1
|
Ellis GI, Coker KE, Winn DW, Deng MZ, Shukla D, Bhoj V, Milone MC, Wang W, Liu C, Naji A, Duran-Struuck R, Riley JL. Trafficking and persistence of alloantigen-specific chimeric antigen receptor regulatory T cells in Cynomolgus macaque. Cell Rep Med 2022; 3:100614. [PMID: 35551746 PMCID: PMC9133392 DOI: 10.1016/j.xcrm.2022.100614] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/16/2022] [Accepted: 03/29/2022] [Indexed: 01/13/2023]
Abstract
Adoptive transfer of chimeric antigen receptor regulatory T cells (CAR Tregs) is a promising way to prevent allograft loss without the morbidity associated with current therapies. Non-human primates (NHPs) are a clinically relevant model to develop transplant regimens, but manufacturing and engraftment of NHP CAR Tregs have not been demonstrated yet. Here, we describe a culture system that massively expands CAR Tregs specific for the Bw6 alloantigen. In vitro, these Tregs suppress in an antigen-specific manner without pro-inflammatory cytokine secretion or cytotoxicity. In vivo, Bw6-specific CAR Tregs preferentially traffic to and persist in bone marrow for at least 1 month. Following transplant of allogeneic Bw6+ islets and autologous CAR Tregs into the bone marrow of diabetic recipients, CAR Tregs traffic to the site of islet transplantation and maintain a phenotype of suppressive Tregs. Our results establish a framework for the optimization of CAR Treg therapy in NHP disease models.
Collapse
Affiliation(s)
- Gavin I Ellis
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly E Coker
- Department of Pathobiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Delaine W Winn
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Mosha Z Deng
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Divanshu Shukla
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
| | - Vijay Bhoj
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wei Wang
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Chengyang Liu
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Ali Naji
- Department of Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | | | - James L Riley
- Department of Microbiology and Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
2
|
Thomson AW, Sasaki K, Ezzelarab MB. Non-human Primate Regulatory T Cells and Their Assessment as Cellular Therapeutics in Preclinical Transplantation Models. Front Cell Dev Biol 2021; 9:666959. [PMID: 34211972 PMCID: PMC8239398 DOI: 10.3389/fcell.2021.666959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/21/2021] [Indexed: 11/13/2022] Open
Abstract
Non-human primates (NHP) are an important resource for addressing key issues regarding the immunobiology of regulatory T cells (Treg), their in vivo manipulation and the translation of adoptive Treg therapy to clinical application. In addition to their phenotypic and functional characterization, particularly in cynomolgus and rhesus macaques, NHP Treg have been isolated and expanded successfully ex vivo. Their numbers can be enhanced in vivo by administration of IL-2 and other cytokines. Both polyclonal and donor antigen (Ag) alloreactive NHP Treg have been expanded ex vivo and their potential to improve long-term outcomes in organ transplantation assessed following their adoptive transfer in combination with various cytoreductive, immunosuppressive and "Treg permissive" agents. In addition, important insights have been gained into the in vivo fate/biodistribution, functional stability, replicative capacity and longevity of adoptively-transferred Treg in monkeys. We discuss current knowledge of NHP Treg immunobiology, methods for their in vivo expansion and functional validation, and results obtained testing their safety and efficacy in organ and pancreatic islet transplantation models. We compare and contrast results obtained in NHP and mice and also consider prospects for future, clinically relevant studies in NHP aimed at improved understanding of Treg biology, and innovative approaches to promote and evaluate their therapeutic potential.
Collapse
Affiliation(s)
- Angus W. Thomson
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Kazuki Sasaki
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Mohamed B. Ezzelarab
- Department of Surgery, Thomas E. Starzl Transplantation Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| |
Collapse
|
3
|
Cook L, Reid KT, Häkkinen E, de Bie B, Tanaka S, Smyth DJ, White MP, Wong MQ, Huang Q, Gillies JK, Ziegler SF, Maizels RM, Levings MK. Induction of stable human FOXP3 + Tregs by a parasite-derived TGF-β mimic. Immunol Cell Biol 2021; 99:833-847. [PMID: 33929751 PMCID: PMC8453874 DOI: 10.1111/imcb.12475] [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: 01/21/2021] [Revised: 03/31/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022]
Abstract
Immune homeostasis in the intestine is tightly controlled by FOXP3+ regulatory T cells (Tregs), defects of which are linked to the development of chronic conditions, such as inflammatory bowel disease (IBD). As a mechanism of immune evasion, several species of intestinal parasites boost Treg activity. The parasite Heligmosomoides polygyrus is known to secrete a molecule (Hp-TGM) that mimics the ability of TGF-β to induce FOXP3 expression in CD4+ T cells. The study aimed to investigate whether Hp-TGM could induce human FOXP3+ Tregs as a potential therapeutic approach for inflammatory diseases. CD4+ T cells from healthy volunteers were expanded in the presence of Hp-TGM or TGF-β. Treg induction was measured by flow cytometric detection of FOXP3 and other Treg markers, such as CD25 and CTLA-4. Epigenetic changes were detected using ChIP-Seq and pyrosequencing of FOXP3. Treg phenotype stability was assessed following inflammatory cytokine challenge and Treg function was evaluated by cellular co-culture suppression assays and cytometric bead arrays for secreted cytokines. Hp-TGM efficiently induced FOXP3 expression (> 60%), in addition to CD25 and CTLA-4, and caused epigenetic modification of the FOXP3 locus to a greater extent than TGF-β. Hp-TGM-induced Tregs had superior suppressive function compared with TGF-β-induced Tregs, and retained their phenotype following exposure to inflammatory cytokines. Furthermore, Hp-TGM induced a Treg-like phenotype in in vivo differentiated Th1 and Th17 cells, indicating its potential to re-program memory cells to enhance immune tolerance. These data indicate Hp-TGM has potential to be used to generate stable human FOXP3+ Tregs to treat IBD and other inflammatory diseases.
Collapse
Affiliation(s)
- Laura Cook
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Kyle T Reid
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Elmeri Häkkinen
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Brett de Bie
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Shigeru Tanaka
- Department of Translational Research, Benaroya Research Institute, Virginia Mason, Seattle, WA, USA
| | - Danielle J Smyth
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Madeleine Pj White
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - May Q Wong
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Qing Huang
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Jana K Gillies
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Steven F Ziegler
- Department of Translational Research, Benaroya Research Institute, Virginia Mason, Seattle, WA, USA
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Megan K Levings
- BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada.,Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
4
|
Park JA, Santich BH, Xu H, Lum LG, Cheung NKV. Potent ex vivo armed T cells using recombinant bispecific antibodies for adoptive immunotherapy with reduced cytokine release. J Immunother Cancer 2021; 9:e002222. [PMID: 33986124 PMCID: PMC8126293 DOI: 10.1136/jitc-2020-002222] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND T cell-based immunotherapies using chimeric antigen receptors (CAR) or bispecific antibodies (BsAb) have produced impressive responses in hematological malignancies. However, major hurdles remained, including cytokine release syndrome, neurotoxicity, on-target off-tumor effects, reliance on autologous T cells, and failure in most solid tumors. BsAb armed T cells offer a safe alternative. METHODS We generated ex vivo armed T cells (EATs) using IgG-[L]-scFv-platformed BsAb, where the anti-CD3 (huOKT3) scFv was attached to the light chain of a tumor-binding IgG. BsAb density on EAT, in vitro cytotoxicity, cytokine release, in vivo trafficking into tumors, and their antitumor activities were evaluated in multiple cancer cell lines and patient-derived xenograft mouse models. The efficacy of EATs after cryopreservation was studied, and gamma delta (γδ) T cells were investigated as unrelated alternative effector T cells. RESULTS The antitumor potency of BsAb armed T cells was substantially improved using the IgG-[L]-scFv BsAb platform. When compared with separate BsAb and T cell injection, EATs released less TNF-α, and infiltrated tumors faster, while achieving robust antitumor responses. The in vivo potency of EAT therapy depended on BsAb dose for arming, EAT cell number per injection, total number of EAT doses, and treatment schedule intensity. The antitumor efficacy of EATs was preserved following cryopreservation, and EATs using γδ T cells were safe and as effective as αβ T cell-EATs. CONCLUSIONS EATs exerted potent antitumor activities against a broad spectrum of human cancer targets with remarkable safety. The antitumor potency of EATs depended on BsAb dose, cell number and total dose, and schedule. EATs were equally effective after cryopreservation, and the feasibility of third-party γδ-EATs offered an alternative for autologous T cell sources.
Collapse
MESH Headings
- Animals
- Antibodies, Bispecific/genetics
- Antibodies, Bispecific/immunology
- Antibodies, Bispecific/metabolism
- Cell Line, Tumor
- Cell Movement
- Coculture Techniques
- Cytokines/metabolism
- Cytotoxicity, Immunologic
- Humans
- Immunotherapy, Adoptive
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Intraepithelial Lymphocytes/transplantation
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/transplantation
- Male
- Mice, Inbred BALB C
- Mice, Knockout
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Phenotype
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- Jeong A Park
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Brian H Santich
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Hong Xu
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lawrence G Lum
- Medicine, University of Virginia, Charlottesville, Virginia, USA
| | - Nai-Kong V Cheung
- Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| |
Collapse
|
5
|
Mukhatayev Z, Dellacecca ER, Cosgrove C, Shivde R, Jaishankar D, Pontarolo-Maag K, Eby JM, Henning SW, Ostapchuk YO, Cedercreutz K, Issanov A, Mehrotra S, Overbeck A, Junghans RP, Leventhal JR, Le Poole IC. Antigen Specificity Enhances Disease Control by Tregs in Vitiligo. Front Immunol 2020; 11:581433. [PMID: 33335528 PMCID: PMC7736409 DOI: 10.3389/fimmu.2020.581433] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Vitiligo is an autoimmune skin disease characterized by melanocyte destruction. Regulatory T cells (Tregs) are greatly reduced in vitiligo skin, and replenishing peripheral skin Tregs can provide protection against depigmentation. Ganglioside D3 (GD3) is overexpressed by perilesional epidermal cells, including melanocytes, which prompted us to generate GD3-reactive chimeric antigen receptor (CAR) Tregs to treat vitiligo. Mice received either untransduced Tregs or GD3-specific Tregs to test the hypothesis that antigen specificity contributes to reduced autoimmune reactivity in vitro and in vivo. CAR Tregs displayed increased IL-10 secretion in response to antigen, provided superior control of cytotoxicity towards melanocytes, and supported a significant delay in depigmentation compared to untransduced Tregs and vehicle control recipients in a TCR transgenic mouse model of spontaneous vitiligo. The latter findings were associated with a greater abundance of Tregs and melanocytes in treated mice versus both control groups. Our data support the concept that antigen-specific Tregs can be prepared, used, and stored for long-term control of progressive depigmentation.
Collapse
Affiliation(s)
- Zhussipbek Mukhatayev
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States.,Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan.,Laboratory of Molecular immunology and Immunobiotechnology, M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Emilia R Dellacecca
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Cormac Cosgrove
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Rohan Shivde
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | - Dinesh Jaishankar
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| | | | - Jonathan M Eby
- Oncology Research Institute, Loyola University, Maywood, IL, United States
| | - Steven W Henning
- Oncology Research Institute, Loyola University, Maywood, IL, United States
| | - Yekaterina O Ostapchuk
- Laboratory of Molecular immunology and Immunobiotechnology, M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | - Kettil Cedercreutz
- Department of Dermatology, Northwestern University, Chicago, IL, United States
| | - Alpamys Issanov
- Department of Medicine, School of Medicine, Nazarbayev University, Nur-Sultan, Kazakhstan
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Andreas Overbeck
- Department for Surgery of Pigment Disorders, Lumiderm, Madrid, Spain
| | - Richard P Junghans
- Department of Hematology/Oncology, Boston University, Boston MA, United States
| | - Joseph R Leventhal
- Comprehensive Transplant Center, Northwestern Memorial Hospital, Chicago, IL, United States
| | - I Caroline Le Poole
- Department of Dermatology, Northwestern University, Chicago, IL, United States.,Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, United States
| |
Collapse
|
6
|
Xenotransplantation tolerance: applications for recent advances in modified swine. Curr Opin Organ Transplant 2019; 23:642-648. [PMID: 30379724 DOI: 10.1097/mot.0000000000000585] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The aim of this study was to review the recent progress in xenotransplantation achieved through genetic engineering and discuss the potential of tolerance induction to overcome remaining barriers to extended xenograft survival. RECENT FINDINGS The success of life-saving allotransplantation has created a demand for organ transplantation that cannot be met by the supply of human organs. Xenotransplantation is one possible solution that would allow for a nearly unlimited supply of organs. Recent genetic engineering of swine has decreased the reactivity of preformed antibodies to some, but not all, potential human recipients. Experiments using genetically modified swine organs have now resulted in survival of life-supporting kidneys for over a year. However, the grafts show evidence of antibody-mediated rejection on histology, suggesting additional measures will be required for further extension of graft survival. Tolerance induction through mixed chimerism or thymic transplantation across xenogeneic barriers would be well suited for patients with a positive crossmatch to genetically modified swine or relatively negative crossmatches to genetically modified swine, respectively. SUMMARY This review highlights the current understanding of the immunologic processes in xenotransplantation and describes the development and application of strategies designed to overcome them from the genetic modification of the source animal to the induction of tolerance to xenografts.
Collapse
|
7
|
Regulatory T-cell Number in Peripheral Blood at 1 Year Posttransplant as Predictor of Long-term Kidney Graft Survival. Transplant Direct 2019; 5:e426. [PMID: 30882031 PMCID: PMC6411222 DOI: 10.1097/txd.0000000000000871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2018] [Revised: 01/17/2019] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Supplemental digital content is available in the text. Background Regulatory T (Treg) cells play a role in limiting kidney transplant rejection and can potentially promote long-term transplant tolerance. There are no large prospective studies demonstrating the utility of peripheral blood Treg cells as biomarkers for long-term graft outcome in kidney transplantation. The aim of our study was to analyze the influence of the absolute number of peripheral blood Treg cells after transplantation on long-term death-censored graft survival. Methods We monitored the absolute numbers of Treg cells by flow cytometry in nonfrozen samples of peripheral blood in 133 kidney transplant recipients, who were prospectively followed up to 2 years after transplantation. Death-censored graft survival was determined retrospectively in January 2017. Results The mean time of clinical follow-up was 7.4 ± 2.9 years and 24.1% patients suffered death-censored graft loss (DCGL). Patients with high Treg cells 1 year after transplantation and above the median value (14.57 cells/mm3), showed better death-censored graft survival (5-year survival, 92.5% vs 81.4%, Log-rank P = .030). One-year Treg cells showed a receiver operating characteristic - area under curve of 63.1% (95% confidence interval, 52.9–73.2%, P = 0.026) for predicting DCGL. After multivariate Cox regression analysis, an increased number of peripheral blood Treg cells was a protective factor for DCGL (hazard ratio, 0.961, 95% confidence interval, 0.924–0.998, P = 0.041), irrespectively of 1-year proteinuria and renal function. Conclusions Peripheral blood absolute numbers of Treg cells 1 year after kidney transplantation predict a better long-term graft outcome and may be used as prognostic biomarkers.
Collapse
|
8
|
Gołąb K, Grose R, Placencia V, Wickrema A, Solomina J, Tibudan M, Konsur E, Ciepły K, Marek-Trzonkowska N, Trzonkowski P, Millis JM, Fung J, Witkowski P. Cell banking for regulatory T cell-based therapy: strategies to overcome the impact of cryopreservation on the Treg viability and phenotype. Oncotarget 2018; 9:9728-9740. [PMID: 29515766 PMCID: PMC5839397 DOI: 10.18632/oncotarget.23887] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 11/10/2017] [Indexed: 12/16/2022] Open
Abstract
The first clinical trials with adoptive Treg therapy have shown safety and potential efficacy. Feasibility of such therapy could be improved if cells are cryopreserved and stored until optimal timing for infusion. Herein, we report the evaluation of two cell-banking strategies for Treg therapy: 1) cryopreservation of CD4+ cells for subsequent Treg isolation/expansion and 2) cryopreservation of ex-vivo expanded Tregs (CD4+CD25hiCD127lo/- cells). First, we checked how cryopreservation affects cell viability and Treg markers expression. Then, we performed Treg isolation/expansion with the final products release testing. We observed substantial decrease in cell number recovery after thawing and overnight culture. This observation might be explained by the high percentage of necrotic and apoptotic cells found just after thawing. Furthermore, we noticed fluctuations in percentage of CD4+CD25hiCD127- and CD4+FoxP3+ cells obtained from cryopreserved CD4+ as well as Treg cells. However, after re-stimulation Tregs expanded well, presented a stable phenotype and fulfilled the release criteria at the end of expansions. Cryopreservation of CD4+ cells for subsequent Treg isolation/expansion and cryopreservation of expanded Tregs with re-stimulation and expansion after thawing, are promising solutions to overcome detrimental effects of cryopreservation. Both of these cell-banking strategies for Treg therapy can be applied when designing new clinical trials.
Collapse
Affiliation(s)
- Karolina Gołąb
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Randall Grose
- South Australian Health and Medical Research Institute, University of Adelaide, SA, Australia
| | - Veronica Placencia
- Department of Medicine, Hematology-Oncology, Cancer Research Center, University of Chicago, Chicago, IL, USA
| | - Amittha Wickrema
- Department of Medicine, Hematology-Oncology, Cancer Research Center, University of Chicago, Chicago, IL, USA
| | - Julia Solomina
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Martin Tibudan
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Evelyn Konsur
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Kamil Ciepły
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | | | - Piotr Trzonkowski
- Department of Clinical Immunology and Transplantology, Medical University of Gdańsk, Gdańsk, Poland
| | | | - John Fung
- Department of Surgery, University of Chicago, Chicago, IL, USA
| | - Piotr Witkowski
- Department of Surgery, University of Chicago, Chicago, IL, USA
| |
Collapse
|
9
|
Piscopo NJ, Mueller KP, Das A, Hematti P, Murphy WL, Palecek SP, Capitini CM, Saha K. Bioengineering Solutions for Manufacturing Challenges in CAR T Cells. Biotechnol J 2018; 13:10.1002/biot.201700095. [PMID: 28840981 PMCID: PMC5796845 DOI: 10.1002/biot.201700095] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 07/26/2017] [Indexed: 12/13/2022]
Abstract
The next generation of therapeutic products to be approved for the clinic is anticipated to be cell therapies, termed "living drugs" for their capacity to dynamically and temporally respond to changes during their production ex vivo and after their administration in vivo. Genetically engineered chimeric antigen receptor (CAR) T cells have rapidly developed into powerful tools to harness the power of immune system manipulation against cancer. Regulatory agencies are beginning to approve CAR T cell therapies due to their striking efficacy in treating some hematological malignancies. However, the engineering and manufacturing of such cells remains a challenge for widespread adoption of this technology. Bioengineering approaches including biomaterials, synthetic biology, metabolic engineering, process control and automation, and in vitro disease modeling could offer promising methods to overcome some of these challenges. Here, we describe the manufacturing process of CAR T cells, highlighting potential roles for bioengineers to partner with biologists and clinicians to advance the manufacture of these complex cellular products under rigorous regulatory and quality control.
Collapse
Affiliation(s)
- Nicole J Piscopo
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Katherine P Mueller
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Amritava Das
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin, Madison, WI, USA
| | - Christian M Capitini
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA
- Wisconsin Institute for Discovery, University of Wisconsin, Madison, WI, USA
- University of Wisconsin Carbone Cancer Center, Madison, WI, USA
| |
Collapse
|
10
|
Hanada T, Tsuji S, Nakayama M, Wakinoue S, Kasahara K, Kimura F, Mori T, Ogasawara K, Murakami T. Suppressive regulatory T cells and latent transforming growth factor-β-expressing macrophages are altered in the peritoneal fluid of patients with endometriosis. Reprod Biol Endocrinol 2018; 16:9. [PMID: 29391020 PMCID: PMC5796574 DOI: 10.1186/s12958-018-0325-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 01/17/2018] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Endometriosis is a known cause of infertility. Differences in immune tolerance caused by regulatory T cells (Tregs) and transforming growth factor-β (TGF-β) are thought to be involved in the pathology of endometriosis. Evidence has indicated that Tregs can be separated into three functionally and phenotypically distinct subpopulations and that activated TGF-β is released from latency-associated peptide (LAP) on the surfaces of specific cells. The aim of this study was to examine differences in Treg subpopulations and LAP in the peripheral blood (PB) and peritoneal fluid (PF) of patients with and without endometriosis. METHODS PB and PF were collected from 28 women with laparoscopically and histopathologically diagnosed endometriosis and 20 disease-free women who were subjected to laparoscopic surgery. Three subpopulations of CD4+ T lymphocytes (CD45RA+FoxP3low resting Tregs, CD45RA-FoxP3high effector Tregs, and CD45RA-FoxP3low non-Tregs) and CD11b+ mononuclear cells expressing LAP were analyzed by flow cytometry using specific monoclonal antibodies. RESULTS Proportions of suppressive Tregs (resting and effector Tregs) were significantly higher in the PF samples of patients with endometriosis than in those of control women (P = 0.02 and P < 0.01, respectively) but did not differ between the PB samples of patients and controls. The percentage of CD11b+LAP+ macrophages was significantly lower in PF samples of patients with endometriosis than in those of controls (P < 0.01) but was not altered in the PB samples. CONCLUSION Proportions of suppressive Tregs and LAP+ macrophages are altered locally in the PF of endometriosis patients.
Collapse
Affiliation(s)
- Tetsuro Hanada
- 0000 0000 9747 6806grid.410827.8Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Shunichiro Tsuji
- 0000 0000 9747 6806grid.410827.8Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Misako Nakayama
- 0000 0000 9747 6806grid.410827.8Department of Pathology, Division of Pathology and Disease Regulation, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Shiro Wakinoue
- 0000 0000 9747 6806grid.410827.8Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Kyoko Kasahara
- 0000 0000 9747 6806grid.410827.8Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Fuminori Kimura
- 0000 0000 9747 6806grid.410827.8Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Takahide Mori
- Academia for Repro-Regenerative Medicine, Nonprofit Organization, 394-1 Higashi-Hinodono-cho, Ichijo-Shinmachi-Higashiiru, Kamigyo-ku, Kyoto, 602-0917 Japan
| | - Kazumasa Ogasawara
- 0000 0000 9747 6806grid.410827.8Department of Pathology, Division of Pathology and Disease Regulation, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| | - Takashi Murakami
- 0000 0000 9747 6806grid.410827.8Department of Obstetrics and Gynecology, Shiga University of Medical Science, Seta Tsukionowa-cho, Otsu, Shiga 520-2192 Japan
| |
Collapse
|