcel G Naik
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Danilo Schmidt
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Wiebke Duettmann
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Charlotte Hammett
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eva Schrezenmeier
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Frank Friedersdorff
- Department of Urology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kaiyin Wu
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Fabian Halleck
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Klemens Budde
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
13
|
Mazzio E, Barnes A, Badisa R, Council S, Soliman KFA. Plants against cancer: the immune-boosting herbal microbiome: not of the plant, but in the plant. Basic concepts, introduction, and future resource for vaccine adjuvant discovery. Front Oncol 2023; 13:1180084. [PMID: 37588095 PMCID: PMC10426289 DOI: 10.3389/fonc.2023.1180084] [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: 03/05/2023] [Accepted: 05/30/2023] [Indexed: 08/18/2023] Open
Abstract
The presence of microorganism communities (MOCs) comprised of bacteria, fungi, archaea, algae, protozoa, viruses, and the like, are ubiquitous in all living tissue, including plant and animal. MOCs play a significant role in establishing innate and acquired immunity, thereby influencing susceptibility and resistance to disease. This understanding has fostered substantial advancements in several fields such as agriculture, food science/safety, and the development of vaccines/adjuvants, which rely on administering inactivated-attenuated MOC pathogens. Historical evidence dating back to the 1800s, including reports by Drs Busch, Coley, and Fehleisen, suggested that acute febrile infection in response to "specific microbes" could trigger spontaneous tumor remission in humans. This discovery led to the purposeful administration of the same attenuated strains, known as "Coley's toxin," marking the onset of the first microbial (pathogen) associated molecular pattern (MAMPs or PAMPs)-based tumor immunotherapy, used clinically for over four decades. Today, these same MAMPS are consumed orally by billions of consumers around the globe, through "specific" mediums (immune boosting "herbal supplements") as carriers of highly concentrated MOCs accrued in roots, barks, hulls, sea algae, and seeds. The American Herbal Products Association (AHPA) mandates microbial reduction in botanical product processing but does not necessitate the removal of dead MAMP laden microbial debris, which we ingest. Moreover, while existing research has focused on the immune-modulating role of plant phytochemicals, the actual immune-boosting properties might instead reside solely in the plant's MOC MAMP laden biomass. This assertion is logical, considering that antigenic immune-provoking epitopes, not phytochemicals, are known to stimulate immune response. This review explores a neglected area of research regarding the immune-boosting effects of the herbal microbiome - a presence which is indirectly corroborated by various peripheral fields of study and poses a fundamental question: Given that food safety focuses on the elimination of harmful pathogens and crop science acknowledges the existence of plant microbiomes, what precisely are the immune effects of ingesting MAMPs of diverse structural composition and concentration, and where are these distributed in our botanicals? We will discuss the topic of concentrated edible MAMPs as acid and thermally stable motifs found in specific herbs and how these would activate cognate pattern recognition receptors (PPRs) in the upper gut-associated lymphoid tissue (GALT), including Peyer's patches and the lamina propria, to boost antibody titers, CD8+ and CD4+ T cells, NK activity, hematopoiesis, and facilitating M2 to M1 macrophage phenotype transition in a similar manner as vaccines. This new knowledge could pave the way for developing bioreactor-grown/heat-inactivated MOC therapies to boost human immunity against infections and improve tumor surveillance.
Collapse
Affiliation(s)
- Elizabeth Mazzio
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| | - Andrew Barnes
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| | - Ramesh Badisa
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| | - Stevie Council
- John Gnabre Science Research Institute, Baltimore, MD, United States
| | - Karam F. A. Soliman
- Divison of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Institute of Public Health, Florida A & M University, Tallahassee, FL, United States
| |
Collapse
|
14
|
Hu X, Gattis C, Olroyd AG, Friera AM, White K, Young C, Basco R, Lamba M, Wells F, Ankala R, Dowdle WE, Lin A, Egenberger K, Rukstalis JM, Millman JR, Connolly AJ, Deuse T, Schrepfer S. Human hypoimmune primary pancreatic islets avoid rejection and autoimmunity and alleviate diabetes in allogeneic humanized mice. Sci Transl Med 2023; 15:eadg5794. [PMID: 37043559 DOI: 10.1126/scitranslmed.adg5794] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2023]
Abstract
Transplantation of allogeneic pancreatic donor islets has successfully been performed in selected patients with difficult-to-control insulin-dependent diabetes and impaired awareness of hypoglycemia (IAH). However, the required systemic immunosuppression associated with this procedure prevents this cell replacement therapy from more widespread adoption in larger patient populations. We report the editing of primary human islet cells to the hypoimmune HLA class I- and class II-negative and CD47-overexpressing phenotype and their reaggregation into human HIP pseudoislets (p-islets). Human HIP p-islets were shown to survive, engraft, and ameliorate diabetes in immunocompetent, allogeneic, diabetic humanized mice. HIP p-islet cells were further shown to avoid autoimmune killing in autologous, diabetic humanized autoimmune mice. The survival and endocrine function of HIP p-islet cells were not impaired by contamination of unedited or partially edited cells within the p-islets. HIP p-islet cells were eliminated quickly and reliably in this model using a CD47-targeting antibody, thus providing a safety strategy in case HIP cells exert toxicity in a future clinical setting. Transplantation of human HIP p-islets for which no immunosuppression is required has the potential to lead to wider adoption of this therapy and help more diabetes patients with IAH and history of severe hypoglycemic events to achieve insulin independence.
Collapse
Affiliation(s)
- Xiaomeng Hu
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Corie Gattis
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Ari G Olroyd
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Annabelle M Friera
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Kathy White
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Chi Young
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Ron Basco
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Meghan Lamba
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Frank Wells
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Ramya Ankala
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - William E Dowdle
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - August Lin
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Kyla Egenberger
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | | | - Jeffrey R Millman
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Andrew J Connolly
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tobias Deuse
- Department of Surgery, Division of Cardiothoracic Surgery, Transplant and Stem Cell Immunobiology (TSI) Lab, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sonja Schrepfer
- Sana Biotechnology Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| |
Collapse
|
15
|
Cui X, Yan C, Xu Y, Li D, Guo M, Sun L, Zhu Z. Allograft rejection following immune checkpoint inhibitors in solid organ transplant recipients: A safety analysis from a literature review and a pharmacovigilance system. Cancer Med 2023; 12:5181-5194. [PMID: 36504294 PMCID: PMC10028127 DOI: 10.1002/cam4.5394] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 10/13/2022] [Accepted: 10/21/2022] [Indexed: 12/14/2022] Open
Abstract
AIM This study aimed to systematically characterize transplant rejection after immune checkpoint inhibitors (ICIs) initiation in solid organ transplant recipients (SOTRs). METHODS Data were extracted from the US FDA Adverse Event Reporting System (FAERS) database and case reports in the literature. Disproportionality analysis including information component and reported odds ratio (ROR) was performed to access potential risk signals. RESULTS A total of 168 patients with transplant rejection after ICIs usage were identified in the FAERS database, and 89 cases were identified in the literature review. ICIs were significantly associated with transplant rejection (ROR025 : 2.2). A strong risk signal was found for combination therapy with pembrolizumab and ipilimumab compared to monotherapy. CONCLUSION Immune checkpoint inhibitors were significantly associated with transplant rejection in SOTRs.
Collapse
Affiliation(s)
- Xiangli Cui
- Pharmacy Department of Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Cilin Yan
- School of Automation Science and Electrical Engineering, Beihang University, Beijing, China
| | - Ye Xu
- Pharmacy Department of Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Dandan Li
- Pharmacy Department of Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Mingxing Guo
- Pharmacy Department of Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Liying Sun
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Zhijun Zhu
- Liver Transplantation Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
O'Neil A, Brook M, Abdul-Wahab S, Hester J, Lombardi G, Issa F. A GMP Protocol for the Manufacture of Tregs for Clinical Application. Methods Mol Biol 2023; 2559:205-227. [PMID: 36180635 DOI: 10.1007/978-1-0716-2647-4_14] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Infusion of regulatory T cells is a promising therapeutic strategy in organ transplantation to modulate the immune system, prevent rejection, minimize the need for pharmaceutical immunosuppression, and improve long-term transplant outcomes. Here we describe a GMP-compliant method we have used for the manufacture of ex vivo expanded autologous regulatory T cells for use in clinical trials.
Collapse
Affiliation(s)
- Alice O'Neil
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Matthew Brook
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Seetha Abdul-Wahab
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Joanna Hester
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Giovanna Lombardi
- MRC Centre for Transplantation, School of Immunology and Microbial Sciences, King's College London, Guy's Hospital, London, UK
| | - Fadi Issa
- Transplantation Research Immunology Group, Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| |
Collapse
|
17
|
Marra G, Soria F, Peretti F, Oderda M, Dariane C, Timsit MO, Branchereau J, Hedli O, Mesnard B, Tilki D, Olsburgh J, Kulkarni M, Kasivisvanathan V, Lebacle C, Rodriguez-Faba O, Breda A, Soeterik T, Gandaglia G, Todeschini P, Biancone L, Gontero P, on behalf of the Collaborators †. Prostate Cancer in Renal Transplant Recipients: Results from a Large Contemporary Cohort. Cancers (Basel) 2022; 15:189. [PMID: 36612184 PMCID: PMC9818510 DOI: 10.3390/cancers15010189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/18/2022] [Accepted: 11/18/2022] [Indexed: 12/31/2022] Open
Abstract
Objectives: The aim of this study was to assess the natural history of prostate cancer (PCa) in renal transplant recipients (RTRs) and to clarify the controversy over whether RTRs have a higher risk of PCa and poorer outcomes than non-RTRs, due to factors such as immunosuppression. Patients and Methods: We performed a retrospective multicenter study of RTRs diagnosed with cM0 PCa between 2001 and 2019. Primary outcomes were overall (OS) and cancer-specific survival (CSS). Secondary outcomes included biochemical recurrence and/or progression after active surveillance (AS) and evaluation of variables possibly influencing PCa aggressiveness and outcomes. Management modalities included surgery, radiation, cryotherapy, HIFU, AS, and watchful waiting. Results: We included 166 men from nine institutions. Median age and eGFR at diagnosis were 67 (IQR 60−73) and 45.9 mL/min (IQR 31.5−63.4). ASA score was >2 in 58.4% of cases. Median time from transplant to PCa diagnosis was 117 months (IQR 48−191.5), and median PSA at diagnosis was 6.5 ng/mL (IQR 5.02−10). The biopsy Gleason score was ≥8 in 12.8%; 11.6% and 6.1% patients had suspicion of ≥cT3 > cT2 and cN+ disease. The most frequent management method was radical prostatectomy (65.6%), followed by radiation therapy (16.9%) and AS (10.2%). At a median follow-up of 60.5 months (IQR 31−106) 22.9% of men (n = 38) died, with only n = 4 (2.4%) deaths due to PCa. Local and systemic progression rates were 4.2% and 3.0%. On univariable analysis, no major influence of immunosuppression type was noted, with the exception of a protective effect of antiproliferative agents (HR 0.39, 95% CI 0.16−0.97, p = 0.04) associated with a decreased risk of biochemical recurrence (BCR) or progression after AS. Conclusion: PCa diagnosed in RTRs is mainly of low to intermediate risk and organ-confined at diagnosis, with good cancer control and low PCa death at intermediate follow-up. RTRs have a non-negligible risk of death from causes other than PCa. Aggressive upfront management of the majority of RTRs with PCa may, therefore, be avoided.
Collapse
Affiliation(s)
- Giancarlo Marra
- Department of Surgical Sciences, University of Turin and Città della Salute e della Scienza, 10126 Turin, Italy
- Department of Urology, Institut Mutualiste Montsouris and Université Paris Descartes, 75014 Paris, France
- Department of Urology, Hôpital Tenon, 75020 Paris, France
| | - Francesco Soria
- Department of Surgical Sciences, University of Turin and Città della Salute e della Scienza, 10126 Turin, Italy
| | - Federica Peretti
- Department of Surgical Sciences, University of Turin and Città della Salute e della Scienza, 10126 Turin, Italy
| | - Marco Oderda
- Department of Surgical Sciences, University of Turin and Città della Salute e della Scienza, 10126 Turin, Italy
| | - Charles Dariane
- Department of Urology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Marc-Olivier Timsit
- Department of Urology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Julien Branchereau
- Institut de Transplantation Urologie Nèphrologie (ITUN), CHU Nantes, 44093 Nantes, France
- Nuffield Department of Surgical Sciences, Oxford University, Oxford OX1 2JD, UK
| | - Oussama Hedli
- Institut de Transplantation Urologie Nèphrologie (ITUN), CHU Nantes, 44093 Nantes, France
| | - Benoit Mesnard
- Institut de Transplantation Urologie Nèphrologie (ITUN), CHU Nantes, 44093 Nantes, France
| | - Derya Tilki
- Martini-Klinik Prostate Cancer Center, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
- Department of Urology, University Hospital Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | | | | | - Cedric Lebacle
- Department of Urology, Kremlin-Bicêtre Hospital, 94270 Le Kremlin-Bicêtre, France
| | | | - Alberto Breda
- Department of Urology, Fundacio Puigvert, 08025 Barcelona, Spain
| | - Timo Soeterik
- Department of Urology, Saint Antonius Hospital, 3543 AZ Utrecht, The Netherlands
| | | | - Paola Todeschini
- Department of Nephrology, Sant’Orsola Malpighi Hospital, 40138 Bologna, Italy
| | - Luigi Biancone
- Department of Nephrology, Sant’Orsola Malpighi Hospital, 40138 Bologna, Italy
- Department of Nephrology, University of Turin and Città della Salute e della Scienza, 10126 Turin, Italy
| | - Paolo Gontero
- Department of Surgical Sciences, University of Turin and Città della Salute e della Scienza, 10126 Turin, Italy
| | | |
Collapse
|
18
|
Wang Z, Li Y, Sun C, Cui P, Han Y, Wu T, Xu B, Zhang C, Shi L, Dai J. Locally controlled release of immunosuppressive promotes survival of transplanted adult spinal cord tissue. Regen Biomater 2022; 10:rbac097. [PMID: 36683735 PMCID: PMC9845520 DOI: 10.1093/rb/rbac097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/04/2022] [Accepted: 11/19/2022] [Indexed: 12/12/2022] Open
Abstract
Transplantation of adult spinal cord tissue (aSCT) is a promising treatment for spinal cord injury (SCI) basing on various types of neural cells and matrix components inside aSCT. However, long-term systemic administration of immunosuppressors (e.g. tacrolimus, TAC) is required for the survival of allogeneic tissue, which often associated with severe side effects such as infection, liver damageand renal failure. In this study, a triglycerol monostearate (TGM)-based TAC delivery system (e.g. TAC@TGM) with high drug loading concentration was developed, which possessed injectable properties as well as sustainable and immune-responsive drug release behaviors. In complete transected SCI model, locally injected TAC@TGM could reduce the infiltration of inflammation cells, enhance the survival of transplanted aSCT (e.g. Tuj-1+ and NF+ neurons) and promote the recovery of locomotor function. Moreover, controlled release of TAC by TAC@TGM attenuated side effects of TAC on liver and kidneys compared with traditional systemic administration. More importantly, the developed TAC@TGM system provided a facile single dose of long-term immunosuppressive effect not just for aSCT transplantation, but also for other tissue/organ and cell transplantations.
Collapse
Affiliation(s)
| | | | - Chenxuan Sun
- College of Biology, Hunan University, Changsha 410000, China
| | - Pukong Cui
- College of Biology, Hunan University, Changsha 410000, China
| | - Yuanyuan Han
- College of Biology, Hunan University, Changsha 410000, China
| | - Tong Wu
- College of Biology, Hunan University, Changsha 410000, China
| | - Bai Xu
- State Key Laboratory of Molecular, Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Can Zhang
- Correspondence address. E-mail: (J.D.); (L.S.); (C.Z.)
| | - Liyang Shi
- Correspondence address. E-mail: (J.D.); (L.S.); (C.Z.)
| | - Jianwu Dai
- Correspondence address. E-mail: (J.D.); (L.S.); (C.Z.)
| |
Collapse
|
19
|
Hotta K, Hirose T, Kawai T. Clinical trials for renal allograft tolerance induction through combined hematopoietic stem cell transplantation: A narrative review. Int J Urol 2022; 29:1397-1404. [PMID: 36101964 DOI: 10.1111/iju.15035] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/15/2022] [Indexed: 11/30/2022]
Abstract
During the last four decades, development of effective immunosuppressants has significantly improved short-term results of organ transplantation. However, long-term results have not been satisfactory due to chronic rejection or complications caused by immunosuppressive drugs. Therefore, induction of immunological tolerance of the transplanted organ is considered essential to improve the long-term results. Despite numerous tolerance strategies that have been successful in murine models, inducing hematopoietic chimerism through donor hematopoietic stem cell transplantation is the only method that reproducibly induces kidney allograft tolerance in nonhuman primates or humans. Combining kidney and hematopoietic stem cell transplantation to achieve allograft tolerance has now been attempted with different chimerism strategies. This review summarizes the status of current clinical trials on the induction of allograft tolerance. We also summarize recent studies to extend the chimerism approach to deceased donor transplant recipients.
Collapse
Affiliation(s)
- Kiyohiko Hotta
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Takayuki Hirose
- Department of Urology, Hokkaido University Hospital, Sapporo, Japan
| | - Tatsuo Kawai
- Center for Transplantation Sciences, Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Characteristic Genes and Immune Infiltration Analysis for Acute Rejection after Kidney Transplantation. DISEASE MARKERS 2022; 2022:6575052. [PMID: 36393969 PMCID: PMC9646319 DOI: 10.1155/2022/6575052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/05/2022] [Indexed: 11/06/2022]
Abstract
Background Renal transplantation can significantly improve the survival rate and quality of life of patients with end-stage renal disease, but the probability of acute rejection (AR) in adult renal transplant recipients is still approximately 12.2%. Machine learning (ML) is superior to traditional statistical methods in various clinical scenarios. However, the current AR model is constructed only through simple difference analysis or a single queue, which cannot guarantee the accuracy of prediction. Therefore, this study identified and validated new gene sets that contribute to the early prediction of AR and the prognosis prediction of patients after renal transplantation by constructing a more accurate AR gene signature through ML technology. Methods Based on the Gene Expression Omnibus (GEO) database and multiple bioinformatic analyses, we identified differentially expressed genes (DEGs) and built a gene signature via LASSO regression and SVM analysis. Immune cell infiltration and immunocyte association analyses were also conducted. Furthermore, we investigated the relationship between AR genes and graft survival status. Results Twenty-four DEGs were identified. A 5 gene signature (CPA6, EFNA1, HBM, THEM5, and ZNF683) were obtained by LASSO analysis and SVM analysis, which had a satisfied ability to differentiate AR and NAR in the training cohort, internal validation cohort and external validation cohort. Additionally, ZNF683 was associated with graft survival. Conclusion A 5 gene signature, particularly ZNF683, provided insight into a precise therapeutic schedule and clinical applications for AR patients.
Collapse
|
21
|
Alex RA, Mathews SS, Paul RR, Albert RRA. Laryngeal Dysplasia: To Biopsy or Not? Indian J Otolaryngol Head Neck Surg 2022; 74:2331-2333. [PMID: 36452722 PMCID: PMC9702373 DOI: 10.1007/s12070-020-02161-9] [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: 09/09/2020] [Accepted: 09/21/2020] [Indexed: 10/23/2022] Open
Abstract
A renal transplant recipient with chronic hepatitis B underwent multiple laser excisions over 4 years for laryngeal keratosis. From the initial histopathology reports of mild to moderate dysplasia, a progression to squamous carcinoma was noted over 4 years. This case report highlights the possible role of immunosuppressants and hepatitis virus in the aetiopathogenesis of laryngeal carcinoma.
Collapse
Affiliation(s)
- Reshmi Anna Alex
- Department of ENT, Christian Medical College, Vellore, Tamil Nadu 632004 India
| | - Suma Susan Mathews
- Department of ENT, Christian Medical College, Vellore, Tamil Nadu 632004 India
| | - Roshna Rose Paul
- Department of ENT, Christian Medical College, Vellore, Tamil Nadu 632004 India
| | - Rita Ruby A. Albert
- Department of ENT, Christian Medical College, Vellore, Tamil Nadu 632004 India
| |
Collapse
|
22
|
Guillén-Gómez I, Blanco-García M, Aránega-Gavilán S, Crespo-Montero R. Alteraciones neoplásicas en el paciente trasplantado renal. Una revisión sistemática. ENFERMERÍA NEFROLÓGICA 2022. [DOI: 10.37551/52254-28842022021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Introducción: El trasplante renal se ha destacado como la mejor terapia renal sustitutiva para la enfermedad renal crónica terminal, aunque conlleva una serie de riesgos entre los que el cáncer se destaca cada vez más, por lo que parece importante determinar el impacto que tiene en esta población.Objetivo: Conocer y sintetizar la evidencia científica acerca de la incidencia de neoplasias en pacientes trasplantados renales, así como las variables relacionadas con las mismas.Metodología: Se realizó una revisión sistemática en la que se incluyeron 14 artículos procedentes de las bases de Pubmed, Scopus, Scielo, WOS y Google Académico. Como términos MeSH se utilizaron: neoplasias (neoplasms), trasplante de riñón (kidney transplantation), incidencia (incidence), riesgo (risk), factores de riesgo (risk factors). Se incluyeron artículos originales en inglés y español, en población trasplantada adulta.Resultados: Se incluyeron 14 artículos de diseño observacional. De la revisión emergieron como principales variables: incidencia general, datos demográficos y tratamientos previos, otras características asociadas, tumores más frecuentes y medidas preventivas.Conclusiones: La incidencia de neoplasias en el paciente trasplantado es alta, sobre todo en la edad avanzada y pacientes con tratamiento inmunosupresor con ciclosporina y azatioprina siendo este último, junto al trasplante procedente de cadáver, los principales factores de riesgo encontrados. Los cánceres de piel no melanoma son los más prevalentes y como medidas de prevención se destaca llevar a cabo un cribado rutinario, fomento de una vida saludable, y promoción y potenciación de la vacunación de la hepatitis B.
Collapse
|
23
|
Qin H, Sun C, Zhu Y, Qin Y, Ren S, Wang Z, Li C, Li X, Zhang B, Hao J, Li G, Wang H, Shao B, Zhang J, Wang H. IL-37 overexpression promotes endometrial regenerative cell-mediated inhibition of cardiac allograft rejection. Stem Cell Res Ther 2022; 13:302. [PMID: 35841010 PMCID: PMC9284885 DOI: 10.1186/s13287-022-02982-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 04/19/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endometrial regenerative cells (ERCs) play an important role in attenuation of acute allograft rejection, while their effects are limited. IL-37, a newly discovered immunoregulatory cytokine of the IL-1 family, can regulate both innate and adaptive immunity. Whether IL-37 overexpression can enhance the therapeutic effects of ERCs in inhibition of acute cardiac allograft rejection remains unknown and will be explored in this study. METHODS C57BL/6 mice recipients receiving BALB/c mouse heterotopic heart allografts were randomly divided into the phosphate-buffered saline (untreated), ERC treated, negative lentiviral control ERC (NC-ERC) treated, and IL-37 overexpressing ERC (IL-37-ERC) treated groups. Graft pathological changes were assessed by H&E staining. The intra-graft cell infiltration and splenic immune cell populations were analyzed by immunohistochemistry and flow cytometry, respectively. The stimulatory property of recipient DCs was tested by an MLR assay. Furthermore, serum cytokine profiles of recipients were measured by ELISA assay. RESULTS Mice treated with IL-37-ERCs achieved significantly prolonged allograft survival compared with the ERC-treated group. Compared with all the other control groups, IL-37-ERC-treated group showed mitigated inflammatory response, a significant increase in tolerogenic dendritic cells (Tol-DCs), regulatory T cells (Tregs) in the grafts and spleens, while a reduction of Th1 and Th17 cell population. Additionally, there was a significant upregulation of immunoregulatory IL-10, while a reduction of IFN-γ, IL-17A, IL-12 was detected in the sera of IL-37-ERC-treated recipients. CONCLUSION IL-37 overexpression can promote the therapeutic effects of ERCs to inhibit acute allograft rejection and further prolong graft survival. This study suggests that gene-modified ERCs overexpressing IL-37 may pave the way for novel therapeutic options in the field of transplantation.
Collapse
Affiliation(s)
- Hong Qin
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Chenglu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Yanglin Zhu
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Yafei Qin
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Shaohua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Zhaobo Wang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Chuan Li
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Baoren Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Jingpeng Hao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China.,Department of Anorectal Surgery, Tianjin Medical University Second Hospital, Tianjin, China
| | - Guangming Li
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Hongda Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Jingyi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.,Tianjin General Surgery Institute, Tianjin, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China. .,Tianjin General Surgery Institute, Tianjin, China.
| |
Collapse
|
24
|
Sen T, Thummer RP. The Impact of Human Microbiotas in Hematopoietic Stem Cell and Organ Transplantation. Front Immunol 2022; 13:932228. [PMID: 35874759 PMCID: PMC9300833 DOI: 10.3389/fimmu.2022.932228] [Citation(s) in RCA: 6] [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: 04/29/2022] [Accepted: 06/06/2022] [Indexed: 11/18/2022] Open
Abstract
The human microbiota heavily influences most vital aspects of human physiology including organ transplantation outcomes and transplant rejection risk. A variety of organ transplantation scenarios such as lung and heart transplantation as well as hematopoietic stem cell transplantation is heavily influenced by the human microbiotas. The human microbiota refers to a rich, diverse, and complex ecosystem of bacteria, fungi, archaea, helminths, protozoans, parasites, and viruses. Research accumulating over the past decade has established the existence of complex cross-species, cross-kingdom interactions between the residents of the various human microbiotas and the human body. Since the gut microbiota is the densest, most popular, and most studied human microbiota, the impact of other human microbiotas such as the oral, lung, urinary, and genital microbiotas is often overshadowed. However, these microbiotas also provide critical and unique insights pertaining to transplantation success, rejection risk, and overall host health, across multiple different transplantation scenarios. Organ transplantation as well as the pre-, peri-, and post-transplant pharmacological regimens patients undergo is known to adversely impact the microbiotas, thereby increasing the risk of adverse patient outcomes. Over the past decade, holistic approaches to post-transplant patient care such as the administration of clinical and dietary interventions aiming at restoring deranged microbiota community structures have been gaining momentum. Examples of these include prebiotic and probiotic administration, fecal microbial transplantation, and bacteriophage-mediated multidrug-resistant bacterial decolonization. This review will discuss these perspectives and explore the role of different human microbiotas in the context of various transplantation scenarios.
Collapse
Affiliation(s)
| | - Rajkumar P. Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| |
Collapse
|
25
|
Pinto-Filho VA, Nascimento E, Cunha APL, Assis BPS, Lasmar MF, Vianna HR, Fabreti-Oliveira RA. Malignancy Diseases in Kidney Transplantation, Clinical Outcomes, Patient, and Allograft Survival: A Case-Control Study. Transplant Proc 2022; 54:1253-1261. [PMID: 35750515 DOI: 10.1016/j.transproceed.2022.02.063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/09/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND Malignancy is a well-known complication in patients after kidney transplantation (KT), but its effect on posttransplant outcomes, allograft, and patient survival remains unexplored. The aim of this study is to report the impact of the comorbidity on clinical outcome, function, and failure of an allograft kidney. METHODS This case-control study included 101 KT patients. Twenty-six patients who developed cancer (CA) were assigned to the case group and 75 to the control group. Statistical analysis was performed using logistic regression models, and graft survival was analyzed using the Kaplan-Meier curve. RESULTS Non-melanoma skin CA was the most common malignancy, accounting for almost 60% of cases, followed by stomach CA, prostate CA, and lymphoproliferative diseases (7.70% each). Difference in graft and patient survival was not significant between the two groups (P > .05). A tumor in nonfunctioning in the first nonfunctioning KT was identified in 1 KT patient with a second allograft and by anatomopathological was detect Fuhrman grade II renal cell carcinoma. This KT patient was in good clinical condition with serum creatinine level of 1.5 mg/dL. CONCLUSIONS No association was observed between CA development and risk factors, including family history and smoking habit, and no differences in allograft and patient survival were found. Nevertheless, in our data, CA in KT patients occurred early after transplantation. Renal cell carcinoma in allograft failure was identified in a patient; that suggested that nephrectomy of kidney failure must be performed to avoid patient allosensitization and neoplasia. Thus, we suggest continuous screening of malignancy diseases for KT patients.
Collapse
Affiliation(s)
| | - Evaldo Nascimento
- IMUNOLAB - Laboratory of Histocompatibility, Belo Horizonte, Minas Gerais, Brazil; Institute of Research and Education of the Hospital Santa Casa, Belo Horizonte, Minas Gerais, Brazil
| | - Antônio P L Cunha
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil
| | - Bernardo P S Assis
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil
| | - Marcus F Lasmar
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil
| | - Heloísa R Vianna
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; University Hospital of the Faculty of Medical Science, Belo Horizonte, Minas Gerais, Brazil
| | - Raquel A Fabreti-Oliveira
- Faculty of Medical Sciences, Belo Horizonte, Minas Gerais, Brazil; IMUNOLAB - Laboratory of Histocompatibility, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
26
|
Yue C, Gao S, Li S, Xing Z, Qian H, Hu Y, Wang W, Hua C. TIGIT as a Promising Therapeutic Target in Autoimmune Diseases. Front Immunol 2022; 13:911919. [PMID: 35720417 PMCID: PMC9203892 DOI: 10.3389/fimmu.2022.911919] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/13/2022] [Indexed: 12/19/2022] Open
Abstract
Co-inhibitory receptors (IRs) are molecules that protect host against autoimmune reactions and maintain peripheral self-tolerance, playing an essential role in maintaining immune homeostasis. In view of the substantial clinical progresses of negative immune checkpoint blockade in cancer treatment, the role of IRs in autoimmune diseases is also obvious. Several advances highlighted the substantial impacts of T cell immunoglobulin and ITIM domain (TIGIT), a novel IR, in autoimmunity. Blockade of TIGIT pathway exacerbates multiple autoimmune diseases, whereas enhancement of TIGIT function has been shown to alleviate autoimmune settings in mice. These data suggested that TIGIT pathway can be manipulated to achieve durable tolerance to treat autoimmune disorders. In this review, we provide an overview of characteristics of TIGIT and its role in autoimmunity. We then discuss recent approaches and future directions to leverage our knowledge of TIGIT as therapeutic target in autoimmune diseases.
Collapse
Affiliation(s)
- Chenran Yue
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Wenzhou, China
| | - Shuting Li
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Zhouhang Xing
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hengrong Qian
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ying Hu
- School of the Second Clinical Medical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Wenqian Wang
- Department of Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
27
|
Fuhrmann JD, Valkova K, von Moos S, Wüthrich RP, Müller TF, Schachtner T. Cancer among kidney transplant recipients >20 years after transplantation: post-transplant lymphoproliferative disorder remains the most common cancer type in the ultra long-term. Clin Kidney J 2022; 15:1152-1159. [PMID: 35664271 PMCID: PMC9155242 DOI: 10.1093/ckj/sfac013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Indexed: 11/23/2022] Open
Abstract
Background Cancer risk is increased by 2- to 4-fold in kidney transplant recipients (KTRs) compared with the general population. Little attention, however, has been given to KTRs with ultra long-term survival >20 years. Methods We studied 293 of 1241 KTRs (23.6%), transplanted between 1981 and 1999, who showed kidney allograft survival >20 years. These long-term survivors were analysed for cancer development, cancer type, cancer-associated risk factors and patient and allograft outcomes. Results By 10, 20 and 30 years post-transplantation, these long-term KTRs showed a cancer rate of 4.4%, 14.6% and 33.2%, and a non-melanoma skin cancer (NMSC) rate of 10.3%, 33.5% and 76.8%, respectively. By recipients' ages of 40, 60 and 80 years, KTRs showed a cancer rate of 3.4%, 14.5% 55.2%, and a NMSC rate of 1.7%, 31.6% and 85.2%, respectively. By 30 years post-transplantation, post-transplant lymphoproliferative disorder (PTLD) showed the highest incidence of 8.5%, followed by renal cell carcinoma (RCC) with 5.1%. Risk factors associated with the development of cancer were only recipient age (P = 0.016). Smoking history was associated with the risk of lung cancer (P = 0.018). Risk factors related to the development of NMSC included recipient age (P = 0.001) and thiazide diuretics (P = 0.001). Cancer increased the risk of death by 2.4-fold (P = 0.002), and PTLD increased the risk of kidney allograft loss by 6.5-fold (P = 0.001). No differences were observed concerning the development of donor-specific antibodies (P > 0.05). Conclusions In long-term KTRs, cancer is a leading cause of death. PTLD remains the most common cancer type followed by RCC. These results emphasize the need for focused long-term cancer surveillance protocols.
Collapse
Affiliation(s)
- Julia D Fuhrmann
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Kristyna Valkova
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Seraina von Moos
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Rudolf P Wüthrich
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas F Müller
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Schachtner
- Division of Nephrology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
28
|
Sakowska J, Arcimowicz Ł, Jankowiak M, Papak I, Markiewicz A, Dziubek K, Kurkowiak M, Kote S, Kaźmierczak-Siedlecka K, Połom K, Marek-Trzonkowska N, Trzonkowski P. Autoimmunity and Cancer-Two Sides of the Same Coin. Front Immunol 2022; 13:793234. [PMID: 35634292 PMCID: PMC9140757 DOI: 10.3389/fimmu.2022.793234] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 04/12/2022] [Indexed: 02/06/2023] Open
Abstract
Autoimmune disease results from the immune response against self-antigens, while cancer develops when the immune system does not respond to malignant cells. Thus, for years, autoimmunity and cancer have been considered as two separate fields of research that do not have a lot in common. However, the discovery of immune checkpoints and the development of anti-cancer drugs targeting PD-1 (programmed cell death receptor 1) and CTLA-4 (cytotoxic T lymphocyte antigen 4) pathways proved that studying autoimmune diseases can be extremely helpful in the development of novel anti-cancer drugs. Therefore, autoimmunity and cancer seem to be just two sides of the same coin. In the current review, we broadly discuss how various regulatory cell populations, effector molecules, genetic predisposition, and environmental factors contribute to the loss of self-tolerance in autoimmunity or tolerance induction to cancer. With the current paper, we also aim to convince the readers that the pathways involved in cancer and autoimmune disease development consist of similar molecular players working in opposite directions. Therefore, a deep understanding of the two sides of immune tolerance is crucial for the proper designing of novel and selective immunotherapies.
Collapse
Affiliation(s)
- Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Łukasz Arcimowicz
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Martyna Jankowiak
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| | - Ines Papak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Aleksandra Markiewicz
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdańsk and Medical University of Gdańsk, Gdańsk, Poland
| | - Katarzyna Dziubek
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Małgorzata Kurkowiak
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | - Sachin Kote
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
| | | | - Karol Połom
- Department of Surgical Oncology, Medical University of Gdańsk, Gdańsk, Poland
| | - Natalia Marek-Trzonkowska
- International Centre for Cancer Vaccine Science, University of Gdańsk, Gdańsk, Poland
- Laboratory of Immunoregulation and Cellular Therapies, Department of Family Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Gdańsk, Poland
| |
Collapse
|
29
|
Ghandili S, Kluger MA, Leitner T, Grahammer F, Kirchner L, Modemann F, Achilles E, Kreipe HH, Klein J, Steinemann D, Wolschke C, Fischer L, Bokemeyer C, Fiedler W, Huber TB, Alsdorf WH, Mahmud M. Donor‐transmitted extramedullary acute myeloid leukaemia after living donor kidney transplantation. Br J Haematol 2022; 198:199-202. [PMID: 35428972 PMCID: PMC9321064 DOI: 10.1111/bjh.18194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Susanne Ghandili
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Malte A. Kluger
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Theo Leitner
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Florian Grahammer
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Lennart Kirchner
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Franziska Modemann
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
- Mildred Scheel Cancer Career Center, University Cancer Center Hamburg University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Eike‐Gert Achilles
- Department of Visceral Transplantation University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Hans H. Kreipe
- Institute of Pathology, Bone Marrow Reference Center Hannover Medical School Hanover Germany
| | - Janin Klein
- Institute of Human Genetics Hannover Medical School Hanover Germany
| | - Doris Steinemann
- Institute of Human Genetics Hannover Medical School Hanover Germany
| | - Christine Wolschke
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Lutz Fischer
- Department of Visceral Transplantation University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Walter Fiedler
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Tobias B. Huber
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Winfried H. Alsdorf
- Department of Oncology, Hematology, Bone Marrow Transplantation with Section Pneumology University Medical Center Hamburg‐Eppendorf Hamburg Germany
| | - Maida Mahmud
- III. Department of Medicine University Medical Center Hamburg‐Eppendorf Hamburg Germany
| |
Collapse
|
30
|
Brook MO, Hester J, Petchey W, Rombach I, Dutton S, Bottomley MJ, Black J, Abdul-Wahab S, Bushell A, Lombardi G, Wood K, Friend P, Harden P, Issa F. Transplantation Without Overimmunosuppression (TWO) study protocol: a phase 2b randomised controlled single-centre trial of regulatory T cell therapy to facilitate immunosuppression reduction in living donor kidney transplant recipients. BMJ Open 2022; 12:e061864. [PMID: 35428650 PMCID: PMC9014059 DOI: 10.1136/bmjopen-2022-061864] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
INTRODUCTION Regulatory T cell (Treg) therapy has been demonstrated to facilitate long-term allograft survival in preclinical models of transplantation and may permit reduction of immunosuppression and its associated complications in the clinical setting. Phase 1 clinical trials have shown Treg therapy to be safe and feasible in clinical practice. Here we describe a protocol for the TWO study, a phase 2b randomised control trial of Treg therapy in living donor kidney transplant recipients that will confirm safety and explore efficacy of this novel treatment strategy. METHODS AND ANALYSIS 60 patients will be randomised on a 1:1 basis to Treg therapy (TR001) or standard clinical care (control). Patients in the TR001 arm will receive an infusion of autologous polyclonal ex vivo expanded Tregs 5 days after transplantation instead of standard monoclonal antibody induction. Maintenance immunosuppression will be reduced over the course of the post-transplant period to low-dose tacrolimus monotherapy. Control participants will receive a standard basiliximab-based immunosuppression regimen with long-term tacrolimus and mycophenolate mofetil immunosuppression. The primary endpoint is biopsy proven acute rejection over 18 months; secondary endpoints include immunosuppression burden, chronic graft dysfunction and drug-related complications. ETHICS AND DISSEMINATION Ethical approval has been provided by the National Health Service Health Research Authority South Central-Oxford A Research Ethics Committee (reference 18/SC/0054). The study also received authorisation from the UK Medicines and Healthcare products Regulatory Agency and is being run in accordance with the principles of Good Clinical Practice, in collaboration with the registered trials unit Oxford Clinical Trials Research Unit. Results from the TWO study will be published in peer-reviewed scientific/medical journals and presented at scientific/clinical symposia and congresses. TRIAL REGISTRATION NUMBER ISRCTN: 11038572; Pre-results.
Collapse
Affiliation(s)
- Matthew Oliver Brook
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Oxford Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - William Petchey
- Oxford Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Ines Rombach
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Susan Dutton
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Matthew James Bottomley
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Oxford Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Joanna Black
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, UK
| | - Seetha Abdul-Wahab
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- NIHR Biomedical Research Centre GMP unit, Guy's Hospital, London, UK
| | - Andrew Bushell
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Giovanna Lombardi
- NIHR Biomedical Research Centre GMP unit, Guy's Hospital, London, UK
- Peter Gorer Department of Immunobiology, King's College London Faculty of Life Sciences and Medicine, London, UK
| | - Kathryn Wood
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Peter Friend
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
- Oxford Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Paul Harden
- Oxford Transplant Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| |
Collapse
|
31
|
Namestnikov M, Dekel B. Moving To A New Dimension: 3D Kidney Cultures For Kidney Regeneration. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
Podestà MA, Sykes M. Chimerism-Based Tolerance to Kidney Allografts in Humans: Novel Insights and Future Perspectives. Front Immunol 2022; 12:791725. [PMID: 35069574 PMCID: PMC8767096 DOI: 10.3389/fimmu.2021.791725] [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: 10/08/2021] [Accepted: 12/15/2021] [Indexed: 11/18/2022] Open
Abstract
Chronic rejection and immunosuppression-related toxicity severely affect long-term outcomes of kidney transplantation. The induction of transplantation tolerance – the lack of destructive immune responses to a transplanted organ in the absence of immunosuppression – could potentially overcome these limitations. Immune tolerance to kidney allografts from living donors has been successfully achieved in humans through clinical protocols based on chimerism induction with hematopoietic cell transplantation after non-myeloablative conditioning. Notably, two of these protocols have led to immune tolerance in a significant fraction of HLA-mismatched donor-recipient combinations, which represent the large majority of cases in clinical practice. Studies in mice and large animals have been critical in dissecting tolerance mechanisms and in selecting the most promising approaches for human translation. However, there are several key differences in tolerance induction between these models and humans, including the rate of success and stability of donor chimerism, as well as the relative contribution of different mechanisms in inducing donor-specific unresponsiveness. Kidney allograft tolerance achieved through durable full-donor chimerism may be due to central deletion of graft-reactive donor T cells, even though mechanistic data from patient series are lacking. On the other hand, immune tolerance attained with transient mixed chimerism-based protocols initially relies on Treg-mediated suppression, followed by peripheral deletion of donor-reactive recipient T-cell clones under antigenic pressure from the graft. These conclusions were supported by data deriving from novel high-throughput T-cell receptor sequencing approaches that allowed tracking of alloreactive repertoires over time. In this review, we summarize the most important mechanistic studies on tolerance induction with combined kidney-bone marrow transplantation in humans, discussing open issues that still need to be addressed and focusing on techniques developed in recent years to efficiently monitor the alloresponse in tolerance trials. These cutting-edge methods will be instrumental for the development of immune tolerance protocols with improved efficacy and to identify patients amenable to safe immunosuppression withdrawal.
Collapse
Affiliation(s)
- Manuel Alfredo Podestà
- Renal Division, ASST Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milano, Italy
| | - Megan Sykes
- Columbia Center for Translational Immunology, Department of Medicine, Department of Surgery, Department of Microbiology and Immunology, Columbia University, New York, NY, United States
| |
Collapse
|
33
|
Batra J, DeFilippis EM, Golob S, Clerkin K, Topkara VK, Habal MV, Restaino S, Griffin J, Hi Lee S, Latif F, Farr MA, Sayer G, Raikelkar J, Uriel N. Impact of Pretransplant Malignancy on Heart Transplantation Outcomes: Contemporary United Network for Organ Sharing Analysis Amidst Evolving Cancer Therapies. Circ Heart Fail 2022; 15:e008968. [PMID: 35094567 DOI: 10.1161/circheartfailure.121.008968] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND An aging population and improved cancer survivorship have increased the number of individuals with treated malignancy who develop advanced heart failure. The benefits of heart transplantation (HT) in patients with a pretransplant malignancy (PTM) must be balanced against risks of posttransplant malignancy in the setting of immunosuppression. METHODS Adult patients in the United Network for Organ Sharing registry who received HT between January 1, 2010, and December 31, 2020 were included. Trends, patient characteristics, and posttransplant outcomes in HT recipients with PTM were evaluated. RESULTS From 2000 to 2020, the proportion of HT recipients with PTM increased from 3.2% to 8.2%. From 2010 to 2020, 2113 (7.7%) of 27 344 HT recipients had PTM. PTM was associated with higher rates of 1-year mortality after HT (11.9% versus 9.2%; adjusted hazard ratio, 1.25 [95% CI, 1.09-1.44], P=0.001), driven by increased mortality in patients with hematologic PTM (adjusted hazard ratio, 2.00 [95% CI, 1.61-2.48]; P<0.001). For recipients who survived the first year, 5-year survival was similar between patients with and without PTM. Rates of malignancy at 5-years posttransplant were higher in the PTM group (20.4% versus 13.1%; adjusted hazard ratio, 1.57 [95% CI, 1.38-1.79], P<0.001). CONCLUSIONS Prevalence of PTM in HT recipients nearly tripled over the past 2 decades. Patients with hematologic PTM were at increased risk of early mortality after HT. Patients with PTM were also at higher risk for posttransplant malignancy. Guidelines that reflect contemporary oncological care are needed to inform care of this heterogenous and expanding group of individuals.
Collapse
Affiliation(s)
- Jaya Batra
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Ersilia M DeFilippis
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Stephanie Golob
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Kevin Clerkin
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Veli K Topkara
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Marlena V Habal
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Susan Restaino
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Jan Griffin
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Sun Hi Lee
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Farhana Latif
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Maryjane A Farr
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Gabriel Sayer
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Jayant Raikelkar
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| | - Nir Uriel
- Division of Cardiology, Department of Medicine, Columbia University College of Physicians and Surgeons, New York, NY
| |
Collapse
|
34
|
Bottomley MJ, Brook MO, Shankar S, Hester J, Issa F. Towards regulatory cellular therapies in solid organ transplantation. Trends Immunol 2022; 43:8-21. [PMID: 34844848 DOI: 10.1016/j.it.2021.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/10/2021] [Accepted: 11/01/2021] [Indexed: 01/03/2023]
Abstract
Organ transplantation is a modern medical success story. However, since its inception it has been limited by the need for pharmacological immunosuppression. Regulatory cellular therapies offer an attractive solution to these challenges by controlling transplant alloresponses through multiple parallel suppressive mechanisms. A number of cell types have seen an accelerated development into human trials and are now on the threshold of a long-awaited breakthrough in personalized transplant therapeutics. Here we assess recent developments with a focus on the most likely candidates, some of which have already facilitated successful immunosuppression withdrawal in early clinical trials. We propose that this may constitute a promising approach in clinical transplantation but also evaluate outstanding issues in the field, providing cause for cautious optimism.
Collapse
Affiliation(s)
- Matthew J Bottomley
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Matthew O Brook
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Sushma Shankar
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK; Oxford Transplant Centre, Churchill Hospital, Oxford, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.
| |
Collapse
|
35
|
Krendl FJ, Messner F, Bösmüller C, Scheidl S, Cardini B, Resch T, Weissenbacher A, Oberhuber R, Maglione M, Schneeberger S, Öfner D, Margreiter C. Post-Transplant Malignancies following Pancreas Transplantation: Incidence and Implications on Long-Term Outcome from a Single-Center Perspective. J Clin Med 2021; 10:jcm10214810. [PMID: 34768331 PMCID: PMC8584646 DOI: 10.3390/jcm10214810] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/12/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022] Open
Abstract
Chronic immunosuppression is associated with an increased risk of malignancy. The main objective of this study is to evaluate the incidence and effect of post-transplant malignancies (PTMs) following pancreas transplantation. The 348 first pancreas transplants performed between 1985 and 2015 were retrospectively analyzed in this study. Incidences of PTMs, as well as patient and graft survival, were evaluated. Out of 348 patients, 71 (20.4%) developed a PTM. Median time to diagnosis was 130 months. Thirty-six patients (50.7%) developed skin cancers (four patients with melanoma, 32 with NMSCs). Solid organ malignancy occurred in 25 (35.2%), hematologic malignancy in ten patients (14.1%). Affected patients were transplanted earlier [2000 (IQR 1993−2004) vs. 2003 (IQR 1999−2008); p < 0.001]. No differences in induction therapy were seen, both groups demonstrated comparable patient and graft survival. Pancreas transplant recipients with solid organ and hematologic malignancies had a three- and six-fold increased hazard of death compared to those with skin cancers [aHR 3.04 (IQR 1.17–7.91); p = 0.023; aHR 6.07 (IQR 1.87–19.71); p = 0.003]. PTMs affect every fifth patient following pancreas transplantation. Skin cancers are the most common malignancies accounting for 50% of all PTMs. These results underscore the importance of close dermatologic follow-up.
Collapse
|
36
|
Shirazian S, Starakiewicz P, Latcha S. Cancer Screening in End-Stage Kidney Disease. Adv Chronic Kidney Dis 2021; 28:502-508.e1. [PMID: 35190116 DOI: 10.1053/j.ackd.2021.09.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 09/14/2021] [Indexed: 11/11/2022]
Abstract
The incidence of cancer is higher in patients with end-stage kidney disease (ESKD) than among the general population. Despite this, screening for cancer is generally not cost-effective and may worsen quality of life in these patients. This is due to high mortality rates (patients are not living long enough to reap the benefits of screening), the inaccuracy of cancer screening tests, and the increased risks associated with therapy in patients with ESKD. Specific groups of patients with ESKD who have a longer-than-expected life expectancy or higher-than-expected cancer risk may benefit from screening. These groups include patients on peritoneal dialysis, patients on home hemodialysis, Black and Asian-American patients, transplant-eligible patients, and those at higher risk of cancer including patients with acquired cystic kidney disease, those who have been previously exposed to cytotoxic agents or aristolochic acid, and patients with a genetic predisposition to cancer. In this narrative review, we will examine the prevalence of and risk factors for cancer in patients with ESKD and the effectiveness of cancer screening, and discuss specific situations in which cancer screening may be effective.
Collapse
|
37
|
Lee HH, Joung JY, Kim SH. The effect of subsequent immunosuppressant use in organ-transplanted patients on prostate cancer incidence: a retrospective analysis using the Korean National Health Insurance Database. BMC Urol 2021; 21:112. [PMID: 34419041 PMCID: PMC8379856 DOI: 10.1186/s12894-021-00883-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 08/16/2021] [Indexed: 02/06/2023] Open
Abstract
Backgrounds Prostate cancer (PC) is the most common solid organ cancer. However, there is still no definite consensus before and after organ transplantation (TPL). We aimed to analyze whether PC incidence increased in TPL patients with subsequent use of immunosuppressants using the Korean National Health Insurance Database. Methods TPL patients between 2003 and 2015(N = 12,970) were age- and year-matched to non-TPL patients (N = 38,910) in a 1:3 ratio. Multivariate Cox regression analysis adjusted for significant prognostic clinicopathological parameters, including the duration of immunosuppressant agent use (0–300 or > 300 days), and Kaplan–Meier analysis with log-rank test were used to evaluate the association of TPL with PC incidence between the groups. Results Median overall survival was 4.86 years; overall mortality rate was 3.4% (n = 1761). Regardless of differences in baseline characteristics between the groups, multivariate analysis for PC incidence showed that age, immunosuppressant use, and TPL organ subtypes were significant factors for the overall population, whereas only age was significant in the TPL group (p < 0.05). After adjusting for age, underlying disease, and prescribed medication (aspirin, statin), multiple subgroup analysis models for PC incidence were evaluated. PC incidence was increased in the TPL group (hazard ratio [HR] 1.965, p < 0.001); however, PC incidence in the TPL group became insignificant after adjusting for immunosuppressant use (p = 0.194). Kaplan–Meier curves also showed that PC incidence was significantly different according to age and TPL with the use of immunosuppressants between the TPL and non-TPL groups. Conclusions PC incidence was higher in the TPL group using immunosuppressants than in the non-TPL group. Trial registration: The study was retrospectively registered. Supplementary Information The online version contains supplementary material available at 10.1186/s12894-021-00883-8.
Collapse
Affiliation(s)
- Hyung Ho Lee
- Department of Urology, Urologic Cancer Center, Research Institute and Hospital of National Cancer Center, 323 Ilsanro Ilsandonggu Madoodong, Goyang, 10408, Republic of Korea
| | - Jae Young Joung
- Department of Urology, Urologic Cancer Center, Research Institute and Hospital of National Cancer Center, 323 Ilsanro Ilsandonggu Madoodong, Goyang, 10408, Republic of Korea
| | - Sung Han Kim
- Department of Urology, Urologic Cancer Center, Research Institute and Hospital of National Cancer Center, 323 Ilsanro Ilsandonggu Madoodong, Goyang, 10408, Republic of Korea.
| |
Collapse
|
38
|
Kim B, Kang M, Kim Y, Lee HS, Kim B, Lee JJ, Park Y, Lee KA. De Novo Cancer Incidence after Kidney Transplantation in South Korea from 2002 to 2017. J Clin Med 2021; 10:3530. [PMID: 34441826 PMCID: PMC8396914 DOI: 10.3390/jcm10163530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/05/2021] [Accepted: 08/10/2021] [Indexed: 12/26/2022] Open
Abstract
Advances in patient care and immunosuppressive drugs have improved graft survival, resulting in an increase in kidney transplantation (KT); however, persistent immunosuppression is thought to cause late occurrence of cancer. This population-based study consisted of a total of 14,842 patients whose data from the years 2002 to 2017 were collected from the National Health Information Database in South Korea. Malignancies occurred in 7.6% of the total KT patients. Prostate and thyroid cancers were the most common in males and females, respectively. From the age-adjusted incidence analysis, Kaposi's sarcoma showed the highest standardized incidence ratio in both male and female patients. According to the linear regression model, cancer incidence in KT recipients under immunosuppressive conditions increased by approximately 0.1% each month. Patients' age over 39 and the use of prednisolone as an initial steroid regimen were associated with increased risk of cancer development after KT. Our regression and proportional hazards models will help clinicians to predict the approximate cancer incidence risk when monitoring KT recipients. Based on the largest available national database, screening or monitoring methods for cancer detection and prevention can be established for KT patients by considering the factors involved in cancer development.
Collapse
Affiliation(s)
- Boyeon Kim
- Department of Laboratory Medicine, National Health Insurance Service Ilsan Hospital, Goyang-si 10444, Korea; (B.K.); (B.K.)
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (Y.K.); (K.-A.L.)
| | - Minjin Kang
- Research Institute, National Health Insurance Service Ilsan Hospital, Goyang 10444, Korea;
| | - Yoonjung Kim
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (Y.K.); (K.-A.L.)
| | - Hyung Soon Lee
- Department of Surgery, National Health Insurance Service Ilsan Hospital, Goyang 10444, Korea;
| | - Banseok Kim
- Department of Laboratory Medicine, National Health Insurance Service Ilsan Hospital, Goyang-si 10444, Korea; (B.K.); (B.K.)
| | - Jung Jun Lee
- Department of Surgery, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea
| | - Yongjung Park
- Department of Laboratory Medicine, National Health Insurance Service Ilsan Hospital, Goyang-si 10444, Korea; (B.K.); (B.K.)
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (Y.K.); (K.-A.L.)
| | - Kyung-A Lee
- Department of Laboratory Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Korea; (Y.K.); (K.-A.L.)
| |
Collapse
|
39
|
Zheng G, Sundquist K, Sundquist J, Chen T, Försti A, Hemminki A, Liska V, Hemminki K. Second Primary Cancers After Liver, Gallbladder and Bile Duct Cancers, and These Cancers as Second Primary Cancers. Clin Epidemiol 2021; 13:683-691. [PMID: 34377034 PMCID: PMC8349530 DOI: 10.2147/clep.s318737] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background Second primary cancers (SPCs) are important clinically as they may negatively influence patient survival and they may tell about therapeutic side effects and general causes of cancer. Population-based literature concerning SPCs after hepatobiliary cancers is limited and here we assess risks of SPCs after hepatocellular cancer (HCC), and cancers of the gallbladder, bile ducts and ampulla of Vater. In reverse order, we consider the risk of hepatobiliary cancers as SPCs after any cancer. Methods We used standardized incidence ratios (SIRs) to estimate bidirectional relative risks of subsequent cancers associated with hepatobiliary cancers. Cancer diagnoses were obtained from the Swedish Cancer Registry from years 1990 through 2015. Results We identified 9997 primary HCCs, 1365 gallbladder cancers and 4721 bile duct cancers. After HCC, risks of four SPCs were increased: gallbladder (SIR = 4.38; 95% confidence interval 1.87-8.67), thyroid (4.13; 1.30-9.70), kidney (2.92; 1.66-4.47) and squamous cell skin (1.55; 1.02-2.26) cancers. In reverse order, HCC as SPC, in addition to the above cancers, associations included upper aerodigestive tract, esophageal, small intestinal and bladder cancers and non-Hodgkin lymphoma. For gallbladder and bile duct cancers, associations were found with small intestinal and pancreatic cancers. Conclusion The results suggested that HCC is associated with two types of SPC, one related to shared environmental risk factors, such as alcohol, exemplified by upper aerodigestive tract and esophageal cancer, and the other related to immune dysfunction, exemplified by squamous cell skin cancer. SPCs associated with gallbladder and bile duct cancers suggest predisposition to mutations in the mismatch repair gene MLH1.
Collapse
Affiliation(s)
- Guoqiao Zheng
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany.,Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany.,Center for Primary Health Care Research, Lund University, Malmö, 205 02, Sweden
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, 205 02, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, 205 02, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Center for Community-based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Japan
| | - Tianhui Chen
- Department of Cancer Prevention, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Vaclav Liska
- Department of Surgery, University Hospital, School of Medicine in Pilsen, Pilsen, Czech Republic.,Biomedical Center, Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, Pilsen, 30605, Czech Republic
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany.,Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, D-69120, Germany.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Biomedical Center, Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, Pilsen, 30605, Czech Republic
| |
Collapse
|
40
|
Bilek O, Holanek M, Jurica J, Stepankova S, Vasina J, Selingerova I, Poprach A, Borilova S, Kazda T, Kiss I, Zdrazilova-Dubska L. Drug interaction profile of TKI alectinib allows effective and safe treatment of ALK+ lung cancer in the kidney transplant recipient. Int Immunopharmacol 2021; 99:108012. [PMID: 34339964 DOI: 10.1016/j.intimp.2021.108012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/08/2021] [Accepted: 07/20/2021] [Indexed: 01/15/2023]
Abstract
ALK targeting with tyrosine kinase inhibitors (TKIs) is a highly potent treatment option for the therapy of ALK positive non-small cell lung cancer (NSCLC). However, pharmacokinetics of TKIs leads to clinically significant drug interactions, and the interfering co-medication may hamper the anti-cancer therapeutic management. Here, we present for the first time a drug interaction profile of ALK-TKIs, crizotinib and alectinib, and immunosuppressive agent cyclosporine A in kidney transplant recipients diagnosed with ALK+ lung cancer. Based on therapeutic drug monitoring of cyclosporin A plasma level, the dose of cyclosporine A has been adjusted to achieve a safe and effective therapeutic level in terms of both cancer treatment and kidney transplant condition. Particularly, 15 years upon the kidney transplantation, the stage IV lung cancer patient was treated with the 1st-line chemotherapy, the 2nd-line ALK-TKI crizotinib followed by ALK-TKI alectinib. The successful therapy with ALK-TKIs has been continuing for more than 36 months, including the period when the patient was treated for COVID-19 bilateral pneumonia. Hence, the therapy of ALK+ NSCLC with ALK-TKIs in organ transplant recipients treated with cyclosporine A may be feasible and effective.
Collapse
Affiliation(s)
- Ondrej Bilek
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic; Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic
| | - Milos Holanek
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic
| | - Jan Jurica
- Department of Pharmacology, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic; Hospital Pharmacy, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic
| | - Sona Stepankova
- Center of Cardiovascular and Transplant Surgery, Pekarska 53, 656 91 Brno, the Czech Republic
| | - Jiri Vasina
- Department of Nuclear Medicine, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic
| | - Iveta Selingerova
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Pharmacology, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic
| | - Alexandr Poprach
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic
| | - Simona Borilova
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic
| | - Tomas Kazda
- Department of Radiation oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Radiation oncology, Faculty of Medicine, Faculty of Medicine, Masaryk University, Brno, 625 00 Brno, the Czech Republic
| | - Igor Kiss
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Comprehensive Cancer Care, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic
| | - Lenka Zdrazilova-Dubska
- Research Centre for Applied Molecular Oncology, Masaryk Memorial Cancer Institute, 656 53 Brno, the Czech Republic; Department of Pharmacology, Faculty of Medicine, Masaryk University, 625 00 Brno, the Czech Republic; Department of Clinical Microbiology and Immunology, University Hospital Brno, Jihlavská 25, 625 00 Brno, the Czech Republic.
| |
Collapse
|
41
|
Zheng G, Sundquist K, Sundquist J, Chen T, Försti A, Hemminki A, Hemminki K. Second Primary Cancers After Gastric Cancer, and Gastric Cancer as Second Primary Cancer. Clin Epidemiol 2021; 13:515-525. [PMID: 34239328 PMCID: PMC8260108 DOI: 10.2147/clep.s304332] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/01/2021] [Indexed: 12/15/2022] Open
Abstract
Background Second primary cancers (SPCs) are increasing, which may negatively influence patient survival. Gastric cancer (GC) has poor survival and when it is diagnosed as SPC it is often the cause of death. We wanted to analyze the risk of SPCs after GC and the risk of GC as SPC after any cancer. Such bidirectional analysis is important in relation to fatal cancers because SPCs may be under-reported in the short-term survival period. Methods Cancers were obtained from the Swedish Cancer Registry from years 1990 through 2015. Standardized incidence ratios (SIRs) were used to estimate bidirectional relative. Results We identified 23,137 GC patients who developed 1042 SPCs (4.5%); 2158 patients had GC as SPC. While the risk for three SPCs was increased after GC, seven first primary cancers were followed by an increased risk of GC as SPC, including esophageal, colorectal, bladder, squamous cell skin and breast cancers and non-Hodgkin lymphoma. Breast cancer, which was followed by a diagnosis of second GC, showed an excess of lobular histology. Conclusion Multiple primary cancers in the same individuals may signal genetic predisposition. Accordingly, the association of GC with breast cancer may be related to mutations in the CDH1 gene, and clustering of colorectal, small intestinal and bladder cancers could be related to Lynch syndrome. The third line of findings supports a contribution of immune dysfunction on the increased risk of GC as SPC after skin cancer and non-Hodgkin lymphoma. Early detection of GC in the risk groups could save lives.
Collapse
Affiliation(s)
- Guoqiao Zheng
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Department of Family Medicine and Community Health.,Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Department of Family Medicine and Community Health.,Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Center for Community-Based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Matsue, Shimane, Japan
| | - Tianhui Chen
- Department of Cancer Prevention, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, 310022, People's Republic of China
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Akseli Hemminki
- Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Comprehensive Cancer Center, Helsinki University Hospital, Helsinki, Finland
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Biomedical Center, Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, Pilsen, 30605, Czech Republic
| |
Collapse
|
42
|
Zheng G, Sundquist K, Sundquist J, Försti A, Hemminki O, Hemminki K. Bladder and upper urinary tract cancers as first and second primary cancers. Cancer Rep (Hoboken) 2021; 4:e1406. [PMID: 34114732 PMCID: PMC8714543 DOI: 10.1002/cnr2.1406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/29/2021] [Accepted: 04/09/2021] [Indexed: 11/24/2022] Open
Abstract
Background Previous population‐based studies on second primary cancers (SPCs) in urothelial cancers have focused on known risk factors in bladder cancer patients without data on other urothelial sites of the renal pelvis or ureter. Aims To estimate sex‐specific risks for any SPCs after urothelial cancers, and in reverse order, for urothelial cancers as SPCs after any cancer. Such two‐way analysis may help interpret the results. Methods We employed standardized incidence ratios (SIRs) to estimate bidirectional relative risks of subsequent cancer associated with urothelial cancers. Patient data were obtained from the Swedish Cancer Registry from years 1990 through 2015. Results We identified 46 234 urinary bladder cancers (75% male), 940 ureteral cancers (60% male), and 2410 renal pelvic cancers (57% male). After male bladder cancer, SIRs significantly increased for 9 SPCs, most for ureteral (SIR 41.9) and renal pelvic (17.2) cancers. In the reversed order (bladder cancer as SPC), 10 individual FPCs were associated with an increased risk; highest associations were noted after renal pelvic (21.0) and ureteral (20.9) cancers. After female bladder cancer, SIRs of four SPCs were significantly increased, most for ureteral (87.8) and pelvic (35.7) cancers. Female bladder, ureteral, and pelvic cancers associated are with endometrial cancer. Conclusions The risks of recurrent urothelial cancers were very high, and, at most sites, female risks were twice over the male risks. Risks persisted often to follow‐up periods of >5 years, motivating an extended patient follow‐up. Lynch syndrome‐related cancers were associated with particularly female urothelial cancers, calling for clinical vigilance.
Collapse
Affiliation(s)
- Guoqiao Zheng
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kristina Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Center for Community-Based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Shimane, Japan
| | - Jan Sundquist
- Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Department of Family Medicine and Community Health, Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, New York, USA.,Center for Community-Based Healthcare Research and Education (CoHRE), Department of Functional Pathology, School of Medicine, Shimane University, Shimane, Japan
| | - Asta Försti
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Hopp Children's Cancer Center (KiTZ), Heidelberg, Germany.,Division of Pediatric Neurooncology, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Otto Hemminki
- Department of Surgery, University Health Network, Toronto, Ontario, Canada.,Department of Urology, Helsinki University Hospital, Helsinki, Finland.,Cancer Gene Therapy Group, Translational Immunology Research Program, University of Helsinki, Helsinki, Finland
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Division of Cancer Epidemiology, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center for Primary Health Care Research, Lund University, Malmö, Sweden.,Biomedical Center, Faculty of Medicine and Biomedical Center in Pilsen, Charles University in Prague, Pilsen, Czech Republic
| |
Collapse
|
43
|
Namestnikov M, Pleniceanu O, Dekel B. Mixing Cells for Vascularized Kidney Regeneration. Cells 2021; 10:1119. [PMID: 34066487 PMCID: PMC8148539 DOI: 10.3390/cells10051119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/26/2021] [Accepted: 05/01/2021] [Indexed: 02/06/2023] Open
Abstract
The worldwide rise in prevalence of chronic kidney disease (CKD) demands innovative bio-medical solutions for millions of kidney patients. Kidney regenerative medicine aims to replenish tissue which is lost due to a common pathological pathway of fibrosis/inflammation and rejuvenate remaining tissue to maintain sufficient kidney function. To this end, cellular therapy strategies devised so far utilize kidney tissue-forming cells (KTFCs) from various cell sources, fetal, adult, and pluripotent stem-cells (PSCs). However, to increase engraftment and potency of the transplanted cells in a harsh hypoxic diseased environment, it is of importance to co-transplant KTFCs with vessel forming cells (VFCs). VFCs, consisting of endothelial cells (ECs) and mesenchymal stem-cells (MSCs), synergize to generate stable blood vessels, facilitating the vascularization of self-organizing KTFCs into renovascular units. In this paper, we review the different sources of KTFCs and VFCs which can be mixed, and report recent advances made in the field of kidney regeneration with emphasis on generation of vascularized kidney tissue by cell transplantation.
Collapse
Affiliation(s)
- Michael Namestnikov
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- ediatric Nephrology Division, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
| | - Oren Pleniceanu
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
- The Kidney Research Lab, Institute of Nephrology and Hypertension, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel Hashomer, Ramat Gan 52621, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Ramat Aviv, Tel Aviv 69978, Israel;
| |
Collapse
|
44
|
Liauw SL, Ham SA, Das LC, Rudra S, Packiam VT, Koshy M, Weichselbaum RR, Becker YT, Bodzin AS, Eggener SE. Prostate Cancer Outcomes Following Solid-Organ Transplantation: A SEER-Medicare Analysis. J Natl Cancer Inst 2021; 112:847-854. [PMID: 31728517 DOI: 10.1093/jnci/djz221] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 10/01/2019] [Accepted: 11/01/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Immunosuppressive regimens associated with organ transplantation increase the risk of developing cancer. Transplant candidates and recipients with prostate cancer are often treated, even if low-risk features would ordinarily justify active surveillance. METHODS Using SEER-Medicare, we identified 163 676 men aged 66 years and older diagnosed with nonmetastatic prostate cancer. History of solid organ transplant was identified using diagnosis or procedure codes. A propensity score-matched cohort was identified by matching transplanted men to nontransplanted controls by age, race, region, year, T-stage, grade, comorbidity, and cancer therapy. Fine-Gray competing risk models assessed associations between transplant status and prostate cancer-specific mortality (PCSM) and overall mortality (OM). RESULTS We identified 620 men (0.4%) with transplant up to 10 years before (n = 320) or 5 years after (n = 300) prostate cancer diagnosis and matched them to 3100 men. At 10 years, OM was 55.7% and PCSM was 6.0% in the transplant cohort compared with 42.4% (P < .001) and 7.6% (P = .70) in the nontransplant cohort, respectively. Adjusted models showed no difference in PCSM for transplanted men (hazard ratio = 0.88, 95% confidence interval = 0.61 to 1.27, P = .70) or differences by prostate cancer therapy. Among 334 transplanted men with T1-2N0, well or moderately differentiated "low-risk" prostate cancer, PCSM was similar for treated and untreated men (hazard ratio = 0.92, 95% confidence interval = 0.47 to 1.81). CONCLUSIONS Among men aged 66 years and older with prostate cancer, an organ transplant is associated with higher OM but no observable difference in PCSM. These findings suggest men with prostate cancer and previous or future organ transplantation should be managed per usual standards of care, including consideration of active surveillance for low-risk cancer characteristics.
Collapse
Affiliation(s)
| | | | - Lauren C Das
- Department of Radiation and Cellular Oncology, Beacon Health System, Elkhart, IN
| | - Sonali Rudra
- University of Chicago, Chicago, IL.,Department of Radiation Oncology, Georgetown University, Washington, DC
| | | | | | | | - Yolanda T Becker
- Department of Transplant Surgery, University of Chicago, Chicago, IL
| | - Adam S Bodzin
- Department of Transplant Surgery, Jefferson University Hospitals, Philadelphia, PA
| | | |
Collapse
|
45
|
Johnstone BH, Messner F, Brandacher G, Woods EJ. A Large-Scale Bank of Organ Donor Bone Marrow and Matched Mesenchymal Stem Cells for Promoting Immunomodulation and Transplant Tolerance. Front Immunol 2021; 12:622604. [PMID: 33732244 PMCID: PMC7959805 DOI: 10.3389/fimmu.2021.622604] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Induction of immune tolerance for solid organ and vascular composite allografts is the Holy Grail for transplantation medicine. This would obviate the need for life-long immunosuppression which is associated with serious adverse outcomes, such as infections, cancers, and renal failure. Currently the most promising means of tolerance induction is through establishing a mixed chimeric state by transplantation of donor hematopoietic stem cells; however, with the exception of living donor renal transplantation, the mixed chimerism approach has not achieved durable immune tolerance on a large scale in preclinical or clinical trials with other solid organs or vascular composite allotransplants (VCA). Ossium Health has established a bank of cryopreserved bone marrow (BM), termed "hematopoietic progenitor cell (HPC), Marrow," recovered from deceased organ donor vertebral bodies. This new source for hematopoietic cell transplant will be a valuable resource for treating hematological malignancies as well as for inducing transplant tolerance. In addition, we have discovered and developed a large source of mesenchymal stem (stromal) cells (MSC) tightly associated with the vertebral body bone fragment byproduct of the HPC, Marrow recovery process. Thus, these vertebral bone adherent MSC (vBA-MSC) are matched to the banked BM obtained from each donor, as opposed to third-party MSC, which enhances safety and potentially efficacy. Isolation and characterization of vBA-MSC from over 30 donors has demonstrated that the cells are no different than traditional BM-MSC; however, their abundance is >1,000-fold higher than obtainable from living donor BM aspirates. Based on our own unpublished data as well as reports published by others, MSC facilitate chimerism, especially at limiting hematopoietic stem and progenitor cell (HSPC) numbers and increase safety by controlling and/or preventing graft-vs.-host-disease (GvHD). Thus, vBA-MSC have the potential to facilitate mixed chimerism, promote complementary peripheral immunomodulatory functions and increase safety of BM infusions. Both HPC, Marrow and vBA-MSC have potential use in current VCA and solid organ transplant (SOT) tolerance clinical protocols that are amenable to "delayed tolerance." Current trials with HPC, Marrow are planned with subsequent phases to include vBA-MSC for tolerance of both VCA and SOT.
Collapse
Affiliation(s)
- Brian H. Johnstone
- Ossium Health, Indianapolis, IN, United States
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, United States
| | - Franka Messner
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Visceral, Transplant and Thoracic Surgery, Center of Operative Medicine, Medical University of Innsbruck, Innsbruck, Austria
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Erik J. Woods
- Ossium Health, Indianapolis, IN, United States
- Department of Biomedical Sciences, College of Osteopathic Medicine, Marian University, Indianapolis, IN, United States
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
46
|
Podestà MA, Remuzzi G, Casiraghi F. Mesenchymal Stromal Cell Therapy in Solid Organ Transplantation. Front Immunol 2021; 11:618243. [PMID: 33643298 PMCID: PMC7902912 DOI: 10.3389/fimmu.2020.618243] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/29/2020] [Indexed: 12/29/2022] Open
Abstract
Transplantation is the gold-standard treatment for the failure of several solid organs, including the kidneys, liver, heart, lung and small bowel. The use of tailored immunosuppressive agents has improved graft and patient survival remarkably in early post-transplant stages, but long-term outcomes are frequently unsatisfactory due to the development of chronic graft rejection, which ultimately leads to transplant failure. Moreover, prolonged immunosuppression entails severe side effects that severely impact patient survival and quality of life. The achievement of tolerance, i.e., stable graft function without the need for immunosuppression, is considered the Holy Grail of the field of solid organ transplantation. However, spontaneous tolerance in solid allograft recipients is a rare and unpredictable event. Several strategies that include peri-transplant administration of non-hematopoietic immunomodulatory cells can safely and effectively induce tolerance in pre-clinical models of solid organ transplantation. Mesenchymal stromal cells (MSC), non-hematopoietic cells that can be obtained from several adult and fetal tissues, are among the most promising candidates. In this review, we will focus on current pre-clinical evidence of the immunomodulatory effect of MSC in solid organ transplantation, and discuss the available evidence of their safety and efficacy in clinical trials.
Collapse
Affiliation(s)
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Bergamo, Italy
| | - Federica Casiraghi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Aldo & Cele Daccò Clinical Research Center for Rare Diseases, Bergamo, Italy
| |
Collapse
|
47
|
Uterine Malakoplakia Identified by Diagnostic Hysteroscopy. J Minim Invasive Gynecol 2020; 28:924-926. [PMID: 33238208 DOI: 10.1016/j.jmig.2020.11.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/12/2020] [Accepted: 11/14/2020] [Indexed: 11/23/2022]
|
48
|
Hu Y, Kong D, Qin Y, Yu D, Jin W, Li X, Zhao Y, Wang H, Li G, Hao J, Zhang B, Pang Z, Wang H. CD73 expression is critical to therapeutic effects of human endometrial regenerative cells in inhibition of cardiac allograft rejection in mice. Stem Cells Transl Med 2020; 10:465-478. [PMID: 33124777 PMCID: PMC7900594 DOI: 10.1002/sctm.20-0154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 08/23/2020] [Accepted: 10/05/2020] [Indexed: 12/20/2022] Open
Abstract
The newly found mesenchymal‐like endometrial regenerative cells (ERCs) have been proved to induce immune tolerance in cardiac allograft transplantation. However, the therapeutic mechanism is not clear. The present study was undertaken to investigate whether ecto‐5′‐nucleotidase (CD73) expression on ERCs is critical to cardiac allograft protection. C57BL/6 mouse recipients receiving BALB/c mouse cardiac allografts were treated with unmodified ERCs or anti‐CD73 monoclonal antibodies (mAb) pretreated ERCs, respectively. It has been found that CD73 expression was critical to ERC‐induced attenuation of graft pathology. The blockage of CD73 expression on ERCs was related to the percentage decline of tolerogenic dendritic cells (Tol‐DCs), macrophages type 2 (M2), and regulatory T cells (Tregs). As compared with anti‐CD73 mAb pretreated ERCs group, CD73 expressing ERCs significantly increased the level of anti‐inflammatory cytokine IL‐10 but decreased levels of pro‐inflammatory cytokines including IFN‐γ and TNF‐α. In addition, CD73 expressing ERCs showed tissue protective function via the regulation of adenosine receptor expression which was related to the infiltration of CD4+ and CD8+ cells in the allografts. Furthermore, significant increase of A2B receptors in the cardiac allograft was also associated with CD73 expressing ERC‐induced prolongation of cardiac allograft survival.
Collapse
Affiliation(s)
- Yonghao Hu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Dejun Kong
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Yafei Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Dingding Yu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Wang Jin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Yiming Zhao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Hongda Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Guangming Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Jingpeng Hao
- Tianjin General Surgery Institute, Tianjin, People's Republic of China.,Department of Anorectal Surgery, The Second Hospital of Tianjin Medical University, Tianjin, People's Republic of China
| | - Baoren Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| | - Zhaoyan Pang
- Department of Nursing, China-Japan Friendship Hospital, Beijing, People's Republic of China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, People's Republic of China.,Tianjin General Surgery Institute, Tianjin, People's Republic of China
| |
Collapse
|
49
|
Roemhild A, Otto NM, Moll G, Abou-El-Enein M, Kaiser D, Bold G, Schachtner T, Choi M, Oellinger R, Landwehr-Kenzel S, Juerchott K, Sawitzki B, Giesler C, Sefrin A, Beier C, Wagner DL, Schlickeiser S, Streitz M, Schmueck-Henneresse M, Amini L, Stervbo U, Babel N, Volk HD, Reinke P. Regulatory T cells for minimising immune suppression in kidney transplantation: phase I/IIa clinical trial. BMJ 2020; 371:m3734. [PMID: 33087345 PMCID: PMC7576328 DOI: 10.1136/bmj.m3734] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To assess whether reshaping of the immune balance by infusion of autologous natural regulatory T cells (nTregs) in patients after kidney transplantation is safe, feasible, and enables the tapering of lifelong high dose immunosuppression, with its limited efficacy, adverse effects, and high direct and indirect costs, along with addressing several key challenges of nTreg treatment, such as easy and robust manufacturing, danger of over immunosuppression, interaction with standard care drugs, and functional stability in an inflammatory environment in a useful proof-of-concept disease model. DESIGN Investigator initiated, monocentre, nTreg dose escalation, phase I/IIa clinical trial (ONEnTreg13). SETTING Charité-University Hospital, Berlin, Germany, within the ONE study consortium (funded by the European Union). PARTICIPANTS Recipients of living donor kidney transplant (ONEnTreg13, n=11) and corresponding reference group trial (ONErgt11-CHA, n=9). INTERVENTIONS CD4+ CD25+ FoxP3+ nTreg products were given seven days after kidney transplantation as one intravenous dose of 0.5, 1.0, or 2.5-3.0×106 cells/kg body weight, with subsequent stepwise tapering of triple immunosuppression to low dose tacrolimus monotherapy until week 48. MAIN OUTCOME MEASURES The primary clinical and safety endpoints were assessed by a composite endpoint at week 60 with further three year follow-up. The assessment included incidence of biopsy confirmed acute rejection, assessment of nTreg infusion related adverse effects, and signs of over immunosuppression. Secondary endpoints addressed allograft functions. Accompanying research included a comprehensive exploratory biomarker portfolio. RESULTS For all patients, nTreg products with sufficient yield, purity, and functionality could be generated from 40-50 mL of peripheral blood taken two weeks before kidney transplantation. None of the three nTreg dose escalation groups had dose limiting toxicity. The nTreg and reference groups had 100% three year allograft survival and similar clinical and safety profiles. Stable monotherapy immunosuppression was achieved in eight of 11 (73%) patients receiving nTregs, while the reference group remained on standard dual or triple drug immunosuppression (P=0.002). Mechanistically, the activation of conventional T cells was reduced and nTregs shifted in vivo from a polyclonal to an oligoclonal T cell receptor repertoire. CONCLUSIONS The application of autologous nTregs was safe and feasible even in patients who had a kidney transplant and were immunosuppressed. These results warrant further evaluation of Treg efficacy and serve as the basis for the development of next generation nTreg approaches in transplantation and any immunopathologies. TRIAL REGISTRATION NCT02371434 (ONEnTreg13) and EudraCT:2011-004301-24 (ONErgt11).
Collapse
Affiliation(s)
- Andy Roemhild
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Natalie Maureen Otto
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Guido Moll
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mohamed Abou-El-Enein
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Daniel Kaiser
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Gantuja Bold
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Schachtner
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mira Choi
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Robert Oellinger
- Department of Abdominal and Transplant Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sybille Landwehr-Kenzel
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Karsten Juerchott
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Cordula Giesler
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anett Sefrin
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Carola Beier
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Dimitrios Laurin Wagner
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Stephan Schlickeiser
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Streitz
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Schmueck-Henneresse
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Leila Amini
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
| | - Ulrik Stervbo
- Medical Department 1, University hospitals of the Ruhr University of Bochum, Herne, Germany
| | - Nina Babel
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
- Medical Department 1, University hospitals of the Ruhr University of Bochum, Herne, Germany
| | - Hans-Dieter Volk
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Institute of Medical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Petra Reinke
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Centre for Advanced Therapies (BeCAT), Charité Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburger Platz 1, D-13353 Berlin, Germany
- Department of Nephrology and Internal Intensive Care Medicine, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
50
|
Repair of acute liver damage with immune evasive hESC derived hepato-blasts. Stem Cell Res 2020; 49:102010. [PMID: 33011360 DOI: 10.1016/j.scr.2020.102010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/23/2020] [Accepted: 09/21/2020] [Indexed: 01/20/2023] Open
Abstract
Human embryonic stem cells (hESCs) can undergo unlimited self-renewal and differentiate into hepatic cells, including expandable hepato-blasts (HBs) and hepatocyte-like cells (HLCs) in vitro. Therefore, hESC-derived HBs have the potential to become a renewable cell source for cell therapy of serious liver damage. However, one of the key challenges for such cell therapy is the allogeneic immune rejection of hESC-derived HBs. To overcome this challenge, we developed a strategy to protect the hESC-derived HBs from allogeneic immune rejection by ectopically expressing immune suppressive molecules CTLA4-Ig and PD-L1, denoted CP HBs. Like HBs derived from normal hESCs, CP HBs are capable of repairing liver damage in animal models. Using humanized mice (Hu-mice) reconstituted with human immune system, we showed that CP HBs are protected from allogeneic immune system and can survive long-term in Hu-mice. These data support the feasibility to develop CP HBs into a cell therapy to treat serious liver damage.
Collapse
|