1
|
Antoney J, Kainrath S, Dubowsky JG, Ahmed FH, Kang SW, Mackie ERR, Granado GB, Soares da Costa TP, Jackson CJ, Janovjak H. A F 420-dependent single domain chemogenetic tool for protein de-dimerization. J Mol Biol 2025:169184. [PMID: 40324743 DOI: 10.1016/j.jmb.2025.169184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/16/2025] [Accepted: 04/29/2025] [Indexed: 05/07/2025]
Abstract
Protein-protein interactions (PPIs) mediate many fundamental cellular processes. Control of PPIs through optically or chemically responsive protein domains has had a profound impact on basic research and some clinical applications. Most chemogenetic methods induce the association, i.e., dimerization or oligomerization, of target proteins, whilst the few available dissociation approaches either break large oligomeric protein clusters or heteromeric complexes. Here, we have exploited the controlled dissociation of a homodimeric oxidoreductase from mycobacteria (MSMEG_2027) by its native cofactor, F420, which is not present in mammals, as a bioorthogonal monomerization switch. Using X-ray crystallography, we found that in the absence of F420, MSMEG_2027 forms a unique domain-swapped dimer that occludes the cofactor binding site. Rearrangement of the N-terminal helix upon F420 binding results in the dissolution of the dimer. We then showed that MSMEG_2027 can be fused to proteins of interest in human cells and applied it as a tool to induce and release MAPK/ERK signalling downstream of a chimeric fibroblast growth factor receptor 1 (FGFR1) tyrosine kinase. This F420-dependent chemogenetic de-homodimerization tool is stoichiometric and based on a single domain and thus represents a novel mechanism to investigate protein complexes in situ.
Collapse
Affiliation(s)
- James Antoney
- Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601 ACT, Australia; ARC Centre of Excellence in Synthetic Biology, Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601 ACT, Australia; ARC Centre of Excellence in Synthetic Biology, Centre for Agriculture and the Bioeconomy, School of Biology and Environmental Science, Queensland University of Technology, 2 George Street, Brisbane 4000 QLD, Australia
| | - Stephanie Kainrath
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 15 Innovation Walk, Clayton 3800 Melbourne, VIC, Australia; European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 15 Innovation Walk, Clayton 3800 Melbourne, VIC, Australia; Institute of Science and Technology Austria (IST Austria), Am Campus 1, 3400 Klosterneuburg, Austria; Proxygen GmbH, Siemensstrasse 89, 1210 Vienna, Austria
| | - Joshua G Dubowsky
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, 1 Flinders Drive, Bedford Park 5042 Adelaide, SA, Australia
| | - F Hafna Ahmed
- Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601 ACT, Australia; CSIRO Environment, Clunies Ross Street, Canberra 2601, ACT, Australia
| | - Suk Woo Kang
- Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601 ACT, Australia; Center for Natural Product Efficacy Optimization, Korea Institute of Science and Technology (KIST), 679, Saimdang-ro, Gangneung 25451, Republic of Korea
| | - Emily R R Mackie
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road, Bundoora 3086 Melbourne, VIC, Australia; School of Agriculture, Food and Wine, Waite Research Institute, The University of Adelaide, Hartley Grove, Urrbrae 5064 Adelaide, SA, Australia
| | - Gustavo Bracho Granado
- Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, 1 Flinders Drive, Bedford Park 5042 Adelaide, SA, Australia
| | - Tatiana P Soares da Costa
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Plenty Road, Bundoora 3086 Melbourne, VIC, Australia; School of Agriculture, Food and Wine, Waite Research Institute, The University of Adelaide, Hartley Grove, Urrbrae 5064 Adelaide, SA, Australia
| | - Colin J Jackson
- Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601 ACT, Australia; ARC Centre of Excellence in Synthetic Biology, Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601 ACT, Australia; ARC Centre of Excellence for Innovations in Peptide & Protein Science, Research School of Chemistry, Australian National University, 137 Sullivans Creek Road, Canberra 2601, ACT, Australia.
| | - Harald Janovjak
- Australian Regenerative Medicine Institute (ARMI), Faculty of Medicine, Nursing and Health Sciences, Monash University, 15 Innovation Walk, Clayton 3800 Melbourne, VIC, Australia; European Molecular Biology Laboratory Australia (EMBL Australia), Monash University, 15 Innovation Walk, Clayton 3800 Melbourne, VIC, Australia; Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, 1 Flinders Drive, Bedford Park 5042 Adelaide, SA, Australia.
| |
Collapse
|
2
|
Mulvey A, Trueb L, Coukos G, Arber C. Novel strategies to manage CAR-T cell toxicity. Nat Rev Drug Discov 2025:10.1038/s41573-024-01100-5. [PMID: 39901030 DOI: 10.1038/s41573-024-01100-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 02/05/2025]
Abstract
The immune-related adverse events associated with chimeric antigen receptor (CAR)-T cell therapy result in substantial morbidity as well as considerable cost to the health-care system, and can limit the use of these treatments. Current therapeutic strategies to manage immune-related adverse events include interleukin-6 receptor (IL-6R) blockade and corticosteroids. However, because these interventions do not always address the side effects, nor prevent progression to higher grades of adverse events, new approaches are needed. A deeper understanding of the cell types involved, and their associated signalling pathways, cellular metabolism and differentiation states, should provide the basis for alternative strategies. To preserve treatment efficacy, cytokine-mediated toxicity needs to be uncoupled from CAR-T cell function, expansion, long-term persistence and memory formation. This may be achieved by targeting CAR or independent cytokine signalling axes transiently, and through novel T cell engineering strategies, such as low-affinity CAR-T cells, reversible on-off switches and versatile adaptor systems. We summarize the current management of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, and review T cell- and myeloid cell-intrinsic druggable targets and cellular engineering strategies to develop safer CAR-T cells.
Collapse
Affiliation(s)
- Arthur Mulvey
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Lionel Trueb
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland.
- Departments of Oncology UNIL-CHUV and Laboratory Medicine and Pathology, Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
3
|
Palazzo L, Pieri V, Berzero G, Filippi M. CAR-T Cells for the Treatment of Central Nervous System Tumours: Known and Emerging Neurotoxicities. Brain Sci 2024; 14:1220. [PMID: 39766419 PMCID: PMC11727498 DOI: 10.3390/brainsci14121220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/24/2024] [Accepted: 11/26/2024] [Indexed: 01/15/2025] Open
Abstract
The advent of chimeric antigen receptor (CAR)-T cells has recently changed the prognosis of relapsing/refractory diffuse large B-cell lymphomas, showing response rates as high as 60 to 80%. Common toxicities reported in the pivotal clinical trials include the cytokine release syndrome (CRS) and the Immune effector Cell-Associated Neurotoxicity Syndrome (ICANS), a stereotyped encephalopathy related to myeloid cell activation and blood-brain barrier dysfunction, presenting with a distinctive cascade of dysgraphia, aphasia, disorientation, attention deficits, vigilance impairment, motor symptoms, seizures, and diffuse brain oedema. The tremendous oncological efficacy of CAR-T cells observed in systemic B-cell malignancies is leading to their growing use in patients with primary or secondary central nervous system (CNS) lymphomas and in patients with solid tumours, including several CNS cancers. Early studies conducted in adult and paediatric patients with solid CNS tumours reported a distinct profile of neurotoxicity referred to as Tumour inflammation-associated neurotoxicity (TIAN), corresponding to local inflammation at the tumour site manifesting with focal neurological deficits or mechanical complications (e.g., obstructive hydrocephalus). The present review summarises available data on the efficacy and safety of CAR-T cells for solid and haematological CNS malignancies, emphasising known and emerging phenotypes, ongoing challenges, and future perspectives.
Collapse
Affiliation(s)
- Leonardo Palazzo
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Valentina Pieri
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Giulia Berzero
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Massimo Filippi
- Neurology Unit, IRCCS Ospedale San Raffaele, 20132 Milan, Italy; (L.P.); (V.P.); (M.F.)
- Faculty of Medicine, Vita-Salute San Raffaele University, 20132 Milan, Italy
- Neurorehabilitation Unit, Neurophysiology Unit, Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| |
Collapse
|
4
|
Colonne CK, Kimble EL, Turtle CJ. Evolving strategies to overcome barriers in CAR-T cell therapy for acute myeloid leukemia. Expert Rev Hematol 2024; 17:797-818. [PMID: 39439295 DOI: 10.1080/17474086.2024.2420614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex and heterogeneous disease characterized by an aggressive clinical course and limited efficacious treatment options in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy is an investigational treatment strategy for R/R AML that has shown some promise. However, obstacles to successful CAR-T cell immunotherapy for AML remain. AREAS COVERED In analyses of clinical trials of CAR-T cell therapy for R/R AML, complete responses without measurable residual disease have been reported, but the durability of those responses remains unclear. Significant barriers to successful CAR-T cell therapy in AML include the scarcity of suitable tumor-target antigens (TTA), inherent T cell functional deficits, and the immunoinhibitory and hostile tumor microenvironment (TME). This review will focus on these barriers to successful CAR-T cell therapy in AML, and discuss scientific advancements and evolving strategies to overcome them. EXPERT OPINION Achieving durable remissions in R/R AML will likely require a multifaceted approach that integrates advancements in TTA selection, enhancement of the intrinsic quality of CAR-T cells, and development of strategies to overcome inhibitory mechanisms in the AML TME.
Collapse
Affiliation(s)
- Chanukya K Colonne
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erik L Kimble
- Translational Science and Therapeutic Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Cameron J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
5
|
Głowacki P, Tręda C, Rieske P. Regulation of CAR transgene expression to design semiautonomous CAR-T. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200833. [PMID: 39184876 PMCID: PMC11344471 DOI: 10.1016/j.omton.2024.200833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Effective transgene expression is critical for genetically engineered cell therapy. Therefore, one of CAR-T cell therapy's critical areas of interest, both in registered products and next-generation approaches is the expression of transgenes. It turns out that various constitutive promoters used in clinical products may influence CAR-T cell antitumor effectiveness and impact the manufacturing process. Furthermore, next-generation CAR-T starts to install remotely controlled inducible promoters or even autonomous expression systems, opening new ways of priming, boosting, and increasing the safety of CAR-T. In this article, a wide range of constitutive and inducible promoters has been grouped and structured, making it possible to compare their pros and cons as well as clinical usage. Finally, logic gates based on Synthetic Notch have been elaborated, demonstrating the coupling of desired external signals with genetically engineered cellular responses.
Collapse
Affiliation(s)
- Paweł Głowacki
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 Street, 90-752 Lodz, Poland
| | - Cezary Tręda
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 Street, 90-752 Lodz, Poland
- Department of Research and Development Personather Ltd, Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Chair of Medical Biology, Medical University of Lodz, Zeligowskiego 7/9 Street, 90-752 Lodz, Poland
- Department of Research and Development Personather Ltd, Inwestycyjna 7, 95-050 Konstantynow Lodzki, Poland
| |
Collapse
|
6
|
Shao W, Yao Y, Yang L, Li X, Ge T, Zheng Y, Zhu Q, Ge S, Gu X, Jia R, Song X, Zhuang A. Novel insights into TCR-T cell therapy in solid neoplasms: optimizing adoptive immunotherapy. Exp Hematol Oncol 2024; 13:37. [PMID: 38570883 PMCID: PMC10988985 DOI: 10.1186/s40164-024-00504-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/21/2024] [Indexed: 04/05/2024] Open
Abstract
Adoptive immunotherapy in the T cell landscape exhibits efficacy in cancer treatment. Over the past few decades, genetically modified T cells, particularly chimeric antigen receptor T cells, have enabled remarkable strides in the treatment of hematological malignancies. Besides, extensive exploration of multiple antigens for the treatment of solid tumors has led to clinical interest in the potential of T cells expressing the engineered T cell receptor (TCR). TCR-T cells possess the capacity to recognize intracellular antigen families and maintain the intrinsic properties of TCRs in terms of affinity to target epitopes and signal transduction. Recent research has provided critical insight into their capability and therapeutic targets for multiple refractory solid tumors, but also exposes some challenges for durable efficacy. In this review, we describe the screening and identification of available tumor antigens, and the acquisition and optimization of TCRs for TCR-T cell therapy. Furthermore, we summarize the complete flow from laboratory to clinical applications of TCR-T cells. Last, we emerge future prospects for improving therapeutic efficacy in cancer world with combination therapies or TCR-T derived products. In conclusion, this review depicts our current understanding of TCR-T cell therapy in solid neoplasms, and provides new perspectives for expanding its clinical applications and improving therapeutic efficacy.
Collapse
Affiliation(s)
- Weihuan Shao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yiran Yao
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Ludi Yang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiaoran Li
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Tongxin Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Yue Zheng
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Qiuyi Zhu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Shengfang Ge
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Xiang Gu
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China
| | - Renbing Jia
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Xin Song
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| | - Ai Zhuang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai JiaoTong University School of Medicine, 639 Zhi Zao Ju Road, Shanghai Ninth People's Hospital, Shanghai, 200011, People's Republic of China.
- Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, 200011, People's Republic of China.
| |
Collapse
|
7
|
Jha R, Kinna A, Hotblack A, Bughda R, Bulek A, Gannon I, Ilca T, Allen C, Lamb K, Dolor A, Scott I, Parekh F, Sillibourne J, Cordoba S, Onuoha S, Thomas S, Ferrari M, Pule M. Designer Small-Molecule Control System Based on Minocycline-Induced Disruption of Protein-Protein Interaction. ACS Chem Biol 2024; 19:308-324. [PMID: 38243811 PMCID: PMC10877577 DOI: 10.1021/acschembio.3c00521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 01/01/2024] [Accepted: 01/04/2024] [Indexed: 01/22/2024]
Abstract
A versatile, safe, and effective small-molecule control system is highly desirable for clinical cell therapy applications. Therefore, we developed a two-component small-molecule control system based on the disruption of protein-protein interactions using minocycline, an FDA-approved antibiotic with wide availability, excellent biodistribution, and low toxicity. The system comprises an anti-minocycline single-domain antibody (sdAb) and a minocycline-displaceable cyclic peptide. Here, we show how this versatile system can be applied to OFF-switch split CAR systems (MinoCAR) and universal CAR adaptors (MinoUniCAR) with reversible, transient, and dose-dependent suppression; to a tunable T cell activation module based on MyD88/CD40 signaling; to a controllable cellular payload secretion system based on IL12 KDEL retention; and as a cell/cell inducible junction. This work represents an important step forward in the development of a remote-controlled system to precisely control the timing, intensity, and safety of therapeutic interventions.
Collapse
Affiliation(s)
- Ram Jha
- Autolus
Therapeutics, London W12 7FP, U.K.
- Research
Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, U.K.
| | | | - Alastair Hotblack
- Research
Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, U.K.
| | | | - Anna Bulek
- Autolus
Therapeutics, London W12 7FP, U.K.
| | | | - Tudor Ilca
- Autolus
Therapeutics, London W12 7FP, U.K.
| | | | | | | | - Ian Scott
- Autolus
Therapeutics, London W12 7FP, U.K.
| | | | | | | | | | | | | | - Martin Pule
- Autolus
Therapeutics, London W12 7FP, U.K.
- Research
Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, U.K.
| |
Collapse
|
8
|
Angelos MG, Patel RP, Ruella M, Barta SK. Progress and Pitfalls of Chimeric Antigen Receptor T Cell Immunotherapy against T Cell Malignancies. Transplant Cell Ther 2024; 30:171-186. [PMID: 37866783 PMCID: PMC10873040 DOI: 10.1016/j.jtct.2023.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/05/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) immunotherapy has revolutionized the treatment of relapsed and refractory B cell-derived hematologic malignancies. Currently, there are 6 Food and Drug Administration-approved commercial CAR-T products that target antigens exclusively expressed on malignant B cells or plasma cells. However, concurrent advancement for patients with rarer and more aggressive T cell-derived hematologic malignancies have not yet been achieved. CAR-T immunotherapies are uniquely limited by challenges related to CAR-T product manufacturing and intrinsic tumor biology. In this review tailored for practicing clinician-scientists, we discuss the major barriers of CAR-T implementation against T cell-derived neoplasms and highlight specific scientific advancements poised to circumvent these obstacles. We summarize salient early-stage clinical trials implementing novel CAR-T immunotherapies specifically for patients with relapsed and/or refractory T cell neoplasms. Finally, we highlight novel manufacturing and treatment strategies that are poised to have a meaningful future clinical impact.
Collapse
Affiliation(s)
- Mathew G Angelos
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ruchi P Patel
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marco Ruella
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Stefan K Barta
- Division of Hematology and Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
9
|
Neeser A, Ramasubramanian R, Wang C, Ma L. Engineering enhanced chimeric antigen receptor-T cell therapy for solid tumors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 19:100385. [PMID: 37483659 PMCID: PMC10362352 DOI: 10.1016/j.iotech.2023.100385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
The early clinical success and subsequent US Food and Drug Administration approval of chimeric antigen receptor (CAR)-T cell therapy for leukemia and lymphoma affirm that engineered T cells can be a powerful treatment for hematologic malignancies. Yet this success has not been replicated in solid tumors. Numerous challenges emerged from clinical experience and well-controlled preclinical animal models must be met to enable safe and efficacious CAR-T cell therapy in solid tumors. Here, we review recent advances in bioengineering strategies developed to enhance CAR-T cell therapy in solid tumors, focusing on targeted single-gene perturbation, genetic circuits design, cytokine engineering, and interactive biomaterials. These bioengineering approaches present a unique set of tools that synergize with CAR-T cells to overcome obstacles in solid tumors and achieve robust and long-lasting therapeutic efficacy.
Collapse
Affiliation(s)
- A. Neeser
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia
| | - R. Ramasubramanian
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia
| | - C. Wang
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia
| | - L. Ma
- The Raymond G. Perelman Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
10
|
Zhu I, Piraner DI, Roybal KT. Synthesizing a Smarter CAR T Cell: Advanced Engineering of T-cell Immunotherapies. Cancer Immunol Res 2023; 11:1030-1043. [PMID: 37429007 PMCID: PMC10527511 DOI: 10.1158/2326-6066.cir-22-0962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 03/15/2023] [Accepted: 06/02/2023] [Indexed: 07/12/2023]
Abstract
The immune system includes an array of specialized cells that keep us healthy by responding to pathogenic cues. Investigations into the mechanisms behind immune cell behavior have led to the development of powerful immunotherapies, including chimeric-antigen receptor (CAR) T cells. Although CAR T cells have demonstrated efficacy in treating blood cancers, issues regarding their safety and potency have hindered the use of immunotherapies in a wider spectrum of diseases. Efforts to integrate developments in synthetic biology into immunotherapy have led to several advancements with the potential to expand the range of treatable diseases, fine-tune the desired immune response, and improve therapeutic cell potency. Here, we examine current synthetic biology advances that aim to improve on existing technologies and discuss the promise of the next generation of engineered immune cell therapies.
Collapse
Affiliation(s)
- Iowis Zhu
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
- These authors contributed equally
| | - Dan I. Piraner
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
- These authors contributed equally
| | - Kole T. Roybal
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA 94143, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, San Francisco, CA 94158, USA 8Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
- Gladstone UCSF Institute for Genetic Immunology, San Francisco, CA 94107, USA
- UCSF Cell Design Institute, San Francisco, CA 94158, USA
| |
Collapse
|
11
|
Xiong X, Yu Y, Jin X, Xie D, Sun R, Lu W, Wei Y, Guo R, Zhao M. Functional Validation of the RQR8 Suicide /Marker Gene in CD19 CAR-T Cells and CLL1CAR-T Cells. Ann Hematol 2023; 102:1523-1535. [PMID: 37086278 DOI: 10.1007/s00277-023-05227-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 04/10/2023] [Indexed: 04/23/2023]
Abstract
Chimeric antigen receptor T cell therapy (CAR-T) is a novel treatment that has produced unprecedented clinical effects in patients with hematological malignancies. Acute adverse events often occur following adoptive immunotherapy. Therefore, a suicide gene is helpful, which is a genetically encoded mechanism that allows selective destruction of adoptively transferred T cells in the face of unacceptable toxicity. RQR8 is a gene that integrates CD34 and CD20 epitopes. In our study, we incorporated the suicide gene RQR8 into CAR-T cells, so it enabled rituximab to eliminate vector/transgene-expressing T cells via antibody-dependent cell-mediated cytotoxicity and complement dependent cytotoxicity. In this work, we explored the functionality of RQR8 CAR-T cells in vitro and in vivo. We believe that RQR8 as a safety switch will make CAR-T cell therapy safer and less costly.
Collapse
Affiliation(s)
- Xia Xiong
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
- Army Medical Center of PLA, Chong Qing, China
| | - Yibing Yu
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
- The First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Xin Jin
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Danni Xie
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Rui Sun
- Nankai University School of Medicine, Tianjin, China
| | - Wenyi Lu
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China
| | - Yunxiong Wei
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Ruiting Guo
- The First Central Clinical College of Tianjin Medical University, Tianjin, China
| | - Mingfeng Zhao
- The First Central Clinical College of Tianjin Medical University, Tianjin, China.
- Department of Hematology, Tianjin First Central Hospital, Tianjin, China.
- Nankai University School of Medicine, Tianjin, China.
| |
Collapse
|
12
|
Celichowski P, Turi M, Charvátová S, Radhakrishnan D, Feizi N, Chyra Z, Šimíček M, Jelínek T, Bago JR, Hájek R, Hrdinka M. Tuning CARs: recent advances in modulating chimeric antigen receptor (CAR) T cell activity for improved safety, efficacy, and flexibility. J Transl Med 2023; 21:197. [PMID: 36922828 PMCID: PMC10015723 DOI: 10.1186/s12967-023-04041-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Cancer immunotherapies utilizing genetically engineered T cells have emerged as powerful personalized therapeutic agents showing dramatic preclinical and clinical results, particularly in hematological malignancies. Ectopically expressed chimeric antigen receptors (CARs) reprogram immune cells to target and eliminate cancer. However, CAR T cell therapy's success depends on the balance between effective anti-tumor activity and minimizing harmful side effects. To improve CAR T cell therapy outcomes and mitigate associated toxicities, scientists from different fields are cooperating in developing next-generation products using the latest molecular cell biology and synthetic biology tools and technologies. The immunotherapy field is rapidly evolving, with new approaches and strategies being reported at a fast pace. This comprehensive literature review aims to provide an up-to-date overview of the latest developments in controlling CAR T cell activity for improved safety, efficacy, and flexibility.
Collapse
Affiliation(s)
- Piotr Celichowski
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Marcello Turi
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Dhwani Radhakrishnan
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Neda Feizi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Zuzana Chyra
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Michal Šimíček
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Tomáš Jelínek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Juli Rodriguez Bago
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Matouš Hrdinka
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.
| |
Collapse
|
13
|
CAR T-Cell Immunotherapy Treating T-ALL: Challenges and Opportunities. Vaccines (Basel) 2023; 11:vaccines11010165. [PMID: 36680011 PMCID: PMC9861718 DOI: 10.3390/vaccines11010165] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/28/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023] Open
Abstract
T-cell acute lymphoblastic leukemia (T-ALL), a form of T-cell malignancy, is a typically aggressive hematological malignancy with high rates of disease relapse and a poor prognosis. Current guidelines do not recommend any specific treatments for these patients, and only allogeneic stem cell transplant, which is associated with potential risks and toxicities, is a curative therapy. Recent clinical trials showed that immunotherapies, including monoclonal antibodies, checkpoint inhibitors, and CAR T therapies, are successful in treating hematologic malignancies. CAR T cells, which specifically target the B-cell surface antigen CD19, have demonstrated remarkable efficacy in the treatment of B-cell acute leukemia, and some progress has been made in the treatment of other hematologic malignancies. However, the development of CAR T-cell immunotherapy targeting T-cell malignancies appears more challenging due to the potential risks of fratricide, T-cell aplasia, immunosuppression, and product contamination. In this review, we discuss the current status of and challenges related to CAR T-cell immunotherapy for T-ALL and review potential strategies to overcome these limitations.
Collapse
|
14
|
Overcoming on-target, off-tumour toxicity of CAR T cell therapy for solid tumours. Nat Rev Clin Oncol 2023; 20:49-62. [PMID: 36418477 DOI: 10.1038/s41571-022-00704-3] [Citation(s) in RCA: 199] [Impact Index Per Article: 99.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2022] [Indexed: 11/25/2022]
Abstract
Therapies with genetically modified T cells that express chimeric antigen receptors (CARs) specific for CD19 or B cell maturation antigen (BCMA) are approved to treat certain B cell malignancies. However, translating these successes into treatments for patients with solid tumours presents various challenges, including the risk of clinically serious on-target, off-tumour toxicity (OTOT) owing to CAR T cell-mediated cytotoxicity against non-malignant tissues expressing the target antigen. Indeed, severe OTOT has been observed in various CAR T cell clinical trials involving patients with solid tumours, highlighting the importance of establishing strategies to predict, mitigate and control the onset of this effect. In this Review, we summarize current clinical evidence of OTOT with CAR T cells in the treatment of solid tumours and discuss the utility of preclinical mouse models in predicting clinical OTOT. We then describe novel strategies being developed to improve the specificity of CAR T cells in solid tumours, particularly the role of affinity tuning of target binders, logic circuits and synthetic biology. Furthermore, we highlight control strategies that can be used to mitigate clinical OTOT following cell infusion such as regulating or eliminating CAR T cell activity, exogenous control of CAR expression, and local administration of CAR T cells.
Collapse
|
15
|
Bernard BE, Landmann E, Jeker LT, Schumann K. CRISPR/Cas-based Human T cell Engineering: Basic Research and Clinical Application. Immunol Lett 2022; 245:18-28. [DOI: 10.1016/j.imlet.2022.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/08/2022] [Accepted: 03/15/2022] [Indexed: 11/05/2022]
|
16
|
Industry update: the latest developments in the field of therapeutic delivery, November 2021. Ther Deliv 2022; 13:141-156. [PMID: 35128933 DOI: 10.4155/tde-2022-0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|