1
|
Farhang Doost N, Srivastava SK. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. BIOSENSORS 2024; 14:225. [PMID: 38785699 PMCID: PMC11118005 DOI: 10.3390/bios14050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that simulates an artificial organ within a microfluidic cell culture chip. Current cell biology research focuses on in vitro cell cultures due to various limitations of in vivo testing. Unfortunately, in-vitro cell culturing fails to provide an accurate microenvironment, and in vivo cell culturing is expensive and has historically been a source of ethical controversy. OOC aims to overcome these shortcomings and provide the best of both in vivo and in vitro cell culture research. The critical component of the OOC design is utilizing microfluidics to ensure a stable concentration gradient, dynamic mechanical stress modeling, and accurate reconstruction of a cellular microenvironment. OOC also has the advantage of complete observation and control of the system, which is impossible to recreate in in-vivo research. Multiple throughputs, channels, membranes, and chambers are constructed in a polydimethylsiloxane (PDMS) array to simulate various organs on a chip. Various experiments can be performed utilizing OOC technology, including drug delivery research and toxicology. Current technological expansions involve multiple organ microenvironments on a single chip, allowing for studying inter-tissue interactions. Other developments in the OOC technology include finding a more suitable material as a replacement for PDMS and minimizing artefactual error and non-translatable differences.
Collapse
Affiliation(s)
| | - Soumya K. Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
2
|
Li QH, Zhao QY, Yang WJ, Jiang AF, Ren CE, Meng YH. Beyond Immune Balance: The Pivotal Role of Decidual Regulatory T Cells in Unexplained Recurrent Spontaneous Abortion. J Inflamm Res 2024; 17:2697-2710. [PMID: 38707955 PMCID: PMC11070170 DOI: 10.2147/jir.s459263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 04/18/2024] [Indexed: 05/07/2024] Open
Abstract
Recurrent spontaneous abortion (RSA) is defined as two or more consecutive pregnancy failures, which brings tremendous stress to women of childbearing age and seriously affects family well-being. However, the reason in about 50% of cases remains unknown and is defined as unexplained recurrent spontaneous abortion (URSA). The immunological perspective in URSA has attracted widespread attention in recent years. The embryo is regarded as a semi-allogeneic graft to the mother. A successful pregnancy requires transition to an immune environment conducive to embryo survival at the maternal-fetal interface. As an important member of regulatory immunity, regulatory T (Treg) cells play a key role in regulating immune tolerance at the maternal-fetal interface. This review will focus on the phenotypic plasticity and lineage stability of Treg cells to illustrate its relationship with URSA.
Collapse
Affiliation(s)
- Qing-Hui Li
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Qiu-Yan Zhao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Wei-Jing Yang
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, 261021, People’s Republic of China
| | - Ai-Fang Jiang
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Chun-E Ren
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| | - Yu-Han Meng
- Center of Reproductive Medicine, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong, 261000, People’s Republic of China
| |
Collapse
|
3
|
Agarwal G, Patel M. Review on Monoclonal Antibodies (mAbs) as a Therapeutic Approach for Type 1 Diabetes. Curr Diabetes Rev 2024; 20:e310823220578. [PMID: 37653635 DOI: 10.2174/1573399820666230831153249] [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: 01/30/2023] [Revised: 06/10/2023] [Accepted: 07/18/2023] [Indexed: 09/02/2023]
Abstract
Monoclonal antibodies have been successfully utilized in a variety of animal models to treat auto-immune illnesses for a long time. Immune system responses will either be less active or more active depending on how the immune system is operating abnormally. Immune system hypoactivity reduces the body's capacity to fight off various invading pathogens, whereas immune system hyperactivity causes the body to attack and kill its own tissues and cells. For maximal patient compliance, we will concentrate on a variety of antibody therapies in this study to treat Type 1 diabetes (an autoimmune condition). T-cells are responsible for the auto-immune condition known as T1D, which causes irregularities in the function of β-cells in the pancreas. As a result, for the treatment and prevention of T1D, immunotherapies that selectively restore continuous beta cellspecific self-tolerance are needed. Utilizing monoclonal antibodies is one way to specifically target immune cell populations responsible for the auto-immune-driven disease (mAb). Numerous mAbs have demonstrated clinical safety and varied degrees of success in modulating autoimmunity, including T1D. A targeted cell population is exhausted by mAb treatments, regardless of antigenic specificity. One drawback of this treatment is the loss of obtained protective immunity. Immune effector cell function is regulated by nondepleting monoclonal antibodies (mAb). The antigenfocused new drug delivery system is made possible by the adaptability of mAbs. For the treatment of T1D and T-cell-mediated autoimmunity, different existing and potential mAb therapy methods are described in this article.
Collapse
Affiliation(s)
- Gaurav Agarwal
- Faculty of Pharmacy, Panipat Institute of Engineering and Technology Panipat, Haryana, India
| | - Mayank Patel
- Neuropharmacology division, Department of Pharmacology, ISF College of Pharmacy, Moga, (Pb.) 142001, India
| |
Collapse
|
4
|
Elalouf A, Elalouf H, Rosenfeld A. Modulatory immune responses in fungal infection associated with organ transplant - advancements, management, and challenges. Front Immunol 2023; 14:1292625. [PMID: 38143753 PMCID: PMC10748506 DOI: 10.3389/fimmu.2023.1292625] [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: 09/11/2023] [Accepted: 11/20/2023] [Indexed: 12/26/2023] Open
Abstract
Organ transplantation stands as a pivotal achievement in modern medicine, offering hope to individuals with end-stage organ diseases. Advancements in immunology led to improved organ transplant survival through the development of immunosuppressants, but this heightened susceptibility to fungal infections with nonspecific symptoms in recipients. This review aims to establish an intricate balance between immune responses and fungal infections in organ transplant recipients. It explores the fundamental immune mechanisms, recent advances in immune response dynamics, and strategies for immune modulation, encompassing responses to fungal infections, immunomodulatory approaches, diagnostics, treatment challenges, and management. Early diagnosis of fungal infections in transplant patients is emphasized with the understanding that innate immune responses could potentially reduce immunosuppression and promise efficient and safe immuno-modulating treatments. Advances in fungal research and genetic influences on immune-fungal interactions are underscored, as well as the potential of single-cell technologies integrated with machine learning for biomarker discovery. This review provides a snapshot of the complex interplay between immune responses and fungal infections in organ transplantation and underscores key research directions.
Collapse
Affiliation(s)
- Amir Elalouf
- Department of Management, Bar-Ilan University, Ramat Gan, Israel
| | - Hadas Elalouf
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| | - Ariel Rosenfeld
- Information Science Department, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
5
|
Ziogas A, Bruno M, van der Meel R, Mulder WJM, Netea MG. Trained immunity: Target for prophylaxis and therapy. Cell Host Microbe 2023; 31:1776-1791. [PMID: 37944491 DOI: 10.1016/j.chom.2023.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 07/27/2023] [Accepted: 10/15/2023] [Indexed: 11/12/2023]
Abstract
Trained immunity is a de facto memory for innate immune responses, leading to long-term functional reprogramming of innate immune cells. In physiological conditions, trained immunity leads to adaptive states that enhance resistance against pathogens and contributes to immunosurveillance. Dysregulated trained immunity can however lead either to defective innate immune responses in severe infections or cancer or to inflammatory and autoimmune diseases if trained immunity is inappropriately activated. Here, we review the immunological and molecular mechanisms that mediate trained immunity induction and propose that trained immunity represents an important target for prophylactic and therapeutic approaches in human diseases. On the one hand, we argue that novel approaches that induce trained immunity may enhance vaccine efficacy. On the other hand, induction of trained immunity in cancer, and inhibition of exaggerated induction of trained immunity in inflammatory disorders, are viable targets amenable for new therapeutic approaches.
Collapse
Affiliation(s)
- Athanasios Ziogas
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, the Netherlands.
| | - Mariolina Bruno
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Roy van der Meel
- Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Willem J M Mulder
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Centre, Nijmegen, the Netherlands; Laboratory of Chemical Biology, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, the Netherlands; Department of Immunology and Metabolism, Life & Medical Sciences Institute, University of Bonn, Bonn, Germany
| |
Collapse
|
6
|
Dai Y, Ren D, He Y, Yi H. Editorial: Epigenetic, metabolic, and transcriptional regulation of immune cell plasticity and functions in cancer and non-cancer diseases. Front Immunol 2023; 14:1284124. [PMID: 37818355 PMCID: PMC10561278 DOI: 10.3389/fimmu.2023.1284124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 10/12/2023] Open
Affiliation(s)
- Yun Dai
- Laboratory of Cancer Precision Medicine, the First Hospital of Jilin University, Changchun, Jilin, China
| | - Dong Ren
- Irvine Medical Center, University of California, Irvine, Irvine, CA, United States
| | - Yafeng He
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Huanfa Yi
- Central Laboratory, the First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
7
|
Hansson C, Lebrero-Fernández C, Schön K, Angeletti D, Lycke N. Tr1 cell-mediated protection against autoimmune disease by intranasal administration of a fusion protein targeting cDC1 cells. Mucosal Immunol 2023; 16:486-498. [PMID: 37192682 DOI: 10.1016/j.mucimm.2023.05.006] [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: 04/19/2023] [Accepted: 05/05/2023] [Indexed: 05/18/2023]
Abstract
Curative therapies against autoimmune diseases are lacking. Indeed, most of the currently available treatments are only targeting symptoms. We have developed a novel strategy for a therapeutic vaccine against autoimmune diseases based on intranasal administration of a fusion protein tolerogen, which consists of a mutant, enzymatically inactive, cholera toxin A1 (CTA1)-subunit genetically fused to disease-relevant high-affinity peptides and a dimer of D-fragments from protein A (DD). The CTA1 R7K mutant - myelin oligodendrocyte glycoprotein (MOG), or proteolipid protein (PLP) - DD (CTA1R7K-MOG/PLP-DD) fusion proteins effectively reduced clinical symptoms in the experimental autoimmune encephalitis model of multiple sclerosis. The treatment induced Tr1 cells, in the draining lymph node, which produced interleukin (IL)-10 and suppressed effector clusters of differentiation 4+ T-cell responses. This effect was dependent on IL-27 signaling because treatment was ineffective in bone marrow chimeras lacking IL-27Ra within their hematopoietic compartment. Single-cell RNA sequencing of dendritic cells in draining lymph nodes demonstrated distinct gene transcriptional changes of classic dendritic cells 1, including enhanced lipid metabolic pathways, induced by the tolerogenic fusion protein. Thus, our results with the tolerogenic fusion protein demonstrate the possibility to vaccinate and protect against disease progression by reinstating tolerance in multiple sclerosis and other autoimmune diseases.
Collapse
Affiliation(s)
- Charlotta Hansson
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Cristina Lebrero-Fernández
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Karin Schön
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Davide Angeletti
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.
| | - Nils Lycke
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
8
|
Keppler M, Straß S, Geiger S, Fischer T, Späth N, Weinstein T, Schwamborn A, Guezguez J, Guse JH, Laufer S, Burnet M. Imidazoquinolines with improved pharmacokinetic properties induce a high IFNα to TNFα ratio in vitro and in vivo. Front Immunol 2023; 14:1168252. [PMID: 37409123 PMCID: PMC10319141 DOI: 10.3389/fimmu.2023.1168252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/31/2023] [Indexed: 07/07/2023] Open
Abstract
TLR Agonists have promising activity in preclinical models of viral infection and cancer. However, clinical use is only in topical application. Systemic uses of TLR-ligands such as Resiquimod, have failed due to adverse effects that limited dose and thus, efficacy. This issue could be related to pharmacokinetic properties that include fast elimination leading to low AUC with simultaneously high cmax at relevant doses. The high cmax is associated with a sharp, poorly tolerated cytokine pulse, suggesting that a compound with a higher AUC/cmax-ratio could provide a more sustained and tolerable immune activation. Our approach was to design TLR7/8-agonist Imidazoquinolines intended to partition to endosomes via acid trapping using a macrolide-carrier. This can potentially extend pharmacokinetics and simultaneously direct the compounds to the target compartment. The compounds have hTLR7/8-agonist activity (EC50 of the most active compound in cellular assays: 75-120 nM hTLR7, 2.8-3.1 µM hTLR8) and maximal hTLR7 activation between 40 and 80% of Resiquimod. The lead candidates induce secretion of IFNα from human Leukocytes in the same range as Resiquimod but induce at least 10-fold less TNFα in this system, consistent with a higher specificity for human TLR7. This pattern was reproduced in vivo in a murine system, where small molecules are thought not to activate TLR8. We found that Imidazoquinolines conjugated to a macrolide or, substances carrying an unlinked terminal secondary amine, had longer exposure compared with Resiquimod. The kinetics of pro-inflammatory cytokine release for these substances in vivo were slower and more extended (for comparable AUCs, approximately half-maximal plasma concentrations). Maximal IFNα plasma levels were reached 4 h post application. Resiquimod-treated groups had by then returned to baseline from a peak at 1 h. We propose that the characteristic cytokine profile is likely a consequence of altered pharmacokinetics and, potentially, enhanced endosomal tropism of the novel substances. In particular, our substances are designed to partition to cellular compartments where the target receptor and a distinct combination of signaling molecules relevant to IFNα-release are located. These properties could address the tolerability issues of TLR7/8 ligands and provide insight into approaches to fine-tune the outcomes of TLR7/8 activation by small molecules.
Collapse
Affiliation(s)
| | - Simon Straß
- Synovo GmbH, Tübingen, Germany
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | | - Stefan Laufer
- Pharmaceutical Chemistry, Institute for Pharmaceutical Sciences, Eberhard Karls University Tübingen, Tübingen, Germany
| | | |
Collapse
|
9
|
Michaels YS, Edgar JM, Major MC, Castle EL, Zimmerman C, Yin T, Hagner A, Lau C, Hsu HH, Ibañez-Rios MI, Durland LJ, Knapp DJHF, Zandstra PW. DLL4 and VCAM1 enhance the emergence of T cell-competent hematopoietic progenitors from human pluripotent stem cells. SCIENCE ADVANCES 2022; 8:eabn5522. [PMID: 36001668 PMCID: PMC9401626 DOI: 10.1126/sciadv.abn5522] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 07/12/2022] [Indexed: 05/13/2023]
Abstract
T cells show tremendous efficacy as cellular therapeutics. However, obtaining primary T cells from human donors is expensive and variable. Pluripotent stem cells (PSCs) have the potential to provide a renewable source of T cells, but differentiating PSCs into hematopoietic progenitors with T cell potential remains an important challenge. Here, we report an efficient serum- and feeder-free system for differentiating human PSCs into hematopoietic progenitors and T cells. This fully defined approach allowed us to study the impact of individual proteins on blood emergence and differentiation. Providing DLL4 and VCAM1 during the endothelial-to-hematopoietic transition enhanced downstream progenitor T cell output by ~80-fold. These two proteins synergized to activate notch signaling in nascent hematopoietic stem and progenitor cells, and VCAM1 additionally promoted an inflammatory transcriptional program. We also established optimized medium formulations that enabled efficient and chemically defined maturation of functional CD8αβ+, CD4-, CD3+, TCRαβ+ T cells with a diverse TCR repertoire.
Collapse
Affiliation(s)
- Yale S. Michaels
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - John M. Edgar
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Matthew C. Major
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Elizabeth L. Castle
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Carla Zimmerman
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Ting Yin
- Institute of Biomedical Engineering, University of Toronto, Toronto, Ontario M5S 3G9, Canada
| | - Andrew Hagner
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Charles Lau
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Han Hsuan Hsu
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - M. Iliana Ibañez-Rios
- Institut de recherche en immunologie et en cancérologie and Département de pathologie et biologie cellulaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Lauren J. Durland
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - David J. H. F. Knapp
- Institut de recherche en immunologie et en cancérologie and Département de pathologie et biologie cellulaire, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Peter W. Zandstra
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
- Michael Smith Laboratories, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
| |
Collapse
|
10
|
Lapp MM, Lin G, Komin A, Andrews L, Knudson M, Mossman L, Raimondi G, Arciero JC. Modeling the Potential of Treg-Based Therapies for Transplant Rejection: Effect of Dose, Timing, and Accumulation Site. Transpl Int 2022; 35:10297. [PMID: 35479106 PMCID: PMC9035492 DOI: 10.3389/ti.2022.10297] [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: 12/10/2021] [Accepted: 03/17/2022] [Indexed: 02/04/2023]
Abstract
Introduction: The adoptive transfer of regulatory T cells (Tregs) has emerged as a method to promote graft tolerance. Clinical trials have demonstrated the safety of adoptive transfer and are now assessing their therapeutic efficacy. Strategies that generate large numbers of antigen specific Tregs are even more efficacious. However, the combinations of factors that influence the outcome of adoptive transfer are too numerous to be tested experimentally. Here, mathematical modeling is used to predict the most impactful treatment scenarios. Methods: We adapted our mathematical model of murine heart transplant rejection to simulate Treg adoptive transfer and to correlate therapeutic efficacy with Treg dose and timing, frequency of administration, and distribution of injected cells. Results: The model predicts that Tregs directly accumulating to the graft are more protective than Tregs localizing to draining lymph nodes. Inhibiting antigen-presenting cell maturation and effector functions at the graft site was more effective at modulating rejection than inhibition of T cell activation in lymphoid tissues. These complex dynamics define non-intuitive relationships between graft survival and timing and frequency of adoptive transfer. Conclusion: This work provides the framework for better understanding the impact of Treg adoptive transfer and will guide experimental design to improve interventions.
Collapse
Affiliation(s)
- Maya M. Lapp
- Department of Mathematics, The College of Wooster, Wooster, OH, United States
| | - Guang Lin
- Department of Mathematics, Purdue University, West Lafayette, IN, United States
| | - Alexander Komin
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States
| | - Leah Andrews
- Department of Mathematics, St. Olaf College, Northfield, MN, United States
| | - Mei Knudson
- Department of Mathematics, Carleton College, Northfield, MN, United States
| | - Lauren Mossman
- Department of Mathematics, St. Olaf College, Northfield, MN, United States
| | - Giorgio Raimondi
- Department of Plastic and Reconstructive Surgery, Johns Hopkins School of Medicine, Baltimore, MD, United States,*Correspondence: Giorgio Raimondi, ; Julia C. Arciero,
| | - Julia C. Arciero
- Department of Mathematical Sciences, Indiana University-Purdue University of Indianapolis, Indianapolis, IN, United States,*Correspondence: Giorgio Raimondi, ; Julia C. Arciero,
| |
Collapse
|
11
|
Schmidt R, Steinhart Z, Layeghi M, Freimer JW, Bueno R, Nguyen VQ, Blaeschke F, Ye CJ, Marson A. CRISPR activation and interference screens decode stimulation responses in primary human T cells. Science 2022; 375:eabj4008. [PMID: 35113687 DOI: 10.1126/science.abj4008] [Citation(s) in RCA: 119] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Regulation of cytokine production in stimulated T cells can be disrupted in autoimmunity, immunodeficiencies, and cancer. Systematic discovery of stimulation-dependent cytokine regulators requires both loss-of-function and gain-of-function studies, which have been challenging in primary human cells. We now report genome-wide CRISPR activation (CRISPRa) and interference (CRISPRi) screens in primary human T cells to identify gene networks controlling interleukin-2 (IL-2) and interferon-γ (IFN-γ) production. Arrayed CRISPRa confirmed key hits and enabled multiplexed secretome characterization, revealing reshaped cytokine responses. Coupling CRISPRa screening with single-cell RNA sequencing enabled deep molecular characterization of screen hits, revealing how perturbations tuned T cell activation and promoted cell states characterized by distinct cytokine expression profiles. These screens reveal genes that reprogram critical immune cell functions, which could inform the design of immunotherapies.
Collapse
Affiliation(s)
- Ralf Schmidt
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Zachary Steinhart
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Madeline Layeghi
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA
| | - Jacob W Freimer
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.,Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Raymund Bueno
- Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Vinh Q Nguyen
- Department of Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Franziska Blaeschke
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA
| | - Chun Jimmie Ye
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.,Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, CA 94129, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA.,Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA 94158, USA.,Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA.,Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexander Marson
- Gladstone-UCSF Institute of Genomic Immunology, San Francisco, CA 94158, USA.,Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.,Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.,Department of Microbiology and Immunology, University of California San Francisco, San Francisco, CA 94143, USA.,Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA.,Innovative Genomics Institute, University of California Berkeley, Berkeley, CA 94720, USA.,UCSF Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA 94158, USA.,Parker Institute for Cancer Immunotherapy, University of California San Francisco, San Francisco, CA 94129, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
12
|
Boardman DA, Levings MK. Emerging strategies for treating autoimmune disorders with genetically modified Treg cells. J Allergy Clin Immunol 2022; 149:1-11. [PMID: 34998473 DOI: 10.1016/j.jaci.2021.11.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 12/17/2022]
Abstract
Gene editing of living cells is a cornerstone of present-day medical research that has enabled scientists to address fundamental biologic questions and identify novel strategies to treat diseases. The ability to manipulate adoptive cell therapy products has revolutionized cancer immunotherapy and promises similar results for the treatment of autoimmune diseases, inflammatory disorders, and transplant rejection. Clinical trials have recently deemed polyclonal regulatory T (Treg) cell therapy to be a safe therapeutic option, but questions remain regarding the efficacy of this approach. In this review, we discuss how gene editing technologies are being applied to transform the future of Treg cell therapy, focusing on the preclinical strategies that are currently being investigated to enhance the efficacy, function, and survival of human Treg cells. We explore approaches that may be used to generate immunoregulatory cells ex vivo, detail emerging strategies that are being used to modify these cells (such as using chimeric antigen receptors to confer antigen specificity), and outline concepts that have been explored to repurpose conventional T cells to target and destroy autoreactive and alloreactive lymphocytes. We also describe the key hurdles that currently hinder the clinical adoption of Treg cell therapy and propose potential future avenues of research for this field.
Collapse
Affiliation(s)
- Dominic A Boardman
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, British Columbia, Canada; BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
13
|
Armaghan F, Hajihassan Z. Engineering a variant of IL-17RA with high binding affinity to IL-17A for optimized immunotherapy. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2021; 32:e00682. [PMID: 34765462 PMCID: PMC8572878 DOI: 10.1016/j.btre.2021.e00682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 11/08/2022]
Abstract
Immunotherapy is one of the most recently used treatments for numerous cancer types and also some autoimmune and inflammatory diseases. One of the valuable targets for immunotherapy is Interleukin-17A (IL-17A) or its receptor (IL-17RA) because overexpression of IL-17A as a pro-inflammatory cytokine is associated with several inflammatory, autoimmune and cancer diseases. In this study, the extracellular domain of IL-17RA involved in binding to IL-17A was mutated by using R software to achieve a variant with increased binding affinity to IL-17A. The ∆∆G value of -30.89 kcal/mol was calculated for the best variant (385) with point mutations of R265N, N91T, and W31K using the FoldX module. Also, the KD for its interaction with IL-17A was calculated 0.06 nM by surface plasmon resonance (SPR) technique. Our results indicated that variant 385 could bind to IL-17A with higher binding affinity than wild-type one, so, it can be a good therapeutic candidate for blocking IL-17A.
Collapse
Affiliation(s)
- Fatemeh Armaghan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Zahra Hajihassan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
14
|
Shanti A, Hallfors N, Petroianu GA, Planelles L, Stefanini C. Lymph Nodes-On-Chip: Promising Immune Platforms for Pharmacological and Toxicological Applications. Front Pharmacol 2021; 12:711307. [PMID: 34483920 PMCID: PMC8415712 DOI: 10.3389/fphar.2021.711307] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022] Open
Abstract
Organs-on-chip are gaining increasing attention as promising platforms for drug screening and testing applications. However, lymph nodes-on-chip options remain limited although the lymph node is one of the main determinants of the immunotoxicity of newly developed pharmacological drugs. In this review, we describe existing biomimetic lymph nodes-on-chip, their design, and their physiological relevance to pharmacology and shed the light on future directions associated with lymph node-on-chip design and implementation in drug discovery and development.
Collapse
Affiliation(s)
- Aya Shanti
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Nicholas Hallfors
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Georg A Petroianu
- College of Medicine and Health Sciences, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Lourdes Planelles
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| | - Cesare Stefanini
- Healthcare Engineering Innovation Center, Biomedical Engineering Department, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
| |
Collapse
|
15
|
Fung VCW, Rosado-Sánchez I, Levings MK. Transduction of Human T Cell Subsets with Lentivirus. Methods Mol Biol 2021; 2285:227-254. [PMID: 33928557 DOI: 10.1007/978-1-0716-1311-5_19] [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] [Indexed: 02/28/2023]
Abstract
Lentivirus-mediated gene transfer is an efficient method to introduce a variety of transgenes to human T cells. Here we describe a protocol to transduce human CD4+, CD8+, or CD4+ regulatory T cells. To illustrate the method, we use transduction with lentivirus encoding an HLA-A2-specific chimeric antigen receptor (CAR) and a transduction marker as an example. Methods to isolate, transduce, purify, and expand CD4+ and CD8+ T cells as well as regulatory T cells are provided. We also describe how to carry out cytotoxicity or suppression assays to assess the function of the resulting CAR T cell or CAR regulatory T cells, respectively.
Collapse
Affiliation(s)
- Vivian C W Fung
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada.,BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Isaac Rosado-Sánchez
- BC Children's Hospital Research Institute, Vancouver, BC, Canada.,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada. .,BC Children's Hospital Research Institute, Vancouver, BC, Canada. .,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
16
|
Nagy NA, de Haas AM, Geijtenbeek TBH, van Ree R, Tas SW, van Kooyk Y, de Jong EC. Therapeutic Liposomal Vaccines for Dendritic Cell Activation or Tolerance. Front Immunol 2021; 12:674048. [PMID: 34054859 PMCID: PMC8155586 DOI: 10.3389/fimmu.2021.674048] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/26/2021] [Indexed: 12/15/2022] Open
Abstract
Dendritic cells (DCs) are paramount in initiating and guiding immunity towards a state of activation or tolerance. This bidirectional capacity of DCs sets them at the center stage for treatment of cancer and autoimmune or allergic conditions. Accordingly, many clinical studies use ex vivo DC vaccination as a strategy to boost anti-tumor immunity or to suppress immunity by including vitamin D3, NF-κB inhibitors or retinoic acid to create tolerogenic DCs. As harvesting DCs from patients and differentiating these cells in vitro is a costly and cumbersome process, in vivo targeting of DCs has huge potential as nanoparticulate platforms equipped with activating or tolerogenic adjuvants can modulate DCs in their natural environment. There is a rapid expansion of the choices of nanoparticles and activation- or tolerance-promoting adjuvants for a therapeutic vaccine platform. In this review we highlight the most recent nanomedical approaches aimed at inducing immune activation or tolerance via targeting DCs, together with novel fundamental insights into the mechanisms inherent to fostering anti-tumor or tolerogenic immunity.
Collapse
Affiliation(s)
- Noémi Anna Nagy
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Aram M. de Haas
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Teunis B. H. Geijtenbeek
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| | - Ronald van Ree
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Otorhinolaryngology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Sander W. Tas
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
- Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Amsterdam Rheumatology and Immunology Center, University of Amsterdam, Amsterdam, Netherlands
| | - Yvette van Kooyk
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center, Cancer Center Amsterdam, Amsterdam Institute for Infection and Immunity, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Esther C. de Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, Amsterdam Institute for Infection and Immunity, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
17
|
Jindal A, Sarkar S, Alam A. Nanomaterials-Mediated Immunomodulation for Cancer Therapeutics. Front Chem 2021; 9:629635. [PMID: 33708759 PMCID: PMC7940769 DOI: 10.3389/fchem.2021.629635] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 01/18/2021] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy holds great promise in overcoming the limitations of conventional regimens for cancer therapeutics. There is growing interest among researchers and clinicians to develop novel immune-strategies for cancer diagnosis and treatment with better specificity and lesser adversity. Immunomodulation-based cancer therapies are rapidly emerging as an alternative approach that employs the host's own defense mechanisms to recognize and selectively eliminate cancerous cells. Recent advances in nanotechnology have pioneered a revolution in the field of cancer therapy. Several nanomaterials (NMs) have been utilized to surmount the challenges of conventional anti-cancer treatments like cytotoxic chemotherapy, radiation, and surgery. NMs offer a plethora of exceptional features such as a large surface area to volume ratio, effective loading, and controlled release of active drugs, tunable dimensions, and high stability. Moreover, they also possess the inherent property of interacting with living cells and altering the immune responses. However, the interaction between NMs and the immune system can give rise to unanticipated adverse reactions such as inflammation, necrosis, and hypersensitivity. Therefore, to ensure a successful and safe clinical application of immunomodulatory nanomaterials, it is imperative to acquire in-depth knowledge and a clear understanding of the complex nature of the interactions between NMs and the immune system. This review is aimed at providing an overview of the recent developments, achievements, and challenges in the application of immunomodulatory nanomaterials (iNMs) for cancer therapeutics with a focus on elucidating the mechanisms involved in the interplay between NMs and the host's immune system.
Collapse
Affiliation(s)
- Ajita Jindal
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sounik Sarkar
- Flowcytometry Facility, Modern Biology Department, University of Calcutta, Kolkata, India
| | - Aftab Alam
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
- Clare Hall, University of Cambridge, Cambridge, United Kingdom
- Charles River Laboratories, Cambridge Biomedical Campus, Cambridge, United Kingdom
| |
Collapse
|
18
|
Wu D, Wong MQ, Vent-Schmidt J, Boardman DA, Steiner TS, Levings MK. A method for expansion and retroviral transduction of mouse regulatory T cells. J Immunol Methods 2021; 488:112931. [PMID: 33221458 DOI: 10.1016/j.jim.2020.112931] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 10/18/2020] [Accepted: 11/16/2020] [Indexed: 02/05/2023]
Abstract
Adoptive cell therapy with genetically modified regulatory T cells (Tregs) is under clinical investigation for the treatment of transplant rejection and various autoimmune conditions. A limitation of modelling this approach in mice is the lack of optimized protocols for expanding and transducing mouse Tregs. Here we describe a protocol for purifying, expanding and retrovirally transducing mouse Tregs with a vector encoding a chimeric antigen receptor as a model transgene. We found that isolation of Tregs from C57Bl/6J Foxp3EGFP mice solely based on eGFP expression resulted in sufficiently pure cells; co-sorting of CD25hi cells was not essential. Although expansion with rapamycin reduced Treg expansion, it promoted maximal in vitro suppressive activity. Retroviral transduction of Tregs following 2 days of stimulation with anti-CD3/CD28 beads achieved a transduction efficiency of ~40% and did not impair their suppressive capacity. When injected into a conventional T cell (Tconv)-transfer-induced colitis model, transduced Tregs inhibited colitis progression at ratios as low as 1 Treg to 100 Tconvs, and maintained Foxp3 and transgene expression throughout an 8-week period. This method facilitates the study of transduced Tregs in animal models and will enable the study of genetically engineered Treg therapy for a variety of inflammatory diseases.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- Cell Proliferation
- Cells, Cultured
- Colitis/genetics
- Colitis/immunology
- Colitis/metabolism
- Colitis/prevention & control
- Disease Models, Animal
- Flow Cytometry
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Genes, T-Cell Receptor beta
- Genetic Vectors
- Green Fluorescent Proteins/genetics
- Green Fluorescent Proteins/metabolism
- Immunomagnetic Separation
- Mice, Inbred C57BL
- Mice, Knockout
- Phenotype
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Retroviridae/genetics
- Retroviridae/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Transduction, Genetic
- Mice
Collapse
Affiliation(s)
- Dan Wu
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - May Q Wong
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Jens Vent-Schmidt
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Dominic A Boardman
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - Theodore S Steiner
- BC Children's Hospital Research Institute, Vancouver, BC, Canada; Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada; BC Children's Hospital Research Institute, Vancouver, BC, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
19
|
Building a CAR-Treg: Going from the basic to the luxury model. Cell Immunol 2020; 358:104220. [DOI: 10.1016/j.cellimm.2020.104220] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 09/13/2020] [Accepted: 09/17/2020] [Indexed: 01/10/2023]
|
20
|
Novel human immunomodulatory T cell receptors and their double-edged potential in autoimmunity, cardiovascular disease and cancer. Cell Mol Immunol 2020; 18:919-935. [PMID: 33235388 DOI: 10.1038/s41423-020-00586-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
In the last decade, approaches based on T cells and their immunomodulatory receptors have emerged as a solid improvement in treatments for various types of cancer. However, the roles of these molecules in the therapeutic context of autoimmune and cardiovascular diseases are still relatively unexplored. Here, we review the best known and most commonly used immunomodulatory T cell receptors in clinical practice (PD-1 and CTLA-4), along with the rest of the receptors with known functions in animal models, which have great potential as modulators in human pathologies in the medium term. Among these other receptors is the receptor CD69, which has recently been described to be expressed in mouse and human T cells in autoimmune and cardiovascular diseases and cancer. However, inhibition of these receptors individually or in combination by drugs or monoclonal antibodies generates a loss of immunological tolerance and can trigger multiple autoimmune disorders in different organs and immune-related adverse effects. In the coming decades, knowledge on the functions of different immunomodulatory receptors will be pivotal for the development of new and better therapies with less harmful side effects. In this review, we discuss the roles of these receptors in the control of immunity from a perspective focused on therapeutic potential in not only cancer but also autoimmune diseases, such as systemic lupus erythematosus, autoimmune diabetes and rheumatoid arthritis, and cardiovascular diseases, such as atherosclerosis, acute myocardial infarction, and myocarditis.
Collapse
|
21
|
De Paula Pohl A, Schmidt A, Zhang AH, Maldonado T, Königs C, Scott DW. Engineered regulatory T cells expressing myelin-specific chimeric antigen receptors suppress EAE progression. Cell Immunol 2020; 358:104222. [PMID: 33053469 DOI: 10.1016/j.cellimm.2020.104222] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/25/2022]
Abstract
The expansion of polyclonal T regulatory cells (Tregs) offers great promise for the treatment of immune-mediated diseases, such as multiple sclerosis (MS). However, polyclonal Tregs can be non-specifically immunosuppressive. Based on the advancements with chimeric antigen receptor (CAR) therapy in leukemia, we previously engineered Tregs to express a T-cell receptor (TCR) specific for a myelin basic protein (MBP) peptide. These TCR-engineered specific Tregs suppressed the proliferation of MBP-reactive T effector cells and ameliorated myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE). Herein, we extend this approach by creating human regulatory T cells expressing functional single-chain chimeric antigen receptors (scFv CAR), targeting either MBP or MOG. These scFv CAR-transduced Tregs retained FoxP3 and Helios, characteristic of Treg cells, after long-term expansion in vitro. Importantly, these engineered CNS targeting CAR-Tregs were able to suppress autoimmune pathology in EAE, demonstrating that these Tregs have the potential to be used as a cellular therapy for MS patients.
Collapse
Affiliation(s)
- Alessandra De Paula Pohl
- Department of Medicine, Uniformed Services University of the Health and Sciences, Bethesda, MD, United States
| | - Anja Schmidt
- Department of Pediatrics, Molecular Hemostasis and Immunodeficiency, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Ai-Hong Zhang
- Department of Medicine, Uniformed Services University of the Health and Sciences, Bethesda, MD, United States
| | - Tania Maldonado
- Department of Medicine, Uniformed Services University of the Health and Sciences, Bethesda, MD, United States
| | - Christoph Königs
- Department of Pediatrics, Molecular Hemostasis and Immunodeficiency, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - David W Scott
- Department of Medicine, Uniformed Services University of the Health and Sciences, Bethesda, MD, United States.
| |
Collapse
|
22
|
Dawson NAJ, Rosado-Sánchez I, Novakovsky GE, Fung VCW, Huang Q, McIver E, Sun G, Gillies J, Speck M, Orban PC, Mojibian M, Levings MK. Functional effects of chimeric antigen receptor co-receptor signaling domains in human regulatory T cells. Sci Transl Med 2020; 12:12/557/eaaz3866. [DOI: 10.1126/scitranslmed.aaz3866] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 06/26/2020] [Indexed: 12/12/2022]
Abstract
Antigen-specific regulatory T cells (Tregs) engineered with chimeric antigen receptors (CARs) are a potent immunosuppressive cellular therapy in multiple disease models and could overcome shortcomings of polyclonal Treg therapy. CAR therapy was initially developed with conventional T cells, which have different signaling requirements than do Tregs. To date, most of the CAR Treg studies used second-generation CARs, encoding a CD28 or 4-1BB co-receptor signaling domain and CD3ζ, but it was not known if this CAR design was optimal for Tregs. Using a human leukocyte antigen–A2–specific CAR platform and human Tregs, we compared 10 CARs with different co-receptor signaling domains and systematically tested their function and CAR-stimulated gene expression profile. Tregs expressing a CAR encoding CD28wt were markedly superior to all other CARs tested in an in vivo model of graft-versus-host disease. In vitro assays revealed stable expression of Helios and an ability to suppress CD80 expression on dendritic cells as key in vitro predictors of in vivo function. This comprehensive study of CAR signaling domain variants in Tregs can be leveraged to optimize CAR design for use in antigen-specific Treg therapy.
Collapse
Affiliation(s)
- Nicholas A. J. Dawson
- Department of Medicine, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Isaac Rosado-Sánchez
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - German E. Novakovsky
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Vivian C. W. Fung
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Qing Huang
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Emma McIver
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Grace Sun
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Jana Gillies
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Madeleine Speck
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Paul C. Orban
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Majid Mojibian
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Megan K. Levings
- BC Children’s Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| |
Collapse
|
23
|
Rana J, Biswas M. Regulatory T cell therapy: Current and future design perspectives. Cell Immunol 2020; 356:104193. [PMID: 32823038 DOI: 10.1016/j.cellimm.2020.104193] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 07/30/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Regulatory T cells (Tregs) maintain immune equilibrium by suppressing immune responses through various multistep contact dependent and independent mechanisms. Cellular therapy using polyclonal Tregs in transplantation and autoimmune diseases has shown promise in preclinical models and clinical trials. Although novel approaches have been developed to improve specificity and efficacy of antigen specific Treg based therapies, widespread application is currently restricted. To date, design-based approaches to improve the potency and persistence of engineered chimeric antigen receptor (CAR) Tregs are limited. Here, we describe currently available Treg based therapies, their advantages and limitations for implementation in clinical studies. We also examine various strategies for improving CAR T cell design that can potentially be applied to CAR Tregs, such as identifying co-stimulatory signalling domains that enhance suppressive ability, determining optimal scFv affinity/avidity, and co-expression of accessory molecules. Finally, we discuss the importance of tailoring CAR Treg design to suit the individual disease.
Collapse
Affiliation(s)
- Jyoti Rana
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Moanaro Biswas
- Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
24
|
Klöß S, Dehmel S, Braun A, Parnham MJ, Köhl U, Schiffmann S. From Cancer to Immune-Mediated Diseases and Tolerance Induction: Lessons Learned From Immune Oncology and Classical Anti-cancer Treatment. Front Immunol 2020; 11:1423. [PMID: 32733473 PMCID: PMC7360838 DOI: 10.3389/fimmu.2020.01423] [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: 01/15/2020] [Accepted: 06/02/2020] [Indexed: 12/27/2022] Open
Abstract
Success in cancer treatment over the last four decades has ranged from improvements in classical drug therapy to immune oncology. Anti-cancer drugs have also often proven beneficial for the treatment of inflammatory and autoimmune diseases. In this review, we report on challenging examples that bridge between treatment of cancer and immune-mediated diseases, addressing mechanisms and experimental models as well as clinical investigations. Patient-derived tumor xenograft (PDX) (humanized) mouse models represent useful tools for preclinical evaluation of new therapies and biomarker identification. However, new developments using human ex vivo approaches modeling cancer, for example in microfluidic human organs-on-chips, promise to identify key molecular, cellular and immunological features of human cancer progression in a fully human setting. Classical drugs which bridge the gap, for instance, include cytotoxic drugs, proteasome inhibitors, PI3K/mTOR inhibitors and metabolic inhibitors. Biologicals developed for cancer therapy have also shown efficacy in the treatment of autoimmune diseases. In immune oncology, redirected chimeric antigen receptor (CAR) T cells have achieved spectacular remissions in refractory B cell leukemia and lymphoma and are currently under development for tolerance induction using cell-based therapies such as CAR Tregs or NK cells. Finally, a brief outline will be given of the lessons learned from bridging cancer and autoimmune diseases as well as tolerance induction.
Collapse
Affiliation(s)
- Stephan Klöß
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Cellular Therapeutics, Hannover Medical School (MHH), Hanover, Germany
| | - Susann Dehmel
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine (ITEM), Hanover, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Frankfurt, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany
| | - Ulrike Köhl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Leipzig, Germany.,Institute of Cellular Therapeutics, Hannover Medical School (MHH), Hanover, Germany.,Fraunhofer Cluster of Excellence for Immune-Mediated Diseases (CIMD), Frankfurt, Germany.,Institute of Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Susanne Schiffmann
- Institute of Clinical Pharmacology, University Hospital Frankfurt, Frankfurt, Germany.,Fraunhofer Institute for Molecular Biology and Applied Ecology (IME), Branch for Translational Medicine and Pharmacology (TMP), Frankfurt, Germany
| |
Collapse
|
25
|
Raffin C, Vo LT, Bluestone JA. T reg cell-based therapies: challenges and perspectives. Nat Rev Immunol 2020; 20:158-172. [PMID: 31811270 PMCID: PMC7814338 DOI: 10.1038/s41577-019-0232-6] [Citation(s) in RCA: 408] [Impact Index Per Article: 102.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/07/2019] [Indexed: 12/25/2022]
Abstract
Cellular therapies using regulatory T (Treg) cells are currently undergoing clinical trials for the treatment of autoimmune diseases, transplant rejection and graft-versus-host disease. In this Review, we discuss the biology of Treg cells and describe new efforts in Treg cell engineering to enhance specificity, stability, functional activity and delivery. Finally, we envision that the success of Treg cell therapy in autoimmunity and transplantation will encourage the clinical use of adoptive Treg cell therapy for non-immune diseases, such as neurological disorders and tissue repair.
Collapse
Affiliation(s)
- Caroline Raffin
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Linda T Vo
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA
| | - Jeffrey A Bluestone
- Sean N. Parker Autoimmune Research Laboratory, Diabetes Center, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|