151
|
Rodríguez-Lobato LG, Ganzetti M, Fernández de Larrea C, Hudecek M, Einsele H, Danhof S. CAR T-Cells in Multiple Myeloma: State of the Art and Future Directions. Front Oncol 2020; 10:1243. [PMID: 32850376 PMCID: PMC7399644 DOI: 10.3389/fonc.2020.01243] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 06/16/2020] [Indexed: 01/24/2023] Open
Abstract
Despite recent therapeutic advances, the prognosis of multiple myeloma (MM) patients remains poor. Thus, new strategies to improve outcomes are imperative. Chimeric antigen receptor (CAR) T-cell therapy has changed the treatment landscape of B-cell malignancies, providing a potentially curative option for patients who are refractory to standard treatment. Long-term remissions achieved in patients with acute lymphoblastic leukemia and Non-Hodgkin Lymphoma encouraged its further development in MM. B-cell maturation antigen (BCMA)-targeted CAR T-cells have established outstanding results in heavily pre-treated patients. However, several other antigens such as SLAMF7 and CD44v6 are currently under investigation with promising results. Idecabtagene vicleucel is expected to be approved soon for clinical use. Unfortunately, relapses after CAR T-cell infusion have been reported. Hence, understanding the underlying mechanisms of resistance is essential to promote prevention strategies and to enhance CAR T-cell efficacy. In this review we provide an update of the most recent clinical and pre-clinical data and we elucidate both, the potential and the challenges of CAR T-cell therapy in the future.
Collapse
Affiliation(s)
- Luis Gerardo Rodríguez-Lobato
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maya Ganzetti
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
- Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carlos Fernández de Larrea
- Amyloidosis and Multiple Myeloma Unit, Department of Hematology, Hospital Clínic of Barcelona, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Michael Hudecek
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Division of Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
152
|
Namdari H, Rezaei F, Teymoori-Rad M, Mortezagholi S, Sadeghi A, Akbari A. CAR T cells: Living HIV drugs. Rev Med Virol 2020; 30:1-14. [PMID: 32713110 DOI: 10.1002/rmv.2139] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 06/04/2020] [Accepted: 06/09/2020] [Indexed: 12/29/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1), the virus that causes AIDS (acquired immunodeficiency syndrome), is a major global public health issue. Although the advent of combined antiretroviral therapy (ART) has made significant progress in inhibiting HIV replication in patients, HIV-infected cells remain the principal cellular reservoir of HIV, this allows HIV to rebound immediately upon stopping ART, which is considered the major obstacle to curing HIV infection. Chimeric antigen receptor (CAR) cell therapy has provided new opportunities for HIV treatment. Engineering T cells or hematopoietic stem cells (HSCs) to generate CAR T cells is a rapidly growing approach to develop an efficient immune cell to fight HIV. Herein, we review preclinical and clinical data available for the development of CAR T cells. Further, the advantages and disadvantages of clinical application of anti-HIV CAR T cells will be discussed.
Collapse
Affiliation(s)
- Haideh Namdari
- Iranian Tissue Bank Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Farhad Rezaei
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Sahar Mortezagholi
- Department of Immunology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmadreza Sadeghi
- Iranian Tissue Bank Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Akbari
- Colorectal Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
153
|
Dual Targeting of Mesothelin and CD19 with Chimeric Antigen Receptor-Modified T Cells in Patients with Metastatic Pancreatic Cancer. Mol Ther 2020; 28:2367-2378. [PMID: 32730744 DOI: 10.1016/j.ymthe.2020.07.017] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/12/2020] [Accepted: 07/10/2020] [Indexed: 12/16/2022] Open
Abstract
B cells infiltrate pancreatic ductal adenocarcinoma (PDAC) and in preclinical cancer models, can suppress T cell immunosurveillance in cancer. Here, we conducted a pilot study to assess the safety and feasibility of administering lentiviral-transduced chimeric antigen receptor (CAR)-modified autologous T cells redirected against mesothelin to target tumor cells along with CART cells redirected against CD19 to deplete B cells. Both CARs contained 4-1BB and CD3ζ signaling domains. Three patients with chemotherapy-refractory PDAC received 1.5 g/m2 cyclophosphamide prior to separate infusions of lentiviral-transduced T cells engineered to express chimeric anti-mesothelin immunoreceptor SS1 (CART-Meso, 3 × 107/m2) and chimeric anti-CD19 immunoreceptor (CART-19, 3 × 107/m2). Treatment was well tolerated without dose-limiting toxicities. Best response was stable disease (1 of 3 patients). CART-19 (compared to CART-Meso) cells showed the greatest expansion in the blood, although persistence was transient. B cells were successfully depleted in all subjects, became undetectable by 7-10 days post-infusion, and remained undetectable for at least 28 days. Together, concomitant delivery of CART-Meso and CART-19 cells in patients with PDAC is safe. CART-19 cells deplete normal B cells but at the dose tested in these 3 subjects did not improve CART-Meso cell persistence.
Collapse
|
154
|
Chen GM, Azzam A, Ding YY, Barrett DM, Grupp SA, Tan K. Dissecting the Tumor-Immune Landscape in Chimeric Antigen Receptor T-cell Therapy: Key Challenges and Opportunities for a Systems Immunology Approach. Clin Cancer Res 2020; 26:3505-3513. [PMID: 32127393 PMCID: PMC7367708 DOI: 10.1158/1078-0432.ccr-19-3888] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 01/15/2020] [Accepted: 02/27/2020] [Indexed: 12/17/2022]
Abstract
The adoptive transfer of genetically engineered chimeric antigen receptor (CAR) T cells has opened a new frontier in cancer therapy. Unlike the paradigm of targeted therapies, the efficacy of CAR T-cell therapy depends not only on the choice of target but also on a complex interplay of tumor, immune, and stromal cell communication. This presents both challenges and opportunities from a discovery standpoint. Whereas cancer consortia have traditionally focused on the genomic, transcriptomic, epigenomic, and proteomic landscape of cancer cells, there is an increasing need to expand studies to analyze the interactions between tumor, immune, and stromal cell populations in their relevant anatomical and functional compartments. Here, we focus on the promising application of systems biology to address key challenges in CAR T-cell therapy, from understanding the mechanisms of therapeutic resistance in hematologic and solid tumors to addressing important clinical challenges in biomarker discovery and therapeutic toxicity. We propose a systems biology view of key clinical objectives in CAR T-cell therapy and suggest a path forward for a biomedical discovery process that leverages modern technological approaches in systems biology.
Collapse
Affiliation(s)
- Gregory M Chen
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew Azzam
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Yang-Yang Ding
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - David M Barrett
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stephan A Grupp
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Division of Oncology, Cancer Immunotherapy Program, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kai Tan
- Division of Oncology, Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Graduate Group in Genomics and Computational Biology, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
155
|
Lee PH, Keller MD, Hanley PJ, Bollard CM. Virus-Specific T Cell Therapies for HIV: Lessons Learned From Hematopoietic Stem Cell Transplantation. Front Cell Infect Microbiol 2020; 10:298. [PMID: 32775304 PMCID: PMC7381350 DOI: 10.3389/fcimb.2020.00298] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/19/2020] [Indexed: 12/20/2022] Open
Abstract
Human immunodeficiency virus (HIV) has caused millions of deaths and continues to threaten the health of millions of people worldwide. Despite anti-retroviral therapy (ART) substantially alleviating severity and limiting transmission, HIV has not been eradicated and its persistence can lead to other health concerns such as cancer. The only two cases of HIV cure to date are HIV+ cancer patients receiving an allogeneic hematopoietic stem cell transplantation (allo-HSCT) from a donor with the CCR5 Δ32 mutation. While this approach has not led to such success in other patients and is not applicable to HIV+ individuals without cancer, the encouraging results may point toward a breakthrough in developing a cure strategy for HIV. Adoptive transfer of virus-specific T cells (VSTs) post HSCT has been effectively used to treat and prevent reactivation of latent viral infections such as cytomegalovirus (CMV) and Epstein-Barr virus (EBV), making VSTs an attractive therapeutic to control HIV rebound. Here we will discuss the potential of using adoptive T cell therapies in combination with other treatments such as HSCT and latency reversing agents (LRAs) to achieve a functional cure for HIV.
Collapse
Affiliation(s)
- Ping-Hsien Lee
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, United States
| | - Michael D Keller
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, United States.,Division of Allergy & Immunology, Children's National Hospital, Washington, DC, United States
| | - Patrick J Hanley
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, United States.,Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, United States.,GW Cancer Center, The George Washington University, Washington, DC, United States
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital, Washington, DC, United States.,Division of Blood and Marrow Transplantation, Children's National Hospital, Washington, DC, United States.,GW Cancer Center, The George Washington University, Washington, DC, United States
| |
Collapse
|
156
|
Yang F, Shi K, Jia YP, Hao Y, Peng JR, Qian ZY. Advanced biomaterials for cancer immunotherapy. Acta Pharmacol Sin 2020; 41:911-927. [PMID: 32123302 PMCID: PMC7468530 DOI: 10.1038/s41401-020-0372-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023] Open
Abstract
Immunotherapy, as a powerful strategy for cancer treatment, has achieved tremendous efficacy in clinical trials. Despite these advancements, there is much to do in terms of enhancing therapeutic benefits and decreasing the side effects of cancer immunotherapy. Advanced nanobiomaterials, including liposomes, polymers, and silica, play a vital role in the codelivery of drugs and immunomodulators. These nanobiomaterial-based delivery systems could effectively promote antitumor immune responses and simultaneously reduce toxic adverse effects. Furthermore, nanobiomaterials may also combine with each other or with traditional drugs via different mechanisms, thus giving rise to more accurate and efficient tumor treatment. Here, an overview of the latest advancement in these nanobiomaterials used for cancer immunotherapy is given, describing outstanding systems, including lipid-based nanoparticles, polymer-based scaffolds or micelles, inorganic nanosystems, and others.
Collapse
Affiliation(s)
- Fan Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Kun Shi
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Yan-Peng Jia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Ying Hao
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Jin-Rong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China
| | - Zhi-Yong Qian
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
157
|
El-Galaly TC, Cheah CY, Kristensen D, Hutchison A, Hay K, Callréus T, Villa D. Potentials, challenges and future of chimeric antigen receptor T-cell therapy in non-Hodgkin lymphomas. Acta Oncol 2020; 59:766-774. [PMID: 32189546 DOI: 10.1080/0284186x.2020.1741680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background: Diffuse large B-cell lymphoma (DLBCL) is the most common lymphoma subtype. Disease progression or relapse following frontline chemoimmunotherapy, largely in the form of standard R-CHOP, occurs in 30-40% patients. Relapsed/refractory (R/R) DLBCL represents a major unmet medical need. In particular, patients with primary refractory disease or those whose lymphoma relapses after autologous stem cell transplantation have historically had poor outcomes.Material and methods: Chimeric antigen receptor T-cell (CART) therapy is a promising novel treatment with curative potential in this setting. CART is based on ex vivo genetic modification of autologous T-cells to express chimeric receptors targeting antigens highly expressed in tumors such as CD19 in DLBCL. After lymphocyte-depleting therapy, patients are infused with CARTs that expand in vivo and target CD19-positive lymphoma cells.Results: In initial phase I-II trials, investigators have demonstrated complete responses in 40-50% of patients with R/R DLBCL, resulting in durable remission approaching 3 years of follow-up in most of these patients without further treatment. The logistics of delivery are complex as cell products require timely long-distance transfer between hospitals and production facilities. The unique toxicity profile of CARTs, including the risk of fatal immunological and neurologic events, also requires specific hospital wide management approaches and education. The substantial direct and indirect costs of CART will limit access even in countries with well resourced health care systems.Conclusions: While only two products are commercially available at present, further approvals in coming years appear likely. Future directions include CARTs with reactivity to tumor antigens other than CD19 and products targeting multiple tumor antigens to overcome resistance. The availability of CART has altered the current treatment algorithm for R/R DLBCL, and indications will likely expand to earlier lines of therapy and other hematologic malignancies.
Collapse
Affiliation(s)
- Tarec Christoffer El-Galaly
- Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
- Clinical Cancer Research Centre, Aalborg University Hospital, Aalborg, Denmark
- Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Chan Yoon Cheah
- Department of Haematology, Sir Gairdner Hospital, Nedlands, Australia
- Department of Haematology, Pathwest Laboratory Medicine, Nedlands, Australia
- Medical School, University of Western Australia, Crawley, Australia
| | - Daniel Kristensen
- Department of Haematology, Aalborg University Hospital, Aalborg, Denmark
| | - Andrew Hutchison
- Department of Haematology, Fiona Stanley Hospital, Murdoch, Australia
| | - Kevin Hay
- Leukemia/Bone Marrow Transplant Program of BC and BC Cancer Research Centre, Vancouver, Canada
| | - Torbjörn Callréus
- Department of Drug Design and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | - Diego Villa
- BC Cancer Centre for Lymphoid Cancer, Vancouver, Canada
| |
Collapse
|
158
|
Schober K, Müller TR, Busch DH. Orthotopic T-Cell Receptor Replacement-An "Enabler" for TCR-Based Therapies. Cells 2020; 9:E1367. [PMID: 32492858 PMCID: PMC7348731 DOI: 10.3390/cells9061367] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/25/2020] [Accepted: 05/28/2020] [Indexed: 12/18/2022] Open
Abstract
Natural adaptive immunity co-evolved with pathogens over millions of years, and adoptive transfer of non-engineered T cells to fight infections or cancer so far exhibits an exceptionally safe and functional therapeutic profile in clinical trials. However, the personalized nature of therapies using virus-specific T cells, donor lymphocyte infusion, or tumor-infiltrating lymphocytes makes implementation in routine clinical care difficult. In principle, genetic engineering can be used to make T-cell therapies more broadly applicable, but so far it significantly alters the physiology of cells. We recently demonstrated that orthotopic T-cell receptor (TCR) replacement (OTR) by clustered regularly interspaced short palindromic repeats (CRISPR)/ CRISPR-associated protein 9 (Cas9) can be used to generate engineered T cells with preservation of near-physiological function. In this review, we present the current status of OTR technology development and discuss its potential for TCR-based therapies. By providing the means to combine the therapeutic efficacy and safety profile of physiological T cells with the versatility of cell engineering, OTR can serve as an "enabler" for TCR-based therapies.
Collapse
Affiliation(s)
- Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), 81675 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Thomas R. Müller
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), 81675 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| | - Dirk H. Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), 81675 Munich, Germany;
- German Center for Infection Research (DZIF), Partner Site Munich, 81675 Munich, Germany
| |
Collapse
|
159
|
Xue T, Budde LE. Immunotherapies Targeting CD123 for Blastic Plasmacytoid Dendritic Cell Neoplasm. Hematol Oncol Clin North Am 2020; 34:575-587. [DOI: 10.1016/j.hoc.2020.01.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
160
|
HIV-1-Specific Chimeric Antigen Receptor T Cells Fail To Recognize and Eliminate the Follicular Dendritic Cell HIV Reservoir In Vitro. J Virol 2020; 94:JVI.00190-20. [PMID: 32161179 DOI: 10.1128/jvi.00190-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
The major obstacle to a cure for HIV infection is the persistence of replication-competent viral reservoirs during antiretroviral therapy. HIV-specific chimeric antigen receptor (CAR) T cells have been developed to target latently infected CD4+ T cells that express virus either spontaneously or after intentional latency reversal. Whether HIV-specific CAR-T cells can recognize and eliminate the follicular dendritic cell (FDC) reservoir of HIV-bound immune complexes (ICs) is unknown. We created HIV-specific CAR-T cells using human peripheral blood mononuclear cells (PBMCs) and a CAR construct that enables the expression of CD4 (domains 1 and 2) and the carbohydrate recognition domain of mannose binding lectin (MBL) to target native HIV Env (CD4-MBL CAR). We assessed CAR-T cell cytotoxicity using a carboxyfluorescein succinimidyl ester (CFSE) release assay and evaluated CAR-T cell activation through interferon gamma (IFN-γ) production and CD107a membrane accumulation by flow cytometry. CD4-MBL CAR-T cells displayed potent lytic and functional responses to Env-expressing cell lines and HIV-infected CD4+ T cells but were ineffective at targeting FDC bearing HIV-ICs. CD4-MBL CAR-T cells were unresponsive to cell-free HIV or concentrated, immobilized HIV-ICs in cell-free experiments. Blocking intercellular adhesion molecule-1 (ICAM-1) inhibited the cytolytic response of CD4-MBL CAR-T cells to Env-expressing cell lines and HIV-infected CD4+ T cells, suggesting that factors such as adhesion molecules are necessary for the stabilization of the CAR-Env interaction to elicit a cytotoxic response. Thus, CD4-MBL CAR-T cells are unable to eliminate the FDC-associated HIV reservoir, and alternative strategies to eradicate this reservoir must be sought.IMPORTANCE Efforts to cure HIV infection have focused primarily on the elimination of latently infected CD4+ T cells. Few studies have addressed the unique reservoir of infectious HIV that exists on follicular dendritic cells (FDCs), persists in vivo during antiretroviral therapy, and likely contributes to viral rebound upon cessation of antiretroviral therapy. We assessed the efficacy of a novel HIV-specific chimeric antigen receptor (CAR) T cell to target both HIV-infected CD4+ T cells and the FDC reservoir in vitro Although CAR-T cells eliminated CD4+ T cells that express HIV, they did not respond to or eliminate FDC bound to HIV. These findings reveal a fundamental limitation to CAR-T cell therapy to eradicate HIV.
Collapse
|
161
|
Abdalla AME, Xiao L, Miao Y, Huang L, Fadlallah GM, Gauthier M, Ouyang C, Yang G. Nanotechnology Promotes Genetic and Functional Modifications of Therapeutic T Cells Against Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903164. [PMID: 32440473 PMCID: PMC7237845 DOI: 10.1002/advs.201903164] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/23/2020] [Indexed: 05/24/2023]
Abstract
Growing experience with engineered chimeric antigen receptor (CAR)-T cells has revealed some of the challenges associated with developing patient-specific therapy. The promising clinical results obtained with CAR-T therapy nevertheless demonstrate the urgency of advancements to promote and expand its uses. There is indeed a need to devise novel methods to generate potent CARs, and to confer them and track their anti-tumor efficacy in CAR-T therapy. A potentially effective approach to improve the efficacy of CAR-T cell therapy would be to exploit the benefits of nanotechnology. This report highlights the current limitations of CAR-T immunotherapy and pinpoints potential opportunities and tremendous advantages of using nanotechnology to 1) introduce CAR transgene cassettes into primary T cells, 2) stimulate T cell expansion and persistence, 3) improve T cell trafficking, 4) stimulate the intrinsic T cell activity, 5) reprogram the immunosuppressive cellular and vascular microenvironments, and 6) monitor the therapeutic efficacy of CAR-T cell therapy. Therefore, genetic and functional modifications promoted by nanotechnology enable the generation of robust CAR-T cell therapy and offer precision treatments against cancer.
Collapse
Affiliation(s)
- Ahmed M. E. Abdalla
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
- Department of BiochemistryCollege of Applied ScienceUniversity of BahriKhartoum1660/11111Sudan
| | - Lin Xiao
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| | - Yu Miao
- Department of Vascular SurgeryGeneral Hospital of Ningxia Medical UniversityYinchuan750004China
| | - Lixia Huang
- Hubei Key Laboratory of Purification and Application of Plant Anti‐Cancer Active IngredientsSchool of Chemistry and Life SciencesHubei University of EducationWuhan430205China
| | - Gendeal M. Fadlallah
- Department of Chemistry and BiologyFaculty of EducationUniversity of GeziraWad‐Medani2667Sudan
| | - Mario Gauthier
- Department of ChemistryUniversity of WaterlooWaterlooN2L 3G1Canada
| | - Chenxi Ouyang
- Department of Vascular SurgeryFuwai HospitalNational Center for Cardiovascular DiseaseChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijing100037China
| | - Guang Yang
- Department of Biomedical EngineeringCollege of Life Science and TechnologyHuazhong University of Science and TechnologyWuhan430074China
| |
Collapse
|
162
|
Namuduri M, Brentjens RJ. Enhancing CAR T cell efficacy: the next step toward a clinical revolution? Expert Rev Hematol 2020; 13:533-543. [PMID: 32267181 DOI: 10.1080/17474086.2020.1753501] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Introduction: The field of immunotherapy has witnessed considerable progress over the last two decades. Beginning with the ability to conceptualize CAR T cell therapy as immunotherapeutic approach, to effortlessly genetically modifying T cells, we have now reached the stage of mass production for clinical needs, all within less than quarter of a century.Areas covered: CAR T cell therapy has been tremendously successful in acute leukemia patients, specifically even in relapsed/refractory disease states. However, similar success is yet to be realized in other malignancies. This review article covers the challenges encountered with the current CD19-targeted CARs, as well as specific obstacles faced by adoptive therapy in solid tumors. It also discusses various strategies to counteract these problems.Expert opinion: CD19-directed trials in the past decade have exposed vulnerabilities in the current CAR T cell design, particularly concerning safety aspects, antigen escape, and T cell persistence. Building on these lessons and factoring in the unique challenges associated with immunotherapy in solid tumors will help generate CARs designed for future trials. Also, research related to the production of allogeneic CAR T cell products will boost the patient reach of this unique technology and possibly reduce financial burden.
Collapse
Affiliation(s)
- Manjusha Namuduri
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Renier J Brentjens
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
163
|
Weber EW, Maus MV, Mackall CL. The Emerging Landscape of Immune Cell Therapies. Cell 2020; 181:46-62. [PMID: 32243795 PMCID: PMC8900215 DOI: 10.1016/j.cell.2020.03.001] [Citation(s) in RCA: 250] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/21/2022]
Abstract
Cell therapies present an entirely new paradigm in drug development. Within this class, immune cell therapies are among the most advanced, having already demonstrated definitive evidence of clinical benefits in cancer and infectious disease. Numerous features distinguish these "living therapies" from traditional medicines, including their ability to expand and contract in proportion to need and to mediate therapeutic benefits for months or years following a single application. Continued advances in fundamental immunology, genetic engineering, gene editing, and synthetic biology exponentially expand opportunities to enhance the sophistication of immune cell therapies, increasing potency and safety and broadening their potential for treatment of disease. This perspective will summarize the current status of immune cell therapies for cancer, infectious disease, and autoimmunity, and discuss advances in cellular engineering to overcome barriers to progress.
Collapse
Affiliation(s)
- Evan W Weber
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
164
|
Pampusch MS, Haran KP, Hart GT, Rakasz EG, Rendahl AK, Berger EA, Connick E, Skinner PJ. Rapid Transduction and Expansion of Transduced T Cells with Maintenance of Central Memory Populations. Mol Ther Methods Clin Dev 2020; 16:1-10. [PMID: 31673565 PMCID: PMC6816036 DOI: 10.1016/j.omtm.2019.09.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 09/12/2019] [Indexed: 02/02/2023]
Abstract
Chimeric antigen receptor (CAR)-T cells show great promise in treating cancers and viral infections. However, most protocols developed to expand T cells require relatively long periods of time in culture, potentially leading to progression toward populations of terminally differentiated effector memory cells. Here, we describe in detail a 9-day protocol for CAR gene transduction and expansion of primary rhesus macaque peripheral blood mononuclear cells (PBMCs). Cells produced and expanded with this method show high levels of viability, high levels of co-expression of two transduced genes, retention of the central memory phenotype, and sufficient quantity for immunotherapeutic infusion of 1-2 × 108 cells/kg in a 10 kg rhesus macaque. This 9-day protocol may be broadly used for CAR-T cell and other T cell immunotherapy approaches to decrease culture time and increase maintenance of central memory populations.
Collapse
Affiliation(s)
- Mary S. Pampusch
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Kumudhini Preethi Haran
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Geoffrey T. Hart
- Division of Infectious Disease and International Medicine, Department of Medicine, Center for Immunology, University of Minnesota, Minneapolis, MN 55455, USA
| | - Eva G. Rakasz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI 53715, USA
| | - Aaron K. Rendahl
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| | - Edward A. Berger
- Laboratory of Viral Diseases, NIAID, NIH, Bethesda, MD 20814, USA
| | - Elizabeth Connick
- Division of Infectious Diseases, University of Arizona, Tucson, AZ 85724, USA
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN 55108, USA
| |
Collapse
|
165
|
Quintás-Cardama A. What CAR Will Win the CD19 Race? Mol Cancer Ther 2020; 18:498-506. [PMID: 30824581 DOI: 10.1158/1535-7163.mct-18-1070] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/27/2018] [Accepted: 01/15/2019] [Indexed: 11/16/2022]
Abstract
Adoptive transfer of T cells engineered with synthetic receptors is emerging as a new pillar in the treatment of cancer. The adoptive cell therapy furthest along in clinical development is the engineering of T cells to express chimeric antigen receptors (CAR) against the CD19 antigen. Several platforms have shown remarkable activity in patients with relapsed or refractory B-cell malignancies. In 2017, the FDA approved the first CAR T cell products tisagenlecleucel (Kymriah, Novartis) and axicabtagene ciloleucel (Yescarta, Gilead), and others are expected to follow shortly. Despite their activity, CAR T cell approaches have limitations that will need to be addressed, including excessive toxicity, relapses mediated via antigen escape, difficulties overcoming the suppressive tumor microenvironment, high manufacturing costs and retail prices, and patient access, among others. The CAR T cell product that better addresses those challenges will obtain a critical competitive advantage.
Collapse
|
166
|
Qi J, Ding C, Jiang X, Gao Y. Advances in Developing CAR T-Cell Therapy for HIV Cure. Front Immunol 2020; 11:361. [PMID: 32210965 PMCID: PMC7076163 DOI: 10.3389/fimmu.2020.00361] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 02/14/2020] [Indexed: 02/05/2023] Open
Abstract
Acquired immune deficiency syndrome (AIDS), which is caused by HIV infection, is an epidemic disease that has killed millions of people in the last several decades. Although combination antiretroviral therapy (cART) has enabled tremendous progress in suppressing HIV replication, it fails to eliminate HIV latently infected cells, and infected individuals remain HIV positive for life. Lifelong antiretroviral therapy is required to maintain control of virus replication, which may result in significant problems, including long-term toxicity, high cost, and stigma. Therefore, novel therapeutic strategies are urgently needed to eliminate the viral reservoir in the host for HIV cure. In this review, we compare several potential strategies regarding HIV cure and focus on how we might utilize chimeric antigen receptor-modified T cells (CAR T) as a therapy to cure HIV infection.
Collapse
Affiliation(s)
- Jinxin Qi
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
| | - Chengchao Ding
- The First Affiliated Hospital, Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| | - Xian Jiang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Gao
- Department of Microbiology and Immunology, The University of Western Ontario, London, ON, Canada
- The First Affiliated Hospital, Department of Life Science and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
167
|
Liu X, Wen J, Yi H, Hou X, Yin Y, Ye G, Wu X, Jiang X. Split chimeric antigen receptor-modified T cells targeting glypican-3 suppress hepatocellular carcinoma growth with reduced cytokine release. Ther Adv Med Oncol 2020; 12:1758835920910347. [PMID: 32215059 PMCID: PMC7065297 DOI: 10.1177/1758835920910347] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 01/30/2020] [Indexed: 12/25/2022] Open
Abstract
Background: Human glypican-3 (hGPC3) is a protein highly expressed in hepatocellular carcinoma (HCC) but limited in normal tissues, making it an ideal target for immunotherapy. The adoptive transfer of hGPC3-specific chimeric antigen receptor T (CAR-T) cells for HCC treatment has been conducted in clinical trials. Due to the rigid construction, conventional CAR-T cells have some intrinsic limitations, like uncontrollable overactivation and inducing severe cytokine release syndrome. Methods: We redesigned the hGPC3-specific CAR by splitting the traditional CAR into two parts. By using coculturing assays and a xenograft mouse model, the in vitro and in vivo cytotoxicity and cytokine release of the split anti-hGPC3 CAR-T cells were evaluated against various HCC cell lines and compared with conventional CAR-T cells. Results: In vitro data demonstrated that split anti-hGPC3 CAR-T cells could recognize and lyse hGPC3+ HepG2 and Huh7 cells in a dose-dependent manner. Impressively, split anti-hGPC3 CAR-T cells produced and released a significantly lower amount of proinflammatory cytokines, including IFN-γ, TNF-α, IL-6, and GM-CSF, than conventional CAR-T cells. When injected into immunodeficient mice inoculated subcutaneously with HepG2 cells, our split anti-hGPC3 CAR-T cells could suppress HCC tumor growth, but released significantly lower levels of cytokines than conventional CAR-T cells. Conclusions: We describe here for the first time the use of split anti-hGPC3 CAR-T cells to treat HCC; split anti-hGPC3 CAR-T cells could suppress tumor growth and reduce cytokine release, and represent a more versatile and safer alternative to conventional CAR-T cells treatment.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianyun Wen
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Honglei Yi
- Department of Orthopedics, General Hospital of Southern Theater Command, Guangzhou, China
| | - Xiaorui Hou
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yue Yin
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Guofu Ye
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xuedong Wu
- Department of Pediatrics, Nanfang Hospital, Southern Medical University, Tonghe Road, Guangzhou, 510515, China
| | - Xiaotao Jiang
- Department of Immunology, School of Basic Medical Sciences, Southern Medical University, Shatai Road, Guangzhou, 510515, China
| |
Collapse
|
168
|
Dash PK, Kevadiya BD, Su H, Banoub MG, Gendelman HE. Pathways towards human immunodeficiency virus elimination. EBioMedicine 2020; 53:102667. [PMID: 32114397 PMCID: PMC7047153 DOI: 10.1016/j.ebiom.2020.102667] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/17/2022] Open
Abstract
Antiretroviral therapy (ART) suppresses human immunodeficiency virus (HIV) infection. Research seeking to transform viral suppression into elimination has generated novel immune, chemical and molecular antiviral agents. However, none, to date, have excised latent integrated proviral DNA or removed infected cells from infected persons. These efforts included, but are not limited to, broadly neutralizing antibodies, "shock" and "kill" latency-reversing agents, innate immune regulators, and sequential long-acting antiretroviral nanoformulated prodrugs and CRISPR-Cas9 gene editing. While, the latter, enabled the complete excision of latent HIV-1 from the host genome success was so far limited. We contend that improvements in antiretroviral delivery, potency, agent specificity, or combinatorial therapies can provide a pathway towards complete HIV elimination.
Collapse
Affiliation(s)
- Prasanta K Dash
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Bhavesh D Kevadiya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Hang Su
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Mary G Banoub
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Howard E Gendelman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
169
|
Cong W, Shi Y, Qi Y, Wu J, Gong L, He M. Viral approaches to study the mammalian brain: Lineage tracing, circuit dissection and therapeutic applications. J Neurosci Methods 2020; 335:108629. [PMID: 32045571 DOI: 10.1016/j.jneumeth.2020.108629] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 02/01/2020] [Accepted: 02/04/2020] [Indexed: 02/09/2023]
Abstract
Viral vectors are widely used to study the development, function and pathology of neural circuits in the mammalian brain. Their flexible payloads with customizable choices of tool genes allow versatile applications ranging from lineage tracing, circuit mapping and functional interrogation, to translational and therapeutic applications. Different applications have distinct technological requirements, therefore, often utilize different types of virus. This review introduces the most commonly used viruses for these applications and some recent advances in improving the resolution and throughput of lineage tracing, the efficacy and selectivity of circuit tracing and the specificity of cell type targeting.
Collapse
Affiliation(s)
- Wei Cong
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yun Shi
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanqing Qi
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinyun Wu
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ling Gong
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Miao He
- Department of Neurology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Zhongshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
170
|
Braendstrup P, Levine BL, Ruella M. The long road to the first FDA-approved gene therapy: chimeric antigen receptor T cells targeting CD19. Cytotherapy 2020; 22:57-69. [PMID: 32014447 PMCID: PMC7036015 DOI: 10.1016/j.jcyt.2019.12.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/27/2019] [Accepted: 12/01/2019] [Indexed: 12/11/2022]
Abstract
Thirty years after initial publications of the concept of a chimeric antigen receptor (CAR), the U.S. Food and Drug Administration (FDA) approved the first anti-CD19 CAR T-cell therapy. Unlike other immunotherapies, such as immune checkpoint inhibitors and bispecific antibodies, CAR T cells are unique as they are "living drugs," that is, gene-edited killer cells that can recognize and kill cancer. During these 30 years of development, the CAR construct, T-cell manufacturing process, and clinical patient management have gone through rounds of failures and successes that drove continuous improvement. Tisagenlecleucel was the first gene therapy to receive approval from the FDA for any indication. The initial approval was for relapsed or refractory (r/r) pediatric and young-adult B-cell acute lymphoblastic leukemia in August 2017 and in May 2018 for adult r/r diffuse large B-cell lymphoma. Here we review the preclinical and clinical development of what began as CART19 at the University of Pennsylvania and later developed into tisagenlecleucel.
Collapse
Affiliation(s)
- Peter Braendstrup
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Hematology, Herlev University Hospital, Denmark; Department of Hematology, Zealand University Hospital Roskilde, Denmark
| | - Bruce L Levine
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Marco Ruella
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA; Department of Medicine, Division of Hematology and Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
171
|
Chulanetra M, Morchang A, Sayour E, Eldjerou L, Milner R, Lagmay J, Cascio M, Stover B, Slayton W, Chaicumpa W, Yenchitsomanus PT, Chang LJ. GD2 chimeric antigen receptor modified T cells in synergy with sub-toxic level of doxorubicin targeting osteosarcomas. Am J Cancer Res 2020; 10:674-687. [PMID: 32195035 PMCID: PMC7061749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 01/17/2020] [Indexed: 06/10/2023] Open
Abstract
Since the prognosis for children with high-risk osteosarcoma (OS) remains suboptimal despite intensive multi-modality therapies, there is a clear and urgent need for the development of targeted therapeutics against these refractory malignancies. Chimeric antigen receptor (CAR) modified T cells can meet this need by utilizing the immune system's potent cytotoxic mechanisms against tumor specific antigen targets with exquisite specificity. Since OS highly expresses the GD2 antigen, a viable immunotherapeutic target, we sought to assess if CAR modified T cells targeting GD2 could induce cytotoxicity against OS tumor cells. We demonstrated that the GD2 CAR modified T cells were highly efficacious for inducing OS tumor cell death. Interestingly, the OS cells were induced to up-regulate expression of PD-L1 upon interaction with GD2 CAR modified T cells, and the specific interaction induced CAR T cells to overexpress the exhaustion marker PD-1 along with increased CAR T cell apoptosis. To further potentiate CAR T cell killing activity against OS, we demonstrated that suboptimal chemotherapeutic treatment with doxorubicin can synergize with CAR T cells to effectively kill OS tumor cells.
Collapse
Affiliation(s)
- Monrat Chulanetra
- Department of Molecular Genetics and Microbiology, University of FloridaGainesville, FL 32610, USA
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine, Siriraj Hospital, Mahidol UniversityBangkok, Thailand
| | - Atthapan Morchang
- Department of Molecular Genetics and Microbiology, University of FloridaGainesville, FL 32610, USA
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Siriraj Medical Research Center, Faculty of Medicine, Siriraj Hospital, Mahidol UniversityBangkok, Thailand
| | - Elias Sayour
- Division of Pediatric Hematology-Oncology, Department of PediatricsSichuan, China
- Brain Tumor Immunotherapy Program, Department of NeurosurgerySichuan, China
| | - Lamis Eldjerou
- Division of Pediatric Hematology-Oncology, Department of PediatricsSichuan, China
| | - Rowan Milner
- Department of Small Animal Clinical Sciences, University of FloridaSichuan, China
| | - Joanne Lagmay
- Division of Pediatric Hematology-Oncology, Department of PediatricsSichuan, China
| | - Matt Cascio
- Division of Pediatric Hematology-Oncology, Department of PediatricsSichuan, China
| | - Brian Stover
- Division of Pediatric Hematology-Oncology, Department of PediatricsSichuan, China
| | - William Slayton
- Division of Pediatric Hematology-Oncology, Department of PediatricsSichuan, China
| | - Wanpen Chaicumpa
- Center of Research Excellence on Therapeutic Proteins and Antibody Engineering, Department of Parasitology, Faculty of Medicine, Siriraj Hospital, Mahidol UniversityBangkok, Thailand
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy, Siriraj Medical Research Center, Faculty of Medicine, Siriraj Hospital, Mahidol UniversityBangkok, Thailand
| | - Lung-Ji Chang
- Department of Molecular Genetics and Microbiology, University of FloridaGainesville, FL 32610, USA
- University of Electronic Science and Technology of ChinaSichuan, China
- Shenzhen Geno-Immune Medical InstituteShenzhen, China
| |
Collapse
|
172
|
Chandran SS, Klebanoff CA. T cell receptor-based cancer immunotherapy: Emerging efficacy and pathways of resistance. Immunol Rev 2020; 290:127-147. [PMID: 31355495 PMCID: PMC7027847 DOI: 10.1111/imr.12772] [Citation(s) in RCA: 194] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 05/09/2019] [Indexed: 12/13/2022]
Abstract
Adoptive cell transfer (ACT) using chimeric antigen receptor (CAR)-modified T cells can induce durable remissions in patients with refractory B-lymphoid cancers. By contrast, results applying CAR-modified T cells to solid malignancies have been comparatively modest. Alternative strategies to redirect T cell specificity and cytolytic function are therefore necessary if ACT is to serve a greater role in human cancer treatments. T cell receptors (TCRs) are antigen recognition structures physiologically expressed by all T cells that have complementary, and in some cases superior, properties to CARs. Unlike CARs, TCRs confer recognition to epitopes derived from proteins residing within any subcellular compartment, including the membrane, cytoplasm and nucleus. This enables TCRs to detect a broad universe of targets, such as neoantigens, cancer germline antigens, and viral oncoproteins. Moreover, because TCRs have evolved to efficiently detect and amplify antigenic signals, these receptors respond to epitope densities many fold smaller than required for CAR-signaling. Herein, we summarize recent clinical data demonstrating that TCR-based immunotherapies can mediate regression of solid malignancies, including immune-checkpoint inhibitor refractory cancers. These trials simultaneously highlight emerging mechanisms of TCR resistance. We conclude by discussing how TCR-based immunotherapies can achieve broader dissemination through innovations in cell manufacturing and non-viral genome integration techniques.
Collapse
Affiliation(s)
- Smita S Chandran
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Parker Institute for Cancer Immunotherapy, New York, NY
| | - Christopher A Klebanoff
- Center for Cell Engineering and Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.,Parker Institute for Cancer Immunotherapy, New York, NY.,Weill Cornell Medical College, New York, NY
| |
Collapse
|
173
|
Tang F, Lu Y, Ge Y, Shang J, Zhu X. Infusion of chimeric antigen receptor T cells against dual targets of CD19 and B-cell maturation antigen for the treatment of refractory multiple myeloma. J Int Med Res 2020; 48:300060519893496. [PMID: 31939323 PMCID: PMC7114292 DOI: 10.1177/0300060519893496] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 11/15/2019] [Indexed: 12/21/2022] Open
Abstract
Objective To investigate the safety and efficacy of chimeric antigen receptor T (CAR-T) cell infusion in patients with refractory multiple myeloma (MM). Methods Sixteen patients diagnosed with refractory MM were included in this study. Patients received initial infusions of T-derived CD19/B-cell maturation antigen (BCMA) CAR-T cells with 100% CD19, followed by second infusions with 40% BCMA and third infusions with 60% BCMA. The total doses were 0.5–1 × 107/kg CD19 and 1.2 − 6.2 × 107/kg BCMA. Patients were monitored after infusion. Levels of interleukin (IL)-2, IL-6, IL-10, tumor necrosis factor-α, and C-reactive protein were determined by enzyme-linked immunosorbent assay. Results Cytokine release syndrome (CRS) was observed in all 16 patients. Thirteen patients with CRS stage II−IV had persistent hyperthermia from 5−14 days after infusion, while most patients developed hyperthermia from 1 day after infusion and their temperatures returned to normal within 2−10 days. Levels of all factors were significantly elevated 2 days after infusion, peaked at 5 days, and then gradually decreased to normal levels. All inflammatory factors showed normal levels by 10 days after infusion. Conclusion Body temperature and levels of inflammatory factors all increased dramatically after infusion of CD19/BCMA CAR-T cells, but recovered to normal levels after appropriate treatment and nursing.
Collapse
Affiliation(s)
- Fang Tang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yin Lu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongqin Ge
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jingjing Shang
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiaming Zhu
- Department of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
174
|
Abreu TR, Fonseca NA, Gonçalves N, Moreira JN. Current challenges and emerging opportunities of CAR-T cell therapies. J Control Release 2019; 319:246-261. [PMID: 31899268 DOI: 10.1016/j.jconrel.2019.12.047] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/27/2019] [Accepted: 12/28/2019] [Indexed: 12/27/2022]
Abstract
Infusion of chimeric antigen receptor (CAR)-genetically modified T cells (CAR-T cells) have led to remarkable clinical responses and cancer remission in patients suffering from relapsed or refractory B-cell malignancies. This is a new form of adoptive T cell therapy (ACT), whereby the artificial CAR enables the redirection of T cells endogenous antitumor activity towards a predefined tumor-associated antigen, leading to the elimination of a specific tumor. The early success in blood cancers has prompted the US Food and Drug Administration (FDA) to approve the first CAR-T cell therapies for the treatment of CD19-positive leukemias and lymphomas in 2017. Despite the emergence of CAR-T cells as one of the latest breakthroughs of cancer immunotherapies, their wider application has been hampered by specific life-threatening toxicities, and a substantial lack of efficacy in the treatment of solid tumors, owing to the strong immunosuppressive tumor microenvironment and the paucity of reliable tumor-specific targets. Herein, besides providing an overview of the emerging CAR-technologies and current clinical applications, the major hurdles of CAR-T cell therapies will be discussed, namely treatment-related life-threatening toxicities and the obstacles posed by the immunosupressive tumor-microenvironment of solid tumors, as well as the next-generation strategies currently designed to simultaneously improve safety and efficacy of CAR-T cell therapies in vivo.
Collapse
Affiliation(s)
- Teresa R Abreu
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Nuno A Fonseca
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; TREAT U, SA, Parque Industrial de Taveiro, Lote 44, 3045-508 Coimbra, Portugal.
| | - Nélio Gonçalves
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal.
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, 3004-504 Coimbra, Portugal; FFUC - Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal.
| |
Collapse
|
175
|
Chimeric antigen receptor -T cell therapy: Applications and challenges in treatment of allergy and asthma. Biomed Pharmacother 2019; 123:109685. [PMID: 31862474 DOI: 10.1016/j.biopha.2019.109685] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 10/28/2019] [Accepted: 11/16/2019] [Indexed: 01/10/2023] Open
Abstract
Despite the current advancements, cancer treatment approaches have limitations restricting their cure rate. Immunotherapy techniques are among novel and promising cancer therapeutic approaches. Therapeutic antibodies and adoptive cell therapy (ACT) are the main branches of immunotherapy. T lymphocytes and genetically engineered cells are among important cells which can be used in ACT. This review has focused on recent advances in engineered cell-based immunotherapy based on T lymphocytes with chimeric antigen receptors (CARs). CARs are recombinant receptors expressing T cell signaling domains with or without co-stimulatory molecules. CAR-T cells are expanded ex vivo and re-infused to patients in order to improve their therapeutic efficacy. Nowadays, the beneficial function of CAR-T cell therapy has been indicated in various diseases including hematological malignancies, solid tumors, autoimmune diseases, and allergic diseases such as asthma. Furthermore, antigen-specific T regulatory cells (Tregs) and gene-edited T cells seem to be beneficial in controlling inflammation in allergic asthma. In fact, dysregulated function of Tregs is responsible for dominance of T helper 2 immune response and progression of allergic asthma. CAR-Treg cells can also be designed and reproduced using iTreg population to manage asthma. In addition, universal CAR-T cells can be modified to selectively target multiple antigens. The fourth generation CAR-T cells (i.e. TRUCK cells) represent novel strategies to cure asthma and allergic diseases as well. Despite the advantages of CAR-T cells, their applications can be associated with some unwanted reactions such as cytokine storm, anaphylaxis, neurotoxicity, etc. For clinical application, there is a need to prevent and manage these complications by optimizing ACT protocols.
Collapse
|
176
|
Straetemans T, Janssen A, Jansen K, Doorn R, Aarts T, van Muyden ADD, Simonis M, Bergboer J, de Witte M, Sebestyen Z, Kuball J. TEG001 Insert Integrity from Vector Producer Cells until Medicinal Product. Mol Ther 2019; 28:561-571. [PMID: 31882320 DOI: 10.1016/j.ymthe.2019.11.030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 11/19/2019] [Accepted: 11/26/2019] [Indexed: 12/20/2022] Open
Abstract
Despite extensive usage of gene therapy medicinal products (GTMPs) in clinical studies and recent approval of chimeric antigen receptor (CAR) T cell therapy, little information has been made available on the precise molecular characterization and possible variations in terms of insert integrity and vector copy numbers of different GTMPs during the complete production chain. Within this context, we characterize αβT cells engineered to express a defined γδT cell engineered to express a defined γδT receptor (TEG) currently used in a first-in-human clinical study (NTR6541). Utilizing targeted locus amplification in combination with next generation sequencing for the vector producer clone and TEG001 products, we report on five single-nucleotide variants and nine intact vector copies integrated in the producer clone. The vector copy number in TEG001 cells was on average a factor 0.72 (SD 0.11) below that of the producer cell clone. All nucleotide variants were transferred to TEG001 without having an effect on cellular proliferation during extensive in vitro culture. Based on an environmental risk assessment of the five nucleotide variants present in the non-coding viral region of the TEG001 insert, there was no altered environmental impact of TEG001 cells. We conclude that TEG001 cells do not have an increased risk for malignant transformation in vivo.
Collapse
Affiliation(s)
- Trudy Straetemans
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| | - Anke Janssen
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Koen Jansen
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Ruud Doorn
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Tineke Aarts
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Anna D D van Muyden
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | | | | | - Moniek de Witte
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Zsolt Sebestyen
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands
| | - Jurgen Kuball
- Department of Hematology, Center of Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
177
|
Gershovich PM, Karabelskii AV, Ulitin AB, Ivanov RA. The Role of Checkpoint Inhibitors and Cytokines in Adoptive Cell-Based Cancer Immunotherapy with Genetically Modified T Cells. BIOCHEMISTRY (MOSCOW) 2019; 84:695-710. [PMID: 31509722 DOI: 10.1134/s0006297919070022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
This review focuses on the structure and molecular action mechanisms of chimeric antigen receptors (CARs) and major aspects of the manufacturing and clinical application of products for the CAR-T (CAR-modified T lymphocyte) therapy of hematological and solid tumors with special emphasis on the strategies for combined use of CAR-T therapy with immuno-oncological monoclonal antibodies (checkpoint inhibitors) and cytokines to boost survival, persistence, and antitumor efficacy of CAR-T therapy. The review also summarizes preclinical and clinical data on the additive effects of the combined use of CAR-T therapy with interleukins and monoclonal antibodies targeting immune checkpoints.
Collapse
Affiliation(s)
- P M Gershovich
- CJSC Biocad, St. Petersburg, 198515, Russia. .,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A V Karabelskii
- CJSC Biocad, St. Petersburg, 198515, Russia.,St. Petersburg State Chemical Pharmaceutical Academy, St. Petersburg, 197376, Russia
| | - A B Ulitin
- CJSC Biocad, St. Petersburg, 198515, Russia
| | - R A Ivanov
- CJSC Biocad, St. Petersburg, 198515, Russia
| |
Collapse
|
178
|
Abstract
: Given the challenges of life-long adherence to suppressive HIV antiretroviral therapy (ART) and possibilities of comorbidities, such as HIV association neurocognitive disorder, HIV remission and eradication are desirable goals for people living with HIV. In some individuals, there is evidence that HIV persists and replicates in the CNS, impacting the success of HIV remission interventions. This article addresses the role of HIV CNS latency on HIV eradication, examines the effects of early ART, latency-modifying agents, antibody-based and T-cell enhancing therapies on the CNS as well as ART interruption in remission studies. We propose the integration of CNS monitoring into such studies in order to clarify the short-term and long-term neurological safety of experimental agents and treatment interruption, and to better characterize their effects on HIV CNS persistence.
Collapse
|
179
|
Zhao Z, Zheng L, Chen W, Weng W, Song J, Ji J. Delivery strategies of cancer immunotherapy: recent advances and future perspectives. J Hematol Oncol 2019; 12:126. [PMID: 31779642 PMCID: PMC6883629 DOI: 10.1186/s13045-019-0817-3] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022] Open
Abstract
Immunotherapy has become an emerging strategy for the treatment of cancer. Immunotherapeutic drugs have been increasing for clinical treatment. Despite significant advances in immunotherapy, the clinical application of immunotherapy for cancer patients has some challenges associated with safety and efficacy, including autoimmune reactions, cytokine release syndrome, and vascular leak syndrome. Novel strategies, particularly improved delivery strategies, including nanoparticles, scaffolds, and hydrogels, are able to effectively target tumors and/or immune cells of interest, increase the accumulation of immunotherapies within the lesion, and reduce off-target effects. Here, we briefly describe five major types of cancer immunotherapy, including their clinical status, strengths, and weaknesses. Then, we introduce novel delivery strategies, such as nanoparticle-based delivery of immunotherapy, implantable scaffolds, injectable biomaterials for immunotherapy, and matrix-binding molecular conjugates, which can improve the efficacy and safety of immunotherapies. Also, the limitations of novel delivery strategies and challenges of clinical translation are discussed.
Collapse
Affiliation(s)
- Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Liyun Zheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Weiqian Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Wei Weng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Jingjing Song
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University /The Central Hospital of Zhejiang Lishui, Lishui, 323000, China. .,Department of Radiology, Affiliated Lishui Hospital of Zhejiang University/the Fifth Affiliated Hospital of Wenzhou Medical University/The Central Hospital of Zhejiang Lishui, Lishui, 323000, China. .,Department of Interventional Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Affiliated Lishui Hospital of Zhejiang University, The Central Hospital of Zhejiang Lishui, Lishui, 323000, China.
| |
Collapse
|
180
|
Seif M, Einsele H, Löffler J. CAR T Cells Beyond Cancer: Hope for Immunomodulatory Therapy of Infectious Diseases. Front Immunol 2019; 10:2711. [PMID: 31824500 PMCID: PMC6881243 DOI: 10.3389/fimmu.2019.02711] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022] Open
Abstract
Infectious diseases are still a significant cause of morbidity and mortality worldwide. Despite the progress in drug development, the occurrence of microbial resistance is still a significant concern. Alternative therapeutic strategies are required for non-responding or relapsing patients. Chimeric antigen receptor (CAR) T cells has revolutionized cancer immunotherapy, providing a potential therapeutic option for patients who are unresponsive to standard treatments. Recently two CAR T cell therapies, Yescarta® (Kite Pharma/Gilead) and Kymriah® (Novartis) were approved by the FDA for the treatments of certain types of non-Hodgkin lymphoma and B-cell precursor acute lymphoblastic leukemia, respectively. The success of adoptive CAR T cell therapy for cancer has inspired researchers to develop CARs for the treatment of infectious diseases. Here, we review the main achievements in CAR T cell therapy targeting viral infections, including Human Immunodeficiency Virus, Hepatitis C Virus, Hepatitis B Virus, Human Cytomegalovirus, and opportunistic fungal infections such as invasive aspergillosis.
Collapse
Affiliation(s)
| | | | - Jürgen Löffler
- Department of Internal Medicine II, University Hospital Wuerzburg, Würzburg, Germany
| |
Collapse
|
181
|
Vagnozzi RJ, Johansen AKZ, Molkentin JD. CARdiac Immunotherapy: T Cells Engineered to Treat the Fibrotic Heart. Mol Ther 2019; 27:1869-1871. [PMID: 31585799 PMCID: PMC6838878 DOI: 10.1016/j.ymthe.2019.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Ronald J Vagnozzi
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Anne Katrine Z Johansen
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
182
|
Lesch S, Benmebarek MR, Cadilha BL, Stoiber S, Subklewe M, Endres S, Kobold S. Determinants of response and resistance to CAR T cell therapy. Semin Cancer Biol 2019; 65:80-90. [PMID: 31705998 DOI: 10.1016/j.semcancer.2019.11.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/28/2019] [Accepted: 11/03/2019] [Indexed: 12/27/2022]
Abstract
The remarkable success of chimeric antigen receptor (CAR)-engineered T cells in pre-B cell acute lymphoblastic leukemia (ALL) and B cell lymphoma led to the approval of anti-CD19 CAR T cells as the first ever CAR T cell therapy in 2017. However, with the number of CAR T cell-treated patients increasing, observations of tumor escape and resistance to CAR T cell therapy with disease relapse are demonstrating the current limitations of this therapeutic modality. Mechanisms hampering CAR T cell efficiency include limited T cell persistence, caused for example by T cell exhaustion and activation-induced cell death (AICD), as well as therapy-related toxicity. Furthermore, the physical properties, antigen heterogeneity and immunosuppressive capacities of solid tumors have prevented the success of CAR T cells in these entities. Herein we review current obstacles of CAR T cell therapy and propose strategies in order to overcome these hurdles and expand CAR T cell therapy to a broader range of cancer patients.
Collapse
Affiliation(s)
- Stefanie Lesch
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Mohamed-Reda Benmebarek
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Bruno L Cadilha
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Stoiber
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany
| | - Marion Subklewe
- German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany; Department of Medicine III, Klinikum der Universität München, LMU Munich, Germany
| | - Stefan Endres
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, LMU Munich, Germany; German Center for Translational Cancer Research (DKTK), partner site Munich, Munich, Germany.
| |
Collapse
|
183
|
Herzig E, Kim KC, Packard TA, Vardi N, Schwarzer R, Gramatica A, Deeks SG, Williams SR, Landgraf K, Killeen N, Martin DW, Weinberger LS, Greene WC. Attacking Latent HIV with convertibleCAR-T Cells, a Highly Adaptable Killing Platform. Cell 2019; 179:880-894.e10. [PMID: 31668804 DOI: 10.1016/j.cell.2019.10.002] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/19/2019] [Accepted: 10/01/2019] [Indexed: 12/20/2022]
Abstract
Current approaches to reducing the latent HIV reservoir entail first reactivating virus-containing cells to become visible to the immune system. A critical second step is killing these cells to reduce reservoir size. Endogenous cytotoxic T-lymphocytes (CTLs) may not be adequate because of cellular exhaustion and the evolution of CTL-resistant viruses. We have designed a universal CAR-T cell platform based on CTLs engineered to bind a variety of broadly neutralizing anti-HIV antibodies. We show that this platform, convertibleCAR-T cells, effectively kills HIV-infected, but not uninfected, CD4 T cells from blood, tonsil, or spleen and only when armed with anti-HIV antibodies. convertibleCAR-T cells also kill within 48 h more than half of the inducible reservoir found in blood of HIV-infected individuals on antiretroviral therapy. The modularity of convertibleCAR-T cell system, which allows multiplexing with several anti-HIV antibodies yielding greater breadth and control, makes it a promising tool for attacking the latent HIV reservoir.
Collapse
Affiliation(s)
- Eytan Herzig
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kaman Chan Kim
- Xyphos Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Thomas A Packard
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Noam Vardi
- Gladstone Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Biochemistry and Biophysics and Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Roland Schwarzer
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Andrea Gramatica
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | | | - Kyle Landgraf
- Xyphos Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Nigel Killeen
- Xyphos Biosciences, Inc., South San Francisco, CA 94080, USA
| | - David W Martin
- Xyphos Biosciences, Inc., South San Francisco, CA 94080, USA
| | - Leor S Weinberger
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Gladstone Center for Cell Circuitry, Gladstone Institutes, San Francisco, CA 94158, USA; Departments of Biochemistry and Biophysics and Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Warner C Greene
- Gladstone Center for HIV Cure Research, Gladstone Institute of Virology and Immunology, San Francisco, CA 94158, USA; Departments of Medicine and Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
184
|
Hao L, Li T, Chang LJ, Chen X. Adoptive Immunotherapy for B-cell Malignancies Using CD19- Targeted Chimeric Antigen Receptor T-Cells: A Systematic Review of Efficacy and Safety. Curr Med Chem 2019; 26:3068-3079. [PMID: 28762313 DOI: 10.2174/0929867324666170801101842] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 06/15/2017] [Accepted: 07/25/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Adoptive infusion of chimeric antigen receptor transduced T- cells (CAR-T) is a powerful tool of immunotherapy for hematological malignancies, as evidenced by recently published and unpublished clinical results. OBJECTIVE In this report, we performed a meta-analysis to evaluate the efficacy and side effects of CAR-T on refractory and/or relapsed B-cell malignancies, including leukemia and lymphoma. METHODS Clinical studies investigating efficacy and safety of CAR-T in acute and chronic lymphocytic leukemia and lymphoma were identified by searching PubMed and EMBASE. Outcomes of efficacy subjected to analysis were the rates of complete remission (CR) and partial remission (PR). The safety parameters were the prevalence of adverse effects including fever, hypotension, and acute renal failure. Meta analyses were performed using R software. Weighted hazard ratio (HR) with 95% confidence intervals was calculated for each outcome. Fixed or random-effects models were employed depending on the heterogeneity across the included studies. RESULTS Nineteen published clinical studies with a total of 391 patients were included for the meta-analysis. The pooled rate of complete remission was 55% (95% CI 41%-69%); the pooled rate of partial remission was 25% (95% CI: 19%-33%). The prevalence of fever was 62% (95% CI: 41%-79%), the hypotension was 22% (95% CI: 15%-31%), and the acute renal failure was 24% (95% CI: 16%-34%). All adverse effects were manageable and no death was reported due to toxicity. CONCLUSION CD19-targeted CAR-T is an effective modality in treating refractory B-cell malignancies including leukemia and lymphoma. However, there is still a need to develop strategies to improve the safety in its clinical use.
Collapse
Affiliation(s)
- Lu Hao
- Shenzhen Geno-Immune Medical Institute, Shenzhen 518057, China.,Institute of Cancer Stem Cells, Dalian Medical University, Dalian 116044, China
| | - Tongtong Li
- Clinical Medicine Program, Nanchang University Medical College, Nanchang 330006, China.,Department of Obstetrics and Gynecology, Anfu People's Hospital, Jiangxi Province 343200, China
| | - Lung-Ji Chang
- Shenzhen Geno-Immune Medical Institute, Shenzhen 518057, China.,Gene Therapy Center, University of Florida College of Medicine, Gainesville, FL 32610, United States
| | - Xiaochuan Chen
- Shenzhen Geno-Immune Medical Institute, Shenzhen 518057, China.,Department of Oriental Medicine, New York College of Health Professions, New York, NY 10016, United States
| |
Collapse
|
185
|
Abstract
While impressive clinical responses have been observed using chimeric antigen receptor (CAR) T cells targeting CD19+ hematologic malignancies, limited clinical benefit has been observed using CAR T cells for a variety of solid tumors. Results of clinical studies have highlighted several obstacles which CAR T cells face in the context of solid tumors, including insufficient homing to tumor sites, lack of expansion and persistence, encountering a highly immunosuppressive tumor microenvironment, and heterogeneous antigen expression. In this review, we review clinical outcomes and discuss strategies to improve the antitumor activity of CAR T cells for solid tumors.
Collapse
|
186
|
Kim GB, Hege K, Riley JL. CAR Talk: How Cancer-Specific CAR T Cells Can Instruct How to Build CAR T Cells to Cure HIV. Front Immunol 2019; 10:2310. [PMID: 31611880 PMCID: PMC6776630 DOI: 10.3389/fimmu.2019.02310] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/12/2019] [Indexed: 01/21/2023] Open
Abstract
Re-directing T cells via chimeric antigen receptors (CARs) was first tested in HIV-infected individuals with limited success, but these pioneering studies laid the groundwork for the clinically successful CD19 CARs that were recently FDA approved. Now there is great interest in revisiting the concept of using CAR-expressing T cells as part of a strategy to cure HIV. Many lessons have been learned on how to best engineer T cells to cure cancer, but not all of these lessons apply when developing CARs to treat and cure HIV. This mini review will focus on how early CAR T cell studies in HIV paved the way for cancer CAR T cell therapy and how progress in cancer CAR therapy has and will continue to be instructive for the development of HIV CAR T cell therapy. Additionally, the unique challenges that must be overcome to develop a successful HIV CAR T cell therapy will be highlighted.
Collapse
Affiliation(s)
- Gloria B. Kim
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kristen Hege
- Celgene Corporation, San Francisco, CA, United States
| | - James L. Riley
- Department of Microbiology, Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
187
|
Mylvaganam G, Yanez AG, Maus M, Walker BD. Toward T Cell-Mediated Control or Elimination of HIV Reservoirs: Lessons From Cancer Immunology. Front Immunol 2019; 10:2109. [PMID: 31552045 PMCID: PMC6746828 DOI: 10.3389/fimmu.2019.02109] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/21/2019] [Indexed: 12/16/2022] Open
Abstract
As the AIDS epidemic unfolded, the appearance of opportunistic infections in at-risk persons provided clues to the underlying problem: a dramatic defect in cell-mediated immunity associated with infection and depletion of CD4+ T lymphocytes. Moreover, the emergence of HIV-associated malignancies in these same individuals was a clear indication of the significant role effective cellular immunity plays in combating cancers. As research in the HIV field progressed, advances included the first demonstration of the role of PD-1 in human T cell exhaustion, and the development of gene-modified T cell therapies, including chimeric antigen receptor (CAR) T cells. In the intervening years, the oncology field has capitalized on these advances, effectively mobilizing the cellular immune response to achieve immune-mediated remission or cure of previously intractable cancers. Although similar therapeutic advances have not yet been achieved in the HIV field, spontaneous CD8+ T cell mediated remission or functional cure of HIV infection does occur in very small subset of individuals in the absence of anti-retroviral therapy (ART). This has many similarities to the CD8+ T cell mediated functional control or elimination of cancers, and indicates that immunotherapy for HIV is a rational goal. In HIV infection, one major barrier to successful immunotherapy is the small, persistent population of infected CD4+ T cells, the viral reservoir, which evades pharmacological and immune-mediated clearance, and is largely maintained in secondary lymphoid tissues at sites where CD8+ T cells have limited access and/or function. The reservoir-enriched lymphoid microenvironment bears a striking resemblance to the tumor microenvironment of many solid tumors–namely high levels of anti-inflammatory cytokines, expression of co-inhibitory receptors, and physical exclusion of immune effector cells. Here, we review the parallels between CD8+ T cell-mediated immune control of HIV and cancer, and how advances in cancer immunotherapy may provide insights to direct the development of effective HIV cure strategies. Specifically, understanding the impact of the tissue microenvironment on T cell function and development of CAR T cells and therapeutic vaccines deserve robust attention on the path toward a CD8+ T cell mediated cure of HIV infection.
Collapse
Affiliation(s)
- Geetha Mylvaganam
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Adrienne G Yanez
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Marcela Maus
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States.,MGH Cancer Center, Boston, MA, United States
| | - Bruce D Walker
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States.,Howard Hughes Medical Institute, Chevy Chase, MD, United States.,Institute for Medical Engineering and Sciences, MIT, Cambridge, MA, United States
| |
Collapse
|
188
|
ElMallah MK, Kalfopolous M, Flotte TR. GENE THERAPY. Cancer 2019. [DOI: 10.1002/9781119645214.ch28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
189
|
Abstract
Chimeric antigen receptors (CARs) have shown remarkable ability to re-direct T cells to target CD19-expressing tumours, resulting in remission rates of up to 90% in individuals with paediatric acute lymphoblastic lymphoma. Lessons learned from these clinical trials of adoptive T cell therapy for cancer, as well as investments made in manufacturing T cells at commercial scale, have inspired researchers to develop CARs for additional applications. Here, we explore the challenges and opportunities of using this technology to target infectious diseases such as with HIV and undesired immune responses such as autoimmunity and transplant rejection. Despite substantial obstacles, the potential of CAR T cells to enable cures for a wide array of disease settings could be transformational for the medical field.
Collapse
|
190
|
Liu X, Zheng J, Sun W, Zhao X, Li Y, Gong N, Wang Y, Ma X, Zhang T, Zhao LY, Hou Y, Wu Z, Du Y, Fan H, Tian J, Liang XJ. Ferrimagnetic Vortex Nanoring-Mediated Mild Magnetic Hyperthermia Imparts Potent Immunological Effect for Treating Cancer Metastasis. ACS NANO 2019; 13:8811-8825. [PMID: 31328922 DOI: 10.1021/acsnano.9b01979] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Cancer metastasis is a serious concern and a major reason for treatment failure. Herein, we have reported the development of an effective and safe nanotherapeutic strategy that can eradicate primary tumors, inhibit metastasizing to lung, and control the metastasis and growth of distant tumors. Briefly, ferrimagnetic vortex-domain iron oxide nanoring (FVIO)-mediated mild magnetic hyperthermia caused calreticulin (CRT) expression on the 4T1 breast cancer cells. The CRT expression transmitted an "eat-me" signal and promoted phagocytic uptake of cancer cells by the immune system to induce an efficient immunogenic cell death, further leading to the macrophage polarization. This mild thermotherapy promoted 88% increase of CD8+ cytotoxic T lymphocyte infiltration in distant tumors and triggered immunotherapy by effectively sensitizing tumors to the PD-L1 checkpoint blockade. The percentage of CD8+ cytotoxic T lymphocytes can be further increased from 55.4% to 64.5% after combining with PD-L1 blockade. Moreover, the combination treatment also inhibited the immunosuppressive response of the tumor, evidenced by significant down-regulation of myeloid-derived suppressor cells (MDSCs). Our results revealed that the FVIO-mediated mild magnetic hyperthermia can activate the host immune systems and efficiently cooperate with PD-L1 blockade to inhibit the potential metastatic spreading as well as the growth of distant tumors.
Collapse
Affiliation(s)
- Xiaoli Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
- The College of Life Sciences , Northwest University , Xi'an , Shaanxi 710069 , China
| | - Jianjun Zheng
- Department of Radiology, Hwa Mei Hospital , University of Chinese Academy of Sciences , Ningbo No.2 Hospital, Ningbo , Zhejiang 315010 , China
| | - Wei Sun
- Department of Radiology, Hwa Mei Hospital , University of Chinese Academy of Sciences , Ningbo No.2 Hospital, Ningbo , Zhejiang 315010 , China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Yao Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Ningqiang Gong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Yanyun Wang
- The College of Life Sciences , Northwest University , Xi'an , Shaanxi 710069 , China
| | - Xiaowei Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
| | - Tingbin Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , China
| | - Ling-Yun Zhao
- Key Laboratory of Advanced Materials, School of Material Science & Engineering , Tsinghua University , Beijing 100084 , China
| | - Yayi Hou
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical School , Nanjing University , Nanjing 210093 , China
- Jiangsu Key Laboratory of Molecular Medicine , Nanjing University , Nanjing , 210093 , China
| | - Zhibing Wu
- Department of Radiation Oncology , Zhejiang Hospital , Hangzhou , Zhejiang 310013 , China
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , China
| | - Haiming Fan
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of the Ministry of Education, College of Chemistry and Materials Science , Northwest University , Xi'an 710127 , China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, the State Key Laboratory of Management and Control for Complex Systems , Institute of Automation, Chinese Academy of Sciences , Beijing 100190 , China
| | - Xing-Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience , National Center for Nanoscience and Technology of China , No. 11, First North Road , Zhongguancun, Beijing 100190 , China
- University of Chinese Academy of Sciences , Beijing 100049 , China
| |
Collapse
|
191
|
Yu S, Yi M, Qin S, Wu K. Next generation chimeric antigen receptor T cells: safety strategies to overcome toxicity. Mol Cancer 2019; 18:125. [PMID: 31429760 PMCID: PMC6701025 DOI: 10.1186/s12943-019-1057-4] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 08/14/2019] [Indexed: 01/06/2023] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy is an emerging and effective cancer immunotherapy. Especially in hematological malignancies, CAR-T cells have achieved exciting results. Two Anti-CD19 CAR-T therapies have been approved for the treatment of CD19-positive leukemia or lymphoma. However, the application of CAR-T cells is obviously hampered by the adverse effects, such as cytokines release syndrome and on-target off-tumor toxicity. In some clinical trials, patients quitted the treatment of CAR-T cells due to life-threatening toxicity. Seeking to alleviate these toxicities or prevent the occurrence, researchers have developed a number of safety strategies of CAR-T cells, including suicide genes, synthetic Notch receptor, on-switch CAR, combinatorial target-antigen recognition, bispecific T cell engager and inhibitory CAR. This review summarized the preclinical studies and clinical trials of the safety strategies of CAR-T cells and their respective strengths and weaknesses.
Collapse
Affiliation(s)
- Shengnan Yu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Shuang Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
192
|
Koristka S, Ziller-Walter P, Bergmann R, Arndt C, Feldmann A, Kegler A, Cartellieri M, Ehninger A, Ehninger G, Bornhäuser M, Bachmann MP. Anti-CAR-engineered T cells for epitope-based elimination of autologous CAR T cells. Cancer Immunol Immunother 2019; 68:1401-1415. [PMID: 31414180 PMCID: PMC6768917 DOI: 10.1007/s00262-019-02376-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Accepted: 08/07/2019] [Indexed: 12/16/2022]
Abstract
Although CAR T-cell therapy has demonstrated tremendous clinical efficacy especially in hematological malignancies, severe treatment-associated toxicities still compromise the widespread application of this innovative technology. Therefore, developing novel approaches to abrogate CAR T-cell-mediated side effects is of great relevance. Several promising strategies pursue the selective antibody-based depletion of adoptively transferred T cells via elimination markers. However, given the limited half-life and tissue penetration, dependence on the patients’ immune system and on-target/off-side effects of proposed monoclonal antibodies, we sought to exploit αCAR-engineered T cells to efficiently eliminate CAR T cells. For comprehensive and specific recognition, a small peptide epitope (E-tag) was incorporated into the extracellular spacer region of CAR constructs. We provide first proof-of-concept for targeting this epitope by αE-tag CAR T cells, allowing an effective killing of autologous E-tagged CAR T cells both in vitro and in vivo whilst sparing cells lacking the E-tag. In addition to CAR T-cell cytotoxicity, the αE-tag-specific T cells can be empowered with cancer-fighting ability in case of relapse, hence, have versatile utility. Our proposed methodology can most probably be implemented in CAR T-cell therapies regardless of the targeted tumor antigen aiding in improving overall safety and survival control of highly potent gene-modified cells.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Autoantigens/immunology
- Cytotoxicity, Immunologic
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/immunology
- Genetic Engineering
- Humans
- Immunotherapy, Adoptive/methods
- Male
- Mice
- Neoplasm Recurrence, Local
- PC-3 Cells
- Peptide Fragments/genetics
- Prostatic Neoplasms/immunology
- Prostatic Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes, Cytotoxic/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Stefanie Koristka
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Pauline Ziller-Walter
- Tumor Immunology, University Cancer Center (UCC), 'Carl Gustav Carus' Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Ralf Bergmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Claudia Arndt
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Anja Feldmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Alexandra Kegler
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany
| | - Marc Cartellieri
- Cellex Patient Treatment GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Armin Ehninger
- GEMoaB Monoclonals GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Gerhard Ehninger
- Cellex Patient Treatment GmbH, Tatzberg 47, 01307, Dresden, Germany
- GEMoaB Monoclonals GmbH, Tatzberg 47, 01307, Dresden, Germany
| | - Martin Bornhäuser
- Medical Clinic and Policlinic I, University Hospital 'Carl Gustav Carus' Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany
| | - Michael P Bachmann
- Department of Radioimmunology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Bautzner Landstraße 400, 01328, Dresden, Germany.
- Tumor Immunology, University Cancer Center (UCC), 'Carl Gustav Carus' Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- National Center for Tumor Diseases (NCT), 'Carl Gustav Carus' Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
193
|
Han C, Kwon BS. Chimeric antigen receptor T-cell therapy for cancer: a basic research-oriented perspective. Immunotherapy 2019; 10:221-234. [PMID: 29370727 DOI: 10.2217/imt-2017-0133] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have outstanding therapeutic potential for treating blood cancers. The prospects for this technology have accelerated basic research, clinical translation and Big Pharma's investment in the field of T-cell therapeutics. This interest has led to the discovery of key factors that affect CAR T-cell efficacy and play pivotal roles in T-cell immunology. Herein, we introduce advances in adoptive immunotherapy and the birth of CAR T cells, and review CAR T-cell studies that focus on three important features: CAR constructs, target antigens and T-cell phenotypes. At last, we highlight novel strategies that overcome the tumor microenvironment and circumvent CAR T-cell side effects, and consider the future direction of CAR T-cell development.
Collapse
Affiliation(s)
- Chungyong Han
- Immunotherapeutics Branch, Division of Convergence Technology Research Institute, National Cancer Center, Goyang 10408, Korea
| | - Byoung S Kwon
- Immunotherapeutics Branch, Division of Convergence Technology Research Institute, National Cancer Center, Goyang 10408, Korea.,Eutilex Co., Ltd, Suite #1401, Daeryung Technotown 17, Gasan digital 1-ro 25, Geumcheon-gu, Seoul 08594, Korea.,Department of Medicine, Tulane University Health Sciences Center, New Orleans, LA 70118, USA
| |
Collapse
|
194
|
Abstract
As the HIV pandemic rapidly spread worldwide in the 1980s and 1990s, a new approach to treat cancer, genetic diseases, and infectious diseases was also emerging. Cell and gene therapy strategies are connected with human pathologies at a fundamental level, by delivering DNA and RNA molecules that could correct and/or ameliorate the underlying genetic factors of any illness. The history of HIV gene therapy is especially intriguing, in that the virus that was targeted was soon co-opted to become part of the targeting strategy. Today, HIV-based lentiviral vectors, along with many other gene delivery strategies, have been used to evaluate HIV cure approaches in cell culture, small and large animal models, and in patients. Here, we trace HIV cell and gene therapy from the earliest clinical trials, using genetically unmodified cell products from the patient or from matched donors, through current state-of-the-art strategies. These include engineering HIV-specific immunity in T-cells, gene editing approaches to render all blood cells in the body HIV-resistant, and most importantly, combination therapies that draw from both of these respective "offensive" and "defensive" approaches. It is widely agreed upon that combinatorial approaches are the most promising route to functional cure/remission of HIV infection. This chapter outlines cell and gene therapy strategies that are poised to play an essential role in eradicating HIV-infected cells in vivo.
Collapse
|
195
|
Abstract
PURPOSE OF REVIEW Combination antiretroviral therapy (ART) has enabled tremendous progress in suppressing HIV replication in infected patients. However, ART alone cannot eradicate HIV and its latent, persisting reservoirs. Novel approaches are needed to eradicate the virus or achieve functional cure in the absence of ART. RECENT FINDINGS Adoptive T-cell therapies were initially tested in HIV-infected individuals with limited efficiency. Benefiting from new and improved methodologies, an increasing array of CAR T-cell therapies has been successfully developed in the cancer immunotherapy field, demonstrating promising new avenues that could be applied to HIV. Numerous studies have characterized various HIV-specific CAR constructs, types of cytolytic effector cells, and CAR-expressing cells' trafficking to the reservoir compartments, warranting further in-vivo efforts. Notably, the ability of CAR cells to persist and function in low-antigen environments in vivo, that is, in ART-suppressed patients, remains unclear. SUMMARY Despite promising results in preclinical studies, only a handful of clinical trials have been initiated worldwide. Several obstacles remain prior to successful application of HIV-specific CAR T-cell therapies in patients. In this review, we survey the current state of the field, and address paths towards realizing the goal of an efficacious HIV CAR T-cell product.
Collapse
|
196
|
Traceless aptamer-mediated isolation of CD8 + T cells for chimeric antigen receptor T-cell therapy. Nat Biomed Eng 2019; 3:783-795. [PMID: 31209354 PMCID: PMC6783348 DOI: 10.1038/s41551-019-0411-6] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 05/02/2019] [Indexed: 12/29/2022]
Abstract
Chimeric antigen receptor T-cell therapies using defined product compositions require high-purity T-cell isolation systems that, unlike immunomagnetic positive enrichment, are inexpensive and leave no trace on the final cell product. Here, we show that DNA aptamers (generated with a modified cell-SELEX procedure to display low-nanomolar affinity for the T-cell marker CD8) enable the traceless isolation of pure CD8+ T cells at low cost and high yield. Captured CD8+ T cells are released label-free by complementary oligonucleotides that undergo toehold-mediated strand displacement with the aptamer. We also show that chimeric antigen receptor T cells manufactured from these cells are comparable to antibody-isolated chimeric antigen receptor T cells in proliferation, phenotype, effector function and antitumour activity in a mouse model of B-cell lymphoma. By employing multiple aptamers and the corresponding complementary oligonucleotides, aptamer-mediated cell selection could enable the fully synthetic, sequential and traceless isolation of desired lymphocyte subsets from a single system.
Collapse
|
197
|
Ghani K, Boivin-Welch M, Roy S, Dakiw-Piaceski A, Barbier M, Pope E, Germain L, Caruso M. Generation of High-Titer Self-Inactivated γ-Retroviral Vector Producer Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:90-99. [PMID: 31312667 PMCID: PMC6610700 DOI: 10.1016/j.omtm.2019.05.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 05/30/2019] [Indexed: 12/01/2022]
Abstract
The γ-retroviral vector is a gene delivery vehicle that is commonly used in gene therapy. Despite its efficacy, its strong enhancers contributed to malignant transformations in some hematopoietic stem cell (HSC) gene therapy trials. A safer version without viral enhancers (SIN) is available, but its production is cumbersome, as high titers can only be obtained in transient transfection. Our aim was to develop a system that could easily generate high-titer SIN vectors from stable producer cells. The use of the cytomegalovirus enhancer-promoter sequence to generate the full-length genomic RNA combined to sequences that decrease transcriptional readthrough (WPRE and strong polyadenylation sequences) led to 6 × 106 infectious units (IU)/mL of a SIN GFP vector in transient transfection. The incorporation of a blasticidin selection cassette to the retroviral plasmid allowed the generation of stable clones in the 293Vec packaging cells that release 2 × 107 IU/mL and 1.4 × 107 IU/mL of a SIN GFP and a SIN PIGA vector, respectively. A titer of 1.8 × 106 IU/mL was obtained with a SIN vector containing the long 8.9-kb COL7A1 cDNA. Thus, an efficient process was established for the generation of stable 293Vec-derived retrovirus producer cells that release high-titer SIN vectors.
Collapse
Affiliation(s)
- Karim Ghani
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| | - Michael Boivin-Welch
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada.,CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Sylvie Roy
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| | - Angela Dakiw-Piaceski
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Martin Barbier
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Elena Pope
- Section of Dermatology, The Hospital for Sick Children and University of Toronto, Toronto, ON, Canada
| | - Lucie Germain
- CHU de Québec-Université Laval Research Center (Regenerative Medicine Division) and Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, and Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, G1J 1Z4, Canada
| | - Manuel Caruso
- CHU de Québec-Université Laval Research Center (Oncology Division), Université Laval Cancer Research Center, and Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC G1R 2J6, Canada
| |
Collapse
|
198
|
Uludag H, Ubeda A, Ansari A. At the Intersection of Biomaterials and Gene Therapy: Progress in Non-viral Delivery of Nucleic Acids. Front Bioeng Biotechnol 2019; 7:131. [PMID: 31214586 PMCID: PMC6558074 DOI: 10.3389/fbioe.2019.00131] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Biomaterials play a critical role in technologies intended to deliver therapeutic agents in clinical settings. Recent explosion of our understanding of how cells utilize nucleic acids has garnered excitement to develop a range of older (e.g., antisense oligonucleotides, plasmid DNA and transposons) and emerging (e.g., short interfering RNA, messenger RNA and non-coding RNAs) nucleic acid agents for therapy of a wide range of diseases. This review will summarize biomaterials-centered advances to undertake effective utilization of nucleic acids for therapeutic purposes. We first review various types of nucleic acids and their unique abilities to deliver a range of clinical outcomes. Using recent advances in T-cell based therapy as a case in point, we summarize various possibilities for utilizing biomaterials to make an impact in this exciting therapeutic intervention technology, with the belief that this modality will serve as a therapeutic paradigm for other types of cellular therapies in the near future. We subsequently focus on contributions of biomaterials in emerging nucleic acid technologies, specifically focusing on the design of intelligent nanoparticles, deployment of mRNA as an alternative to plasmid DNA, long-acting (integrating) expression systems, and in vitro/in vivo expansion of engineered T-cells. We articulate the role of biomaterials in these emerging nucleic acid technologies in order to enhance the clinical impact of nucleic acids in the near future.
Collapse
Affiliation(s)
- Hasan Uludag
- Department of Chemical and Materinals Engineering, University of Alberta, Edmonton, AB, Canada
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Anyeld Ubeda
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Aysha Ansari
- Department of Chemical and Materinals Engineering, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
199
|
Bailey SR, Maus MV. Gene editing for immune cell therapies. Nat Biotechnol 2019; 37:1425-1434. [DOI: 10.1038/s41587-019-0137-8] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Accepted: 04/22/2019] [Indexed: 02/06/2023]
|
200
|
Abstract
PURPOSE OF REVIEW A therapy that might cure HIV is a very important goal for the 30-40 million people living with HIV. Chimeric antigen receptor T cells have recently had remarkable success against certain leukemias, and there are reasons to believe they could be successful for HIV. This manuscript summarizes the published research on HIV CAR T cells and reviews the current anti-HIV chimeric antigen receptor strategies. RECENT FINDINGS Research on anti-HIV chimeric antigen receptor T cells has been going on for at least the last 25 years. First- and second-generation anti-HIV chimeric antigen receptors have been developed. First-generation anti-HIV chimeric antigen receptors were studied in clinical trials more than 15 years ago, but did not have meaningful clinical efficacy. There are some reasons to be optimistic about second-generation anti-HIV chimeric antigen receptor T cells, but they have not yet been tested in vivo.
Collapse
Affiliation(s)
- Thor A Wagner
- Seattle Children's Research Institute, 1900 Ninth Ave, 8th Floor, Seattle, WA, 98101, USA. .,University of Washington, 1959 NE Pacific St., Box 356320, Seattle, WA, 98195-6320, USA.
| |
Collapse
|