1
|
Forbes S. β-Cell Benchmarks: Defining Predictive Outcomes in Islet Transplantation. Diabetes 2025; 74:685-688. [PMID: 40258167 PMCID: PMC12012584 DOI: 10.2337/dbi24-0054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 01/23/2025] [Indexed: 04/23/2025]
Affiliation(s)
- Shareen Forbes
- BHF Centre for Research Excellence, Queen’s Medical Research Institute, University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
2
|
Durgam SS, Rosado-Sánchez I, Yin D, Speck M, Mojibian M, Sayin I, Hynes GE, Alegre ML, Levings MK, Chong AS. CAR Treg synergy with anti-CD154 promotes infectious tolerance and dictates allogeneic heart transplant acceptance. JCI Insight 2025; 10:e188624. [PMID: 40197364 PMCID: PMC11981628 DOI: 10.1172/jci.insight.188624] [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: 11/01/2024] [Accepted: 02/18/2025] [Indexed: 04/10/2025] Open
Abstract
Successful allograft-specific tolerance induction would eliminate the need for daily immunosuppression and improve posttransplant quality of life. Adoptive cell therapy with regulatory T cells expressing donor-specific chimeric antigen receptors (CAR Tregs) is a promising strategy but, as monotherapy, cannot prolong survival with allografts with multiple MHC mismatches. Using an HLA-A2-transgenic haplo-mismatched heart transplantation model in immunocompetent C57BL/6 recipients, we showed that HLA-A2-specific CAR (A2.CAR) Tregs were able to synergize with a low dose of anti-CD154 to enhance graft survival. Using haplo-mismatched grafts expressing the 2W-OVA transgene and tetramer-based tracking of 2W- and OVA-specific T cells, we showed that in mice with accepted grafts, A2.CAR Tregs inhibited donor-specific T cell, B cell, and antibody responses and promoted a substantial increase in endogenous FOXP3+ Tregs with indirect donor specificity. By contrast, in mice where A2.CAR Tregs failed to prolong graft survival, FOXP3- A2.CAR T cells preferentially accumulated in rejecting allografts, and endogenous donor-specific responses were not controlled. This study therefore provides evidence for synergy between A2.CAR Tregs and CD154 blockade to promote infectious tolerance in immunocompetent recipients of haplo-mismatched heart grafts and defines features of A2.CAR Tregs when they fail to reshape host immunity toward allograft tolerance.
Collapse
Affiliation(s)
- Samarth S. Durgam
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Isaac Rosado-Sánchez
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dengping Yin
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Madeleine Speck
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Majid Mojibian
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ismail Sayin
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | - Grace E. Hynes
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| | | | - Megan K. Levings
- BC Children’s Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
| | - Anita S. Chong
- Department of Surgery, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
3
|
Mbaye EHA, Scott EA, Burke JA. From Edmonton to Lantidra and beyond: immunoengineering islet transplantation to cure type 1 diabetes. FRONTIERS IN TRANSPLANTATION 2025; 4:1514956. [PMID: 40182604 PMCID: PMC11965681 DOI: 10.3389/frtra.2025.1514956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025]
Abstract
Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing β cells within pancreatic islets, the specialized endocrine cell clusters of the pancreas. Islet transplantation has emerged as a β cell replacement therapy, involving the infusion of cadaveric islets into a patient's liver through the portal vein. This procedure offers individuals with T1D the potential to restore glucose control, reducing or even eliminating the need for exogenous insulin therapy. However, it does not address the underlying autoimmune condition responsible for T1D. The need for systemic immunosuppression remains the primary barrier to making islet transplantation a more widespread therapy for patients with T1D. Here, we review recent progress in addressing the key limitations of islet transplantation as a viable treatment for T1D. Concerns over systemic immunosuppression arise from its potential to cause severe side effects, including opportunistic infections, malignancies, and toxicity to transplanted islets. Recognizing the risks, the Edmonton protocol (2000) marked a shift away from glucocorticoids to prevent β cell damage specifically. This transition led to the development of combination immunosuppressive therapies and the emergence of less toxic immunosuppressive and anti-inflammatory drugs. More recent advances in islet transplantation derive from islet encapsulation devices, biomaterial platforms releasing immunomodulatory compounds or surface-modified with immune regulating ligands, islet engineering and co-transplantation with accessory cells. While most of the highlighted studies in this review remain at the preclinical stage using mouse and non-human primate models, they hold significant potential for clinical translation if a transdisciplinary research approach is prioritized.
Collapse
Affiliation(s)
- El Hadji Arona Mbaye
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
| | - Evan A. Scott
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, United States
- Department of Biomedical Engineering, NanoSTAR Institute, University of Virginia School of Medicine, Charlottesville, VA, United States
| | | |
Collapse
|
4
|
Rech Tondin A, Lanzoni G. Islet Cell Replacement and Regeneration for Type 1 Diabetes: Current Developments and Future Prospects. BioDrugs 2025; 39:261-280. [PMID: 39918671 PMCID: PMC11906537 DOI: 10.1007/s40259-025-00703-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2025] [Indexed: 03/14/2025]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by the destruction of insulin-producing beta cells in the pancreas, leading to insulin deficiency and chronic hyperglycemia. The main current therapeutic strategies for clinically overt T1D - primarily exogenous insulin administration combined with blood glucose monitoring - fail to fully mimic physiological insulin regulation, often resulting in suboptimal or insufficient glycemic control. Islet cell transplantation has emerged as a promising avenue for functionally replacing endogenous insulin production and achieving long-term glycemic stability. Here, we provide an overview of current islet replacement strategies, ranging from islet transplantation to stem cell-derived islet cell transplantation, and highlight emerging approaches such as immunoengineering. We examine the advancements in immunosuppressive protocols to enhance graft survival, innovative encapsulation, and immunomodulation techniques to protect transplanted islets, and the ongoing challenges in achieving durable and functional islet integration. Additionally, we discuss the latest clinical outcomes, the potential of gene editing technologies, and the emerging strategies for islet cell regeneration. This review aims to highlight the potential of these approaches to transform the management of T1D and improve the quality of life of individuals affected by this condition.
Collapse
Affiliation(s)
- Arthur Rech Tondin
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Giacomo Lanzoni
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
5
|
de Graav GN, Udomkarnjananun S, Baan CC, Reinders MEJ, Roodnat JI, de Winter BCM, Hesselink DA. New Developments and Therapeutic Drug Monitoring Options in Costimulatory Blockade in Solid Organ Transplantation: A Systematic Critical Review. Ther Drug Monit 2025; 47:64-76. [PMID: 39570574 DOI: 10.1097/ftd.0000000000001275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/31/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE In this review, the authors summarized the latest developments in costimulatory blockade to prevent rejection after solid organ transplantation (SOT) and discussed possibilities for future research and the need for therapeutic drug monitoring (TDM) of these agents. METHODS Studies about costimulatory blockers in SOT in humans or animal transplant models in the past decade (2014-2024) were systematically reviewed in PubMed, European Union clinical trials (EudraCT), and ClinicalTrials.gov . RESULTS Seventy-five registered clinical trials and 58 published articles were found on costimulation blockade of the CD28-CD80/86, CD40-CD40L, and OX40-OX40L pathways. Belatacept, an antagonist of the CD28-CD80/86 pathway, is the only approved costimulatory agent in SOT, hence accounting for most of the research. Other identified costimulatory blocking agents included abatacept and CD28 antagonists tegoprubart, dazodalibep, and TNX-1500. Although tegoprubart was unsuccessful in pancreas transplantation in nonhuman primates, trials in human kidney transplantation are underway. Dazodalibep trials faced recruitment challenges. TNX-1500 was unsuccessful in animal studies and is currently not pursued in humans. After discontinuation of iscalimab (CD40-CD154 pathway antagonist) in SOT, the alternatives, bleselumab and KPL404, showed promising results in kidney transplantation and cardiac xenotransplantation. Studies on secondary costimulatory pathway antagonists, such as OX40-OX40L, have only used animal models. Despite the low interindividual variability in pharmacokinetics (PK) in all studied agents, TDM could be useful for optimizing dosing in PK/pharmacodynamic (PD) studies. CONCLUSIONS The routine use of costimulation blockade in SOT is hindered by problems in efficacy compared with the standard of care. Costimulatory inhibitors could be combined in a calcineurin inhibitor-free regimen. Future PK/pharmacodynamic studies in costimulatory agents and personalized medicine could warrant TDM of these agents.
Collapse
Affiliation(s)
- Gretchen N de Graav
- Department of Internal Medicine, Division of Nephrology, Reinier de Graaf Gasthuis, Delft, the Netherlands
| | - Suwasin Udomkarnjananun
- Department of Medicine, Division of Nephrology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Carla C Baan
- Transplant Laboratory & Research Center, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Marlies E J Reinders
- Department of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands ; and
| | - Joke I Roodnat
- Department of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands ; and
| | - Brenda C M de Winter
- Department of Hospital Pharmacy, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Dennis A Hesselink
- Department of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center, Rotterdam, the Netherlands ; and
| |
Collapse
|
6
|
Griffith BP, Grazioli A, Singh AK, Tully A, Galindo J, Saharia KK, Shah A, Strauss ER, Odonkor PN, Williams B, Silverman HJ, Burke A, Drachenberg CB, Wells CL, Dickfeld T, Hong SN, Hicks AJ, Ananthram M, Gupta A, Christenson RH, Tamburro L, Zhang T, Hershfeld A, Lewis B, Feller ED, Kuravi K, Sorrells L, Morgand E, Mezine F, Goutaudier V, Rothblatt M, Lau CL, Taylor B, Perrin S, Loupy A, Ayares D, Mohiuddin MM. Transplantation of a genetically modified porcine heart into a live human. Nat Med 2025; 31:589-598. [PMID: 39779924 DOI: 10.1038/s41591-024-03429-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025]
Abstract
Following our previous experience with cardiac xenotransplantation of a genetically modified porcine heart into a live human, we sought to achieve improved results by selecting a healthier recipient and through more sensitive donor screening for potential zoonotic pathogens. Here we transplanted a 10-gene-edited pig heart into a 58-year-old man with progressive, debilitating inotrope-dependent heart failure due to ischemic cardiomyopathy who was not a candidate for standard advanced heart failure therapies. He was maintained on a costimulation (anti-CD40L, Tegoprubart) blockade-based immunomodulatory regimen. The xenograft initially functioned well, with excellent systolic and diastolic function during the first several weeks posttransplantation. Subsequently, the xenograft developed rapidly progressing diastolic heart failure, biventricular wall thickening and, ultimately, near-complete loss of systolic function necessitating initiation of extracorporeal membranous oxygenation on day 31. Given these setbacks, the patient chose to transition to comfort care after 40 days. As with our first patient, histology did not reveal substantial immune cell infiltration but suggested capillary endothelial injury with interstitial edema and early fibrosis. No evidence of porcine cytomegalovirus replication in the xenograft was observed. Strategies to overcome the obstacle of antibody-mediated rejection are needed to advance the field of xenotransplantation.
Collapse
Affiliation(s)
- Bartley P Griffith
- Cardiothoracic Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alison Grazioli
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Avneesh K Singh
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Andy Tully
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Javier Galindo
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kapil K Saharia
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aakash Shah
- Cardiothoracic Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Erik R Strauss
- Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Patrick N Odonkor
- Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brittney Williams
- Anesthesiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Allen Burke
- Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Chris L Wells
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Timm Dickfeld
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Susie N Hong
- University of Maryland School of Medicine, Baltimore, MD, USA
| | - Albert J Hicks
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Anuj Gupta
- University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Lo Tamburro
- Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tianshu Zhang
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alena Hershfeld
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Billeta Lewis
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | - Erwan Morgand
- Université de Paris Cité, INSERM U970, Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Fariza Mezine
- Université de Paris Cité, INSERM U970, Paris Translational Research Centre for Organ Transplantation, Paris, France
| | - Valentin Goutaudier
- Université Paris Cité, INSERM U970, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | | | - Christine L Lau
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Bradley Taylor
- Cardiothoracic Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | - Alexandre Loupy
- Université Paris Cité, INSERM U970, Paris Institute for Transplantation and Organ Regeneration, Paris, France
| | | | - Muhammad M Mohiuddin
- Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.
- Cardiothoracic Surgery, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
7
|
Anwar IJ, DeLaura I, Ladowski JM, Schilirò D, Gao Q, Manook M, Yoon J, Belloni R, Park A, Schuster DJ, Song M, Lin L, Farris AB, Magnani D, Williams K, Kwun J, Knechtle SJ. CD154 blockade effectively controls antibody-mediated rejection in highly sensitized nonhuman primate kidney transplant recipients. Sci Transl Med 2025; 17:eadn8130. [PMID: 39742504 PMCID: PMC11797747 DOI: 10.1126/scitranslmed.adn8130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 12/05/2024] [Indexed: 01/03/2025]
Abstract
Current desensitization and maintenance immunosuppression regimens for kidney transplantation in sensitized individuals show limited ability to control the posttransplant humoral response, resulting in high rates of antibody-mediated rejection (ABMR) and graft failure. Here, we showed that anti-CD154 monoclonal antibody (mAb)-based immunosuppression more effectively controlled allograft rejection and humoral rebound in a highly sensitized nonhuman primate kidney transplantation model compared with tacrolimus-based standard-of-care (SOC) immunosuppression. Desensitization with an anti-CD154 mAb (5C8) and a proteasome inhibitor led to decreased donor-specific antibodies (DSAs) and disruption of lymph node germinal centers with reduction of proliferating, memory, and class-switched B cells as well as T follicular helper cells. After transplant, the nonhuman primates maintained on 5C8-based immunosuppression had significantly better survival compared with those maintained on SOC immunosuppression (135.2 days versus 32.8 days, P = 0.013). The 5C8-treated group demonstrated better suppression of DSAs after transplant, more robust suppression of B cell populations, and better induction of regulatory T cells. Fewer infectious and welfare complications, including viral reactivation and weight loss, were also observed with 5C8-based immunosuppression compared with SOC immunosuppression. Therefore, anti-CD154 mAbs may improve kidney transplant outcomes through better control of posttransplant immune responses. The superior efficacy of anti-CD154 mAb-based immunosuppression over tacrolimus-based SOC seen in this highly sensitized NHP transplant model suggests that anti-CD154 mAbs could potentially be used to desensitize and treat highly sensitized patients receiving kidney transplantation.
Collapse
Affiliation(s)
- Imran J. Anwar
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Isabel DeLaura
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Joseph M. Ladowski
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Davide Schilirò
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Qimeng Gao
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Miriam Manook
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Janghoon Yoon
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Rafaela Belloni
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Angela Park
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Daniel J. Schuster
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Mingqing Song
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Lin Lin
- Department of Biostatistics & Bioinformatics, Duke University Medical Center, Durham, NC 27710, USA
| | - Alton B. Farris
- Department of Pathology, Emory University School of Medicine; Atlanta, GA 30322, USA
| | - Diogo Magnani
- Nonhuman Primate reagent Resource, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Kyha Williams
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Jean Kwun
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | - Stuart J. Knechtle
- Duke Transplant Center, Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
8
|
Grattoni A, Korbutt G, Tomei AA, García AJ, Pepper AR, Stabler C, Brehm M, Papas K, Citro A, Shirwan H, Millman JR, Melero-Martin J, Graham M, Sefton M, Ma M, Kenyon N, Veiseh O, Desai TA, Nostro MC, Marinac M, Sykes M, Russ HA, Odorico J, Tang Q, Ricordi C, Latres E, Mamrak NE, Giraldo J, Poznansky MC, de Vos P. Harnessing cellular therapeutics for type 1 diabetes mellitus: progress, challenges, and the road ahead. Nat Rev Endocrinol 2025; 21:14-30. [PMID: 39227741 PMCID: PMC11938328 DOI: 10.1038/s41574-024-01029-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a growing global health concern that affects approximately 8.5 million individuals worldwide. T1DM is characterized by an autoimmune destruction of pancreatic β cells, leading to a disruption in glucose homeostasis. Therapeutic intervention for T1DM requires a complex regimen of glycaemic monitoring and the administration of exogenous insulin to regulate blood glucose levels. Advances in continuous glucose monitoring and algorithm-driven insulin delivery devices have improved the quality of life of patients. Despite this, mimicking islet function and complex physiological feedback remains challenging. Pancreatic islet transplantation represents a potential functional cure for T1DM but is hindered by donor scarcity, variability in harvested cells, aggressive immunosuppressive regimens and suboptimal clinical outcomes. Current research is directed towards generating alternative cell sources, improving transplantation methods, and enhancing cell survival without chronic immunosuppression. This Review maps the progress in cell replacement therapies for T1DM and outlines the remaining challenges and future directions. We explore the state-of-the-art strategies for generating replenishable β cells, cell delivery technologies and local targeted immune modulation. Finally, we highlight relevant animal models and the regulatory aspects for advancing these technologies towards clinical deployment.
Collapse
Affiliation(s)
- Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, Houston Methodist Hospital, Houston, TX, USA.
- Department of Radiation Oncology, Houston Methodist Hospital, Houston, TX, USA.
| | - Gregory Korbutt
- Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Alice A Tomei
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Biomedical Engineering, University of Miami, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrés J García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
| | - Andrew R Pepper
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - Cherie Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL, USA
- Diabetes Institute, University of Florida, Gainesville, FL, USA
| | - Michael Brehm
- Program in Molecular Medicine, Diabetes Center of Excellence, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Klearchos Papas
- Department of Surgery, The University of Arizona, Tucson, AZ, USA
| | - Antonio Citro
- Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Haval Shirwan
- Department of Pediatrics, Ellis Fischel Cancer Center, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Jeffrey R Millman
- Division of Endocrinology, Metabolism and Lipid Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Juan Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute, Cambridge, MA, USA
| | - Melanie Graham
- Department of Surgery, University of Minnesota, Minneapolis, MN, USA
- Department of Veterinary Population Medicine, University of Minnesota, St. Paul, MN, USA
| | - Michael Sefton
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, Ithaca, NY, USA
| | - Norma Kenyon
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omid Veiseh
- Department of Bioengineering, Rice University, Houston, TX, USA
| | - Tejal A Desai
- University of California, San Francisco, Department of Bioengineering and Therapeutic Sciences, San Francisco, CA, USA
- Brown University, School of Engineering, Providence, RI, USA
| | - M Cristina Nostro
- McEwen Stem Cell Institute, University Health Network, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | | | - Megan Sykes
- Department of Medicine, Columbia Center for Translational Immunology, Columbia University, New York, NY, USA
- Department of Microbiology and Immunology, Columbia University, New York, NY, USA
- Department of Surgery, Columbia University, New York, NY, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Jon Odorico
- UW Health Transplant Center, Madison, WI, USA
- Division of Transplantation, Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Qizhi Tang
- Diabetes Center, University of California San Francisco, San Francisco, CA, USA
- Department of Surgery, University of California San Francisco, San Francisco, CA, US
- Gladstone Institute of Genomic Immunology, University of California San Francisco, San Francisco, CA, USA
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Esther Latres
- Research Department, Breakthrough T1D, New York, NY, USA
| | | | - Jaime Giraldo
- Research Department, Breakthrough T1D, New York, NY, USA.
| | - Mark C Poznansky
- Vaccine and Immunotherapy Center, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen and University Medical Center Groningen, Groningen, Netherlands.
| |
Collapse
|
9
|
Kundnani NR, Lolescu B, Dinu AR, Berceanu-Vaduva DM, Dumitrescu P, Tamaș TP, Sharma A, Popa MD. Biotechnology Revolution Shaping the Future of Diabetes Management. Biomolecules 2024; 14:1563. [PMID: 39766270 PMCID: PMC11674738 DOI: 10.3390/biom14121563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
INTRODUCTION Diabetes mellitus (DM) has a millennia-long history, with early references dating back to ancient Egypt and India. However, it was not until the 20th century that the connection between diabetes and insulin was fully understood. The sequencing of insulin in the 1950s initiated the convergence of biotechnology and diabetes management, leading to the development of recombinant human insulin in 1982. This marked the start of peptide-based therapies in DM. Recombinant peptides for DM treatment: Numerous recombinant peptides have been developed since, starting with modified insulin molecules, with the aim of bettering DM management through fine-tuning the glycemic response to insulin. Peptide-based therapies in DM have expanded substantially beyond insulin to include agonists of Glucagon-like peptide-1 receptor and Glucose-dependent insulinotropic polypeptide receptor, glucagon receptor antagonists, and even peptides exerting multiple receptor agonist effects, for better metabolic control. Insulin pumps, continuous glucose monitoring, and automated insulin delivery systems: The development of modern delivery systems combined with real-time glucose monitoring has significantly advanced diabetes care. Insulin pumps evolved from early large devices to modern sensor-augmented pumps with automated shutoff features and hybrid closed-loop systems, requiring minimal user input. The second-generation systems have demonstrated superior outcomes, proving highly effective in diabetes management. Islet cell transplantation, organoids, and biological pancreas augmentation represent innovative approaches to diabetes management. Islet cell transplantation aims to restore insulin production by transplanting donor beta cells, though challenges persist regarding graft survival and the need for immunosuppression. Organoids are a promising platform for generating insulin-producing cells, although far from clinical use. Biological pancreas augmentation relies on therapies that promote beta-cell (re)generation, reduce stress, and induce immune tolerance. Further biotechnology-driven perspectives in DM will include metabolic control via biotechnology-enabled tools such as custom-designed insulin hybrid molecules, machine-learning algorithms to control peptide release, and engineering cells for optimal peptide production and secretion.
Collapse
Affiliation(s)
- Nilima Rajpal Kundnani
- Department of Cardiology—Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (N.R.K.)
- Research Centre of Timisoara Institute of Cardiovascular Diseases, “Victor Babeșs” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Bogdan Lolescu
- Doctoral School Medicine-Pharmacy, “Victor Babeș” University of Medicine and Pharmacy from Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Anca-Raluca Dinu
- Department XVI, Medical Recovery, “Victor Babeş” University of Medicine and Pharmacy, 300041 Timisoara, Romania
- Research Center for Assessment of Human Motion and Functionality and Disability, “Victor Babeșs” University of Medicine and Pharmacy, Eftimie Murgu Square, No. 2, 300041 Timisoara, Romania
- “Pius Brinzeu” Emergency Clinical County Hospital, Bld Liviu Rebreanu, No. 156, 300723 Timisoara, Romania
| | - Delia Mira Berceanu-Vaduva
- Discipline of Microbiology, Department XIV Microbiology, University of Medicine and Pharmacy from Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (D.M.B.-V.)
| | - Patrick Dumitrescu
- Faculty of Medicine, University of Medicine and Pharmacy from Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Tudor-Paul Tamaș
- Discipline of Physiology, Department III—Functional Sciences, University of Medicine and Pharmacy from Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania
| | - Abhinav Sharma
- Department of Cardiology—Internal Medicine and Ambulatory Care, Prevention and Cardiovascular Recovery, “Victor Babeș” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (N.R.K.)
| | - Mihaela-Diana Popa
- Discipline of Microbiology, Department XIV Microbiology, University of Medicine and Pharmacy from Timisoara, Eftimie Murgu Sq. No. 2, 300041 Timisoara, Romania; (D.M.B.-V.)
| |
Collapse
|
10
|
Chuang ST, Alcazar O, Watts B, Abdulreda MH, Buchwald P. Small-molecule inhibitors of the CD40-CD40L costimulatory interaction are effective in pancreatic islet transplantation and prevention of type 1 diabetes models. Front Immunol 2024; 15:1484425. [PMID: 39606229 PMCID: PMC11599200 DOI: 10.3389/fimmu.2024.1484425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024] Open
Abstract
As part of our work to develop small-molecule inhibitors (SMIs) of the CD40-CD40L(CD154) costimulatory protein-protein interaction, here, we describe the ability of two of our most promising SMIs, DRI-C21041 and DRI-C21095, to prolong the survival and function of islet allografts in two murine models of islet transplantation (under the kidney capsule and in the anterior chamber of the eye) and to prevent autoimmune type 1 diabetes (T1D) onset in NOD mice. In both transplant models, a significant portion of islet allografts (50%-80%) remained intact and functional long after terminating treatment, suggesting the possibility of inducing operational immune tolerance via inhibition of the CD40-CD40L axis. SMI-treated mice maintained the structural integrity and function of their islet allografts with concomitant reduction in immune cell infiltration as evidenced by direct longitudinal imaging in situ. Furthermore, in female NODs, three-month SMI treatment reduced the incidence of diabetes from 80% to 60% (DRI-C21041) and 25% (DRI-C21095). These results (i) demonstrate the susceptibility of this TNF superfamily protein-protein interaction to small-molecule inhibition, (ii) confirm the in vivo therapeutic potential of these SMIs of a critical immune checkpoint, and (iii) reaffirm the therapeutic promise of CD40-CD40L blockade in islet transplantation and T1D prevention. Thus, CD40L-targeting SMIs could ultimately lead to alternative immunomodulatory therapeutics for transplant recipients and prevention of autoimmune diseases that are safer, less immunogenic, more controllable (shorter half-lives), and more patient-friendly (i.e., suitable for oral administration, which makes them easier to administer) than corresponding antibody-based interventions.
Collapse
Affiliation(s)
- Sung-Ting Chuang
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Oscar Alcazar
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Brandon Watts
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Midhat H. Abdulreda
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Surgery, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Ophthalmology, Miller School of Medicine, University of Miami, Miami, FL, United States
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL, United States
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, United States
| |
Collapse
|
11
|
Durgam SS, Rosado-Sánchez I, Yin D, Speck M, Mojibian M, Sayin I, Hynes GE, Alegre ML, Levings MK, Chong AS. CAR Treg synergy with anti-CD154 mediates infectious tolerance to dictate heart transplant outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.20.614149. [PMID: 39386649 PMCID: PMC11463638 DOI: 10.1101/2024.09.20.614149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Successful allograft specific tolerance induction would eliminate the need for daily immunosuppression and improve post-transplant quality of life. Adoptive cell therapy with regulatory T cells expressing donor-specific Chimeric Antigen Receptors (CAR-Tregs) is a promising strategy, but as monotherapy, cannot prolong the survival with allografts with multiple MHC mismatches. Using an HLA-A2-transgenic haplo-mismatched heart transplantation model in immunocompetent C57Bl/6 recipients, we show that HLA-A2-specific (A2) CAR Tregs was able to synergize with low dose of anti-CD154 to enhance graft survival. Using haplo-mismatched grafts expressing the 2W-OVA transgene and tetramer-based tracking of 2W- and OVA-specific T cells, we showed that in mice with accepted grafts, A2.CAR Tregs inhibited endogenous non-A2 donor- specific T cell, B cell and antibody responses, and promoted a significant increase in endogenous FoxP3 + Tregs with indirect donor-specificity. By contrast, in mice where A2.CAR Tregs failed to prolong graft survival, FoxP3 neg A2.CAR T cells preferentially accumulated in rejecting allografts and endogenous donor-specific responses were not controlled. This study therefore provides the first evidence for synergy between A2.CAR Tregs and CD154 blockade to promote infectious tolerance in immunocompetent recipients of haplo-mismatched heart grafts and defines features of A2.CAR Tregs when they fail to reshape host immunity towards allograft tolerance.
Collapse
|
12
|
Adams AB, Faber D, Lovasik BP, Matar AJ, Kim SC, Burlak C, Tector M, Tector AJ. Iscalimab Combined With Transient Tesidolumab Prolongs Survival in Pig-to-Rhesus Monkey Renal Xenografts. Xenotransplantation 2024; 31:e12880. [PMID: 39185772 DOI: 10.1111/xen.12880] [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: 01/08/2024] [Revised: 07/29/2024] [Accepted: 08/07/2024] [Indexed: 08/27/2024]
Abstract
OBJECTIVE To evaluate the clinically relevant anti-CD40 antibody iscalimab for baseline immunosuppression in a preclinical pig-to-rhesus renal xenograft model. SUMMARY BACKGROUND DATA CD40/CD40L co-stimulation blockade-based immunosuppression has been more successful than calcineurin-based protocols in prolonging xenograft survival in preclinical models. METHODS GGTA1 knockout/CD55 transgenic pig kidneys were transplanted into rhesus monkeys (n = 6) receiving an iscalimab-based immunosuppressive regimen. RESULTS Two grafts were lost early (22 and 26 days) because of ectatic donor ureters with otherwise normal histology. The other recipients survived 171, 315, 422, and 439 days with good renal function throughout the posttransplant course. None of the recipients experienced serious infectious morbidity. CONCLUSIONS It may be reasonable to evaluate an iscalimab-based immunosuppressive regimen in clinical renal xenotransplantation.
Collapse
Affiliation(s)
- Andrew B Adams
- Department of Surgery, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - David Faber
- Department of Surgery, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Brendan P Lovasik
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Abraham J Matar
- Department of Surgery, University of Minnesota School of Medicine, Minneapolis, Minnesota, USA
| | - Steven C Kim
- Department of Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Christopher Burlak
- Department of Surgery, University of Miami School of Medicine, Miami, Florida, USA
| | | | - Alfred J Tector
- Department of Surgery, University of Miami School of Medicine, Miami, Florida, USA
| |
Collapse
|
13
|
Schmoeckel M, Denner J, Reichart B, Wolf E, Hagl C. Does Xenotransplantation Offer a Large Benefit for Human Patients?-A Reply. Thorac Cardiovasc Surg 2024; 72:286-287. [PMID: 38350646 DOI: 10.1055/s-0044-1779344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2024]
Affiliation(s)
- Michael Schmoeckel
- Department of Cardiac Surgery, LMU Klinikum Grosshadern, Munich, Germany
| | - Joachim Denner
- Institute of Virology, Free University of Berlin, Germany
| | - Bruno Reichart
- DFG-Transregio-Sonderforschungsbereich TR127 "Xenotransplantation," Walter-Brendel-Institute for Experimental Medicine, LMU Munich, Germany
| | - Eckhard Wolf
- Gene Centre and Centre for Innovative Medical Models (CiMM), LMU Munich, Germany
| | - Christian Hagl
- Department of Cardiac Surgery, LMU Klinikum Grosshadern, Munich, Germany
- DZHK (Deutsches Zentrum für Herz-Kreislauf-Forschung e.V.), Partner Site Munich, Germany
| |
Collapse
|
14
|
Wallace BI, Cooney L, Fox DA. New molecular targets in the treatment of rheumatoid arthritis. Curr Opin Rheumatol 2024; 36:235-240. [PMID: 38165286 DOI: 10.1097/bor.0000000000001000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
PURPOSE OF REVIEW This review will discuss selected emerging molecular targets and associated potential therapeutic agents for rheumatoid arthritis (RA)-directed treatment. RECENT FINDINGS Agents in active development for RA treatment include those targeted to CD40 and CD40 ligand, programmed death protein 1 (PD-1), and granulocyte-macrophage colony-stimulating factor (GM-CSF). Several other molecules with a strong theoretical role in RA pathogenesis and/or demonstrated efficacy in other autoimmune diseases are also being evaluated as potential drug targets in preclinical or translational studies in RA. These targets include interleukin 1 receptor associated kinases 1 and 4 (IRAK1, IRAK4), tyrosine kinase 2 (Tyk2), bradykinin receptor 1 (B1R), OX40 and OX40 ligand. SUMMARY Identification of molecular targets for RA treatment remains an active area of investigation, with multiple therapeutic agents in clinical and preclinical development.
Collapse
Affiliation(s)
- Beth I Wallace
- Division of Rheumatology, Department of Internal Medicine, University of Michigan
- Center for Clinical Management Research, VA Ann Arbor Healthcare System
- Rheumatology Section, VA Ann Arbor Healthcare System, Ann Arbor, Michigan, USA
| | - Laura Cooney
- Division of Rheumatology, Department of Internal Medicine, University of Michigan
| | - David A Fox
- Division of Rheumatology, Department of Internal Medicine, University of Michigan
| |
Collapse
|
15
|
Ho BX, Teo AKK, Ng NHJ. Innovations in bio-engineering and cell-based approaches to address immunological challenges in islet transplantation. Front Immunol 2024; 15:1375177. [PMID: 38650946 PMCID: PMC11033429 DOI: 10.3389/fimmu.2024.1375177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/11/2024] [Indexed: 04/25/2024] Open
Abstract
Human allogeneic pancreatic islet transplantation is a life-changing treatment for patients with severe Type 1 Diabetes (T1D) who suffer from hypoglycemia unawareness and high risk of severe hypoglycemia. However, intensive immunosuppression is required to prevent immune rejection of the graft, that may in turn lead to undesirable side effects such as toxicity to the islet cells, kidney toxicity, occurrence of opportunistic infections, and malignancies. The shortage of cadaveric human islet donors further limits islet transplantation as a treatment option for widespread adoption. Alternatively, porcine islets have been considered as another source of insulin-secreting cells for transplantation in T1D patients, though xeno-transplants raise concerns over the risk of endogenous retrovirus transmission and immunological incompatibility. As a result, technological advancements have been made to protect transplanted islets from immune rejection and inflammation, ideally in the absence of chronic immunosuppression, to improve the outcomes and accessibility of allogeneic islet cell replacement therapies. These include the use of microencapsulation or macroencapsulation devices designed to provide an immunoprotective environment using a cell-impermeable layer, preventing immune cell attack of the transplanted cells. Other up and coming advancements are based on the use of stem cells as the starting source material for generating islet cells 'on-demand'. These starting stem cell sources include human induced pluripotent stem cells (hiPSCs) that have been genetically engineered to avoid the host immune response, curated HLA-selected donor hiPSCs that can be matched with recipients within a given population, and multipotent stem cells with natural immune privilege properties. These strategies are developed to provide an immune-evasive cell resource for allogeneic cell therapy. This review will summarize the immunological challenges facing islet transplantation and highlight recent bio-engineering and cell-based approaches aimed at avoiding immune rejection, to improve the accessibility of islet cell therapy and enhance treatment outcomes. Better understanding of the different approaches and their limitations can guide future research endeavors towards developing more comprehensive and targeted strategies for creating a more tolerogenic microenvironment, and improve the effectiveness and sustainability of islet transplantation to benefit more patients.
Collapse
Affiliation(s)
- Beatrice Xuan Ho
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- BetaLife Pte Ltd, Singapore, Singapore
| | - Adrian Kee Keong Teo
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Precision Medicine Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Natasha Hui Jin Ng
- Stem Cells and Diabetes Laboratory, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| |
Collapse
|
16
|
Hu X, White K, Young C, Olroyd AG, Kievit P, Connolly AJ, Deuse T, Schrepfer S. Hypoimmune islets achieve insulin independence after allogeneic transplantation in a fully immunocompetent non-human primate. Cell Stem Cell 2024; 31:334-340.e5. [PMID: 38335966 DOI: 10.1016/j.stem.2024.02.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/21/2024] [Accepted: 02/01/2024] [Indexed: 02/12/2024]
Abstract
Allogeneic transplantation of pancreatic islets for patients with difficult-to-control diabetes mellitus is severely hampered by the requirement for continuous immunosuppression and its associated morbidity. We report that allogeneic transplantation of genetically engineered (B2M-/-, CIITA-/-, CD47+), primary, hypoimmune, pseudo-islets (p-islets) results in their engraftment into a fully immunocompetent, diabetic non-human primate wherein they provide stable endocrine function and enable insulin independence without inducing any detectable immune response in the absence of immunosuppression. Hypoimmune primary p-islets may provide a curative cell therapy for type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Xiaomeng Hu
- 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
| | - Ari G Olroyd
- Sana Biotechnology, Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Paul Kievit
- Sana Biotechnology, Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Andrew J Connolly
- Sana Biotechnology, Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Tobias Deuse
- Sana Biotechnology, Inc., 1 Tower Place, South San Francisco, CA 94080, USA
| | - Sonja Schrepfer
- Sana Biotechnology, Inc., 1 Tower Place, South San Francisco, CA 94080, USA.
| |
Collapse
|
17
|
Kamberi S, Meier RPH. Xenotransplantation literature update March 2023-November 2023. Xenotransplantation 2024; 31:e12837. [PMID: 38334060 DOI: 10.1111/xen.12837] [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: 11/15/2023] [Accepted: 11/16/2023] [Indexed: 02/10/2024]
Abstract
We present an updated report highlighting significant developments in the field of xenotransplantation since March 2023. The past six months have witnessed significant strides in the field and the emergence of novel research that is expected to facilitate the journey towards clinical trials. We are reviewing here the most pertinent findings from March 2023 to November 2023.
Collapse
Affiliation(s)
- Shani Kamberi
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Raphael P H Meier
- Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|