1
|
Rankin AW, Duncan BB, Allen C, Silbert SK, Shah NN. Evolving strategies for addressing CAR T-cell toxicities. Cancer Metastasis Rev 2024; 44:17. [PMID: 39674824 DOI: 10.1007/s10555-024-10227-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 10/02/2024] [Indexed: 12/16/2024]
Abstract
The field of chimeric antigen receptor (CAR) T-cell therapy has grown from a fully experimental concept to now boasting a multitude of treatments including six FDA-approved products targeting various hematologic malignancies. Yet, along with their efficacy, these therapies come with side effects requiring timely and thoughtful interventions. In this review, we discuss the most common toxicities associated with CAR T-cells to date, highlighting risk factors, prognostication, implications for critical care management, patient experience optimization, and ongoing work in the field of toxicity mitigation. Understanding the current state of the field and standards of practice is critical in order to improve and manage potential toxicities of both current and novel CAR T-cell therapies as they are applied in the clinic.
Collapse
Affiliation(s)
- Alexander W Rankin
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Brynn B Duncan
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Cecily Allen
- Division of Hematology, Department of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Critical Care Medicine, National Institutes of Health, Bethesda, MD, USA
| | - Sara K Silbert
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
2
|
Geng J, Yang Y, Li B, Yu Z, Qiu S, Zhang W, Gao S, Liu N, Liu Y, Wang B, Fan Y, Xing C, Liu X. Opto-chemogenetic inhibition of L-type Ca V1 channels in neurons through a membrane-assisted molecular linkage. CELL REPORTS METHODS 2024; 4:100898. [PMID: 39515337 PMCID: PMC11705922 DOI: 10.1016/j.crmeth.2024.100898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 08/28/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Genetically encoded inhibitors of CaV1 channels that operate via C-terminus-mediated inhibition (CMI) have been actively pursued. Here, we advance the design of CMI peptides by proposing a membrane-anchoring tag that is sufficient to link the inhibitory modules to the target channel as well as chemical and optogenetic modes of system control. We designed and implemented the constitutive and inducible CMI modules with appropriate dynamic ranges for the short and long variants of CaV1.3, both naturally occurring in neurons. Upon optical (near-infrared-responsive nanoparticles) and/or chemical (rapamycin) induction of FRB/FKBP binding, the designed peptides translocated onto the membrane via FRB-Ras, where the physical linkage requirement for CMI could be satisfied. The peptides robustly produced acute, potent, and specific inhibitions on both recombinant and neuronal CaV1 activities, including Ca2+ influx-neuritogenesis coupling. Validated through opto-chemogenetic induction, this prototype demonstrates Ca2+ channel modulation via membrane-assisted molecular linkage, promising broad applicability to diverse membrane proteins.
Collapse
Affiliation(s)
- Jinli Geng
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Yaxiong Yang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Boying Li
- School of Chemical Engineering, Hebei University of Technology, Tianjin 300401, China
| | - Zhen Yu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Shuang Qiu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Wen Zhang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Shixin Gao
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Nan Liu
- School of Life Sciences, Yunnan University, Kunming Yunnan 650091, China
| | - Yi Liu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Bo Wang
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China
| | - Yubo Fan
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China.
| | - Chengfen Xing
- School of Chemical Engineering, Hebei University of Technology, Tianjin 300401, China.
| | - Xiaodong Liu
- Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, School of Engineering Medicine, Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beihang University, Beijing 100083, China.
| |
Collapse
|
3
|
Acharya L, Garg A, Rai M, Kshetri R, Grewal US, Dhakal P. Novel chimeric antigen receptor targets and constructs for acute lymphoblastic leukemia: Moving beyond CD19. J Investig Med 2024; 72:32-46. [PMID: 37497999 DOI: 10.1177/10815589231191811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Acute lymphoblastic leukemia (ALL) is the second most common acute leukemia in adults with a poor prognosis with relapsed or refractory (R/R) B-cell lineage ALL (B-ALL). Anti-CD19 chimeric antigen receptor (CAR) T-cell therapy has shown excellent response rates in RR B-ALL, but most patients relapse due to poor persistence of CAR T-cell therapy or other tumor-associated escape mechanisms. In addition, anti-CD19 CAR T-cell therapy causes several serious side effects such as cytokine release syndrome and neurotoxicity. In this review, we will discuss novel CAR targets, CAR constructs, and various strategies to boost CARs for the treatment of RR B-ALL. In addition, we discuss a few novel strategies developed to reduce the side effects of CAR.
Collapse
Affiliation(s)
- Luna Acharya
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Alpana Garg
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Manoj Rai
- Department of Internal Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Rupesh Kshetri
- Department of Internal Medicine, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Udhayvir S Grewal
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| | - Prajwal Dhakal
- Division of Hematology, Oncology, and Blood and Marrow Transplantation, University of Iowa Hospitals and Clinics, Iowa City, IA, USA
| |
Collapse
|
4
|
Santiago-Vicente Y, de Jesús Castillejos-López M, Carmona-Aparicio L, Coballase-Urrutia E, Velasco-Hidalgo L, Niembro-Zúñiga AM, Zapata-Tarrés M, Torres-Espíndola LM. Immunotherapy for Pediatric Gliomas: CAR-T Cells Against B7H3: A Review of the Literature. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2024; 23:420-430. [PMID: 37038673 DOI: 10.2174/1871527322666230406094257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND B7H3 is a co-stimulatory molecule for immune reactions found on the surface of tumor cells in a wide variety of tumors. Preclinical and clinical studies have reported it as a tumor target towards which various immunotherapy modalities could be directed. So far, good results have been obtained in hematological neoplasms; however, a contrasting situation is evident in solid tumors, including those of the CNS, which show high refractoriness to current treatments. The appearance of cellular immunotherapies has transformed oncology due to the reinforcement of the immune response that is compromised in people with cancer. OBJECTIVE This article aims to review the literature to describe the advancement in knowledge on B7H3 as a target of CAR-T cells in pediatric gliomas to consider them as an alternative in the treatment of these patients. RESULTS Although B7H3 is considered a suitable candidate as a target agent for various immunotherapy techniques, there are still limitations in using CAR-T cells to achieve the desired success. CONCLUSION Results obtained with CAR-T cells can be further improved by the suggested proposals; therefore, more clinical trials are needed to study this new therapy in children with gliomas.
Collapse
Affiliation(s)
- Yolanda Santiago-Vicente
- Iztacala Faculty of Higher Studies, Tlalnepantla, México
- Laboratory of Pharmacology, National Institute of Pediatrics, Mexico City, México
| | | | | | | | | | | | - Marta Zapata-Tarrés
- Head of Research Coordination at Mexican Social Security Institute Foundation, Mexico City, México
| | | |
Collapse
|
5
|
Rossignoli F, Hoffman D, Atif E, Shah K. Developing and characterizing a two-layered safety switch for cell therapies. Cancer Biol Ther 2023; 24:2232146. [PMID: 37439774 PMCID: PMC10348026 DOI: 10.1080/15384047.2023.2232146] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 05/31/2023] [Accepted: 06/28/2023] [Indexed: 07/14/2023] Open
Abstract
Gene edited and engineered cell-based therapies are a promising approach for treating a variety of disorders, including cancer. However, the ability of engineered cells to persist for prolonged periods along with possible toxicity raises concerns over the safety of these approaches. Although a number of different one-dimensional suicide systems have been incorporated into therapeutic cell types, the incorporation of a two-layered suicide system that allows controlled killing of therapeutic cells at different time points is needed. In this study, we engineered a variety of therapeutic cells to express two different kill switches, RapaCasp9 and HSV-TK and utilized Rapamycin and Ganciclovir respectively to activate these kill switches. We show that the function of both RapaCasp9 and HSV-TK molecules is preserved and can be activated to induce apoptosis detected early (24 h) and late (48 h) post-activation respectively, with no toxicity. In vivo, we show the eradication of a majority of cells after treatment in subcutaneous and orthotopic models. Furthermore, we demonstrate how both suicide switches work independently and can be activated sequentially for an improved killing, thus ensuring a failsafe mechanism in case the activation of a single one of them is not sufficient to eliminate the cells. Our findings highlight the reliability of the double suicide system, effective on a variety of cells with different biological characteristics, independent of their anatomic presence.
Collapse
Affiliation(s)
- Filippo Rossignoli
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Danielle Hoffman
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Emaan Atif
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA, USA
| | - Khalid Shah
- Center for Stem Cell and Translational Immunotherapy (CSTI), Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Brigham and Women’s Hospital, Boston, MA, USA
- Harvard Stem Cell Institute, Harvard University, Boston, MA, USA
| |
Collapse
|
6
|
Bouquet L, Bôle-Richard E, Warda W, Neto Da Rocha M, Trad R, Nicod C, Haderbache R, Genin D, Ferrand C, Deschamps M. RapaCaspase-9-based suicide gene applied to the safety of IL-1RAP CAR-T cells. Gene Ther 2023; 30:706-713. [PMID: 37173386 PMCID: PMC10506905 DOI: 10.1038/s41434-023-00404-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 03/30/2023] [Accepted: 04/18/2023] [Indexed: 05/15/2023]
Abstract
Even if adoptive cell transfer (ACT) has already shown great clinical efficiency in different types of disease, such as cancer, some adverse events consistently occur, and suicide genes are an interesting system to manage these events. Our team developed a new medical drug candidate, a chimeric antigen receptor (CAR) targeting interleukin-1 receptor accessory protein (IL-1RAP), which needs to be evaluated in clinical trials with a clinically applicable suicide gene system. To prevent side effects and ensure the safety of our candidate, we devised two constructs carrying an inducible suicide gene, RapaCasp9-G or RapaCasp9-A, containing a single-nucleotide polymorphism (rs1052576) affecting the efficiency of endogenous caspase 9. These suicide genes are activated by rapamycin and based on the fusion of human caspase 9 with a modified human FK-binding protein, allowing conditional dimerization. RapaCasp9-G- and RapaCasp9-A-expressing gene-modified T cells (GMTCs) were produced from healthy donors (HDs) and acute myeloid leukemia (AML) donors. The RapaCasp9-G suicide gene demonstrated better efficiency, and we showed its in vitro functionality in different clinically relevant culture conditions. Moreover, as rapamycin is not pharmacologically inert, we also demonstrated its safe use as part of our therapy.
Collapse
Affiliation(s)
- Lucie Bouquet
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Elodie Bôle-Richard
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Walid Warda
- CanCell Therapeutics, 25 000, Besançon, France
| | | | - Rim Trad
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Clémentine Nicod
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Rafik Haderbache
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Delphine Genin
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
| | - Christophe Ferrand
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France
- CanCell Therapeutics, 25 000, Besançon, France
| | - Marina Deschamps
- Univ. Bourgogne Franche-Comté, INSERM, EFS BFC, UMR1098, RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, F-25 000, Besançon, France.
- CanCell Therapeutics, 25 000, Besançon, France.
| |
Collapse
|
7
|
Jerez-Longres C, Gómez-Matos M, Becker J, Hörner M, Wieland FG, Timmer J, Weber W. Engineering a material-genetic interface as safety switch for embedded therapeutic cells. BIOMATERIALS ADVANCES 2023; 150:213422. [PMID: 37084636 DOI: 10.1016/j.bioadv.2023.213422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/28/2023] [Accepted: 04/04/2023] [Indexed: 04/23/2023]
Abstract
Encapsulated cell-based therapies involve the use of genetically-modified cells embedded in a material in order to produce a therapeutic agent in a specific location in the patient's body. This approach has shown great potential in animal model systems for treating diseases such as type I diabetes or cancer, with selected approaches having been tested in clinical trials. Despite the promise shown by encapsulated cell therapy, though, there are safety concerns yet to be addressed, such as the escape of the engineered cells from the encapsulation material and the resulting production of therapeutic agents at uncontrolled sites in the body. For that reason, there is great interest in the implementation of safety switches that protect from those side effects. Here, we develop a material-genetic interface as safety switch for engineered mammalian cells embedded into hydrogels. Our switch allows the therapeutic cells to sense whether they are embedded in the hydrogel by means of a synthetic receptor and signaling cascade that link transgene expression to the presence of an intact embedding material. The system design is highly modular, allowing its flexible adaptation to other cell types and embedding materials. This autonomously acting switch constitutes an advantage over previously described safety switches, which rely on user-triggered signals to modulate activity or survival of the implanted cells. We envision that the concept developed here will advance the safety of cell therapies and facilitate their translation to clinical evaluation.
Collapse
Affiliation(s)
- Carolina Jerez-Longres
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany; SGBM - Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstrasse 19a, 79104 Freiburg, Germany
| | - Marieta Gómez-Matos
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Jan Becker
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Maximilian Hörner
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany
| | - Franz-Georg Wieland
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Institute of Physics, University of Freiburg, Hermann-Herder-Strasse 3, 79104 Freiburg, Germany; Freiburg Center for Data Analysis and Modelling (FDM), University of Freiburg, Ernst-Zermelo-Strasse 1, 79104 Freiburg, Germany
| | - Jens Timmer
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Institute of Physics, University of Freiburg, Hermann-Herder-Strasse 3, 79104 Freiburg, Germany; Freiburg Center for Data Analysis and Modelling (FDM), University of Freiburg, Ernst-Zermelo-Strasse 1, 79104 Freiburg, Germany
| | - Wilfried Weber
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany; Department of Materials Science and Materials Engineering, Saarland University, 66123 Saarbrücken, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Schänzlestrasse 18, 79104 Freiburg, Germany; Faculty of Biology, University of Freiburg, Schänzlestrasse 1, 79104 Freiburg, Germany; SGBM - Spemann Graduate School of Biology and Medicine, University of Freiburg, Albertstrasse 19a, 79104 Freiburg, Germany.
| |
Collapse
|
8
|
Rao P, Furst L, Meyran D, Mayoh C, Neeson PJ, Terry R, Khuong-Quang DA, Mantamadiotis T, Ekert PG. Advances in CAR T cell immunotherapy for paediatric brain tumours. Front Oncol 2022; 12:873722. [PMID: 36505819 PMCID: PMC9727400 DOI: 10.3389/fonc.2022.873722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Accepted: 11/02/2022] [Indexed: 11/24/2022] Open
Abstract
Brain tumours are the most common solid tumour in children and the leading cause of cancer related death in children. Current treatments include surgery, chemotherapy and radiotherapy. The need for aggressive treatment means many survivors are left with permanent severe disability, physical, intellectual and social. Recent progress in immunotherapy, including genetically engineered T cells with chimeric antigen receptors (CARs) for treating cancer, may provide new avenues to improved outcomes for patients with paediatric brain cancer. In this review we discuss advances in CAR T cell immunotherapy, the major CAR T cell targets that are in clinical and pre-clinical development with a focus on paediatric brain tumours, the paediatric brain tumour microenvironment and strategies used to improve CAR T cell therapy for paediatric tumours.
Collapse
Affiliation(s)
- Padmashree Rao
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia
| | - Liam Furst
- Department of Microbiology & Immunology, The University of Melbourne, Victoria, VIC, Australia,Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC, Australia
| | - Deborah Meyran
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia,Université de Paris, Inserm, U976 Human Immunology Pathophysiology Immunotherapy (HIPI) Unit, Institut de Recherche Saint-Louis, Paris, France,Children’s Cancer Centre, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Chelsea Mayoh
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,School of Women and Children’s Health, University of New South Wales, Randwick, NSW, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Rachael Terry
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,School of Women and Children’s Health, University of New South Wales, Randwick, NSW, Australia
| | - Dong-Anh Khuong-Quang
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC, Australia,Children’s Cancer Centre, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Theo Mantamadiotis
- Department of Microbiology & Immunology, The University of Melbourne, Victoria, VIC, Australia,Department of Surgery Royal Melbourne Hospital (RMH), The University of Melbourne, Parkville, VIC, Australia,*Correspondence: Theo Mantamadiotis, ; Paul G. Ekert,
| | - Paul G. Ekert
- Translational Tumour Biology, Children’s Cancer Institute, Randwick, NSW, Australia,Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC, Australia,Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia,Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia,School of Women and Children’s Health, University of New South Wales, Randwick, NSW, Australia,Department of Paediatrics, University of Melbourne, Parkville, VIC, Australia,*Correspondence: Theo Mantamadiotis, ; Paul G. Ekert,
| |
Collapse
|
9
|
Zhou Z, Tao C, Li J, Tang JCO, Chan ASC, Zhou Y. Chimeric antigen receptor T cells applied to solid tumors. Front Immunol 2022; 13:984864. [PMID: 36389701 PMCID: PMC9659902 DOI: 10.3389/fimmu.2022.984864] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/18/2022] [Indexed: 12/02/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy is novel tumor immunotherapy that enables autologous T to express synthetic receptors to specifically recognize the surface tumor-associated antigens for exerting subsequent antitumor effects, and eliminating the resistance, metastases and recurrence of cancer. Although CAR T cells have exhibited success in eradicating hematologic malignancies, their applications to solid tumors has not yet been achieved due to obstacles such as the immune-suppressor tumor microenvironment and lack of tumor specific target antigens. In this review, we presented advancements in the development of CAR T cell therapy in solid tumors, and offered a brief summary of the challenges, as well as novel engineering and pharmaceutical interventions to overcome these barriers. Looking forward, we discussed the latest studies which are expected to reach the clinicals in the next few years, including CRISPR screens-based CAR modification and CAR T cells driven from progenitor-like T cells. Collectively, this review may inspire researchers and clinicians to develop clinical available strategies of CAR T cell therapies in solid tumor.
Collapse
Affiliation(s)
- Zhongguo Zhou
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, Australia
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Can Tao
- School of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Jianting Li
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Johnny Cheuk-on Tang
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hong Kong, Hong Kong SAR, China
- Kamford Genetics Company Limited, Hong Kong, Hong Kong SAR, China
| | - Albert Sun-chi Chan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuanyuan Zhou
- School of Biomedical Engineering, Sun Yat-sen University, Guangzhou, Guangdong, China
- *Correspondence: Yuanyuan Zhou,
| |
Collapse
|
10
|
Chiriaco C, Donini C, Cortese M, Ughetto S, Modica C, Martinelli I, Proment A, Vitali L, Fontani L, Casucci M, Comoglio PM, Giordano S, Sangiolo D, Leuci V, Vigna E. Efficacy of CAR-T immunotherapy in MET overexpressing tumors not eligible for anti-MET targeted therapy. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2022; 41:309. [PMID: 36271379 PMCID: PMC9585715 DOI: 10.1186/s13046-022-02479-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 08/30/2022] [Indexed: 11/07/2022]
Abstract
Background Aberrant activation of the MET receptor in cancer is sustained by genetic alterations or, more frequently, by transcriptional upregulations. A fraction of MET-amplified or mutated tumors are sensible to MET targeting agents, but their responsiveness is typically short-lasting, as secondary resistance eventually occurs. Since in the absence of genetic alterations MET is usually not a tumor driver, MET overexpressing tumors are not/poorly responsive to MET targeted therapies. Consequently, the vast majority of tumors exhibiting MET activation still represent an unmet medical need. Methods Here we propose an immunotherapy strategy based on T lymphocytes expressing a Chimeric Antigen Receptor (CAR) targeting MET overexpressing tumors of different histotypes. We engineered two different MET-CAR constructs and tested MET-CAR-T cell cytotoxic activity against different MET overexpressing models, including tumor cell lines, primary cancer cells, organoids, and xenografts in immune-deficient mice. Results We proved that MET-CAR-T exerted a specific cytotoxic activity against MET expressing cells. Cell killing was proportional to the level of MET expressed on the cell surface. While CAR-T cytotoxicity was minimal versus cells carrying MET at physiological levels, essentially sparing normal cells, the activity versus MET overexpressing tumors was robust, significantly controlling tumor cell growth in vitro and in vivo. Notably, MET-CAR-T cells were also able to brake acquired resistance to MET targeting agents in MET amplified cancer cells carrying secondary mutations in downstream signal transducers. Conclusions We set and validated at the pre-clinical level a MET-CAR immunotherapy strategy potentially beneficial for cancers not eligible for MET targeted therapy with inhibitory molecules, including those exhibiting primary or secondary resistance. Supplementary Information The online version contains supplementary material available at 10.1186/s13046-022-02479-y.
Collapse
Affiliation(s)
- Cristina Chiriaco
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,Present address: Anemocyte S.r.l., 21040 Gerenzano, VA Italy
| | - Chiara Donini
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| | - Marco Cortese
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| | - Stefano Ughetto
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy ,Present address: Bios-Therapy, Physiological System for Health S.p.A, 52037 Sansepolcro, AR Italy
| | - Chiara Modica
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.10776.370000 0004 1762 5517Present address: Department of Surgical, Oncological and Stomatological Sciences (DICHIRONS), University of Palermo, 90127 Palermo, Italy
| | - Ilaria Martinelli
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy
| | - Alessia Proment
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| | - Letizia Vitali
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| | - Lara Fontani
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy
| | - Monica Casucci
- grid.18887.3e0000000417581884Innovative Immunotherapies Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Maria Comoglio
- grid.7678.e0000 0004 1757 7797IFOM-FIRC Institute of Molecular Oncology, Milan, Italy
| | - Silvia Giordano
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| | - Dario Sangiolo
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| | - Valeria Leuci
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy
| | - Elisa Vigna
- grid.419555.90000 0004 1759 7675Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, 10060 Candiolo, TO Italy ,grid.7605.40000 0001 2336 6580Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
11
|
Ou Z, Qiu L, Rong H, Li B, Ren S, Kuang S, Lan T, Lin H, Li Q, Wu F, Cai T, Yan L, Ye Y, Fan S, Li J. Bibliometric Analysis of Chimeric Antigen Receptor-Based Immunotherapy in Cancers From 2001 to 2021. Front Immunol 2022; 13:822004. [PMID: 35432345 PMCID: PMC9005877 DOI: 10.3389/fimmu.2022.822004] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/07/2022] [Indexed: 12/21/2022] Open
Abstract
Background Chimeric antigen receptor (CAR)-based immunotherapy has shown great potential for the treatment of both hematopoietic malignancies and solid tumors. Nevertheless, multiple obstacles still block the development of CAR-based immunotherapy in the clinical setting. In this study, we aimed to summarize the research landscape and highlight the front lines and trends of this field. Methods Literature published from 2001 to 2021 was searched in the Web of Science Core Collection database. Full records and cited references of all the documents were extracted and screened. Bibliometric analysis and visualization were conducted using CiteSpace, Microsoft Excel 2019, VOSviewer and R software. Results A total of 5981 articles and reviews were included. The publication and citation results exhibited increasing trends in the last 20 years. Frontiers in Immunology and Blood were the most productive and most co-cited journals, respectively. The United States was the country with the most productive organizations and publications in the comprehensive worldwide cooperation network, followed by China and Germany. June, C.H. published the most papers with the most citations, while Maude, S.L. ranked first among the co-cited authors. The hotspots in CAR-based therapy research were multiple myeloma, safety and toxicity, solid tumors, CAR-engineered immune cells beyond T cells, and gene editing. Conclusion CAR-based immunotherapy is a promising treatment for cancer patients, and there is an emerging movement toward using advanced gene modification technologies to overcome therapeutic challenges, especially in solid tumors, and to generate safer and more effective universal CAR-engineered cell products.
Collapse
Affiliation(s)
- Zhanpeng Ou
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Ling Qiu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Haixu Rong
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Bowen Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Siqi Ren
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Shijia Kuang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Tianjun Lan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Hsinyu Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Qunxing Li
- Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Fan Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Tingting Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Lingjian Yan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Yushan Ye
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China.,Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinsong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Guangzhou, China.,Department of Oral and Maxillofacial Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
12
|
Dai X, Li J, Chen Y, Ostrikov KK. When Onco-Immunotherapy Meets Cold Atmospheric Plasma: Implications on CAR-T Therapies. Front Oncol 2022; 12:837995. [PMID: 35280746 PMCID: PMC8905244 DOI: 10.3389/fonc.2022.837995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/31/2022] [Indexed: 12/03/2022] Open
Abstract
T cells engineered with chimeric antigen receptors (CAR) have demonstrated its widespread efficacy as a targeted immunotherapeutic modality. Yet, concerns on its specificity, efficacy and generalization prevented it from being established into a first-line approach against cancers. By reviewing challenges limiting its clinical application, ongoing efforts trying to resolve them, and opportunities that emerging oncotherapeutic modalities may bring to temper these challenges, we conclude that careful CAR design should be done to avoid the off-tumor effect, enhance the efficacy of solid tumor treatment, improve product comparability, and resolve problems such as differential efficacies of co-stimulatory molecules, cytokine storm, tumor lysis syndrome, myelosuppression and severe hepatotoxicity. As a promising solution, we propose potential synergies between CAR-T therapies and cold atmospheric plasma, an emerging onco-therapeutic strategy relying on reactive species, towards improved therapeutic efficacies and enhanced safety that deserve extensive investigations.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China.,CAPsoul Biotechnology Company, Ltd, Beijing, China
| | - Jitian Li
- Henan Luoyang Orthopedic Hospital (Henan Provincial Orthopedic Hospital)/Henan Provincial Orthopedic Institute, Zhengzhou, China
| | - Yiming Chen
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Kostya Ken Ostrikov
- School of Chemistry and Physics and Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
13
|
Zhong C, Liu M, Pan X, Zhu H. Tumorigenicity Risk of iPSCs in vivo: Nip it in the Bud. PRECISION CLINICAL MEDICINE 2022; 5:pbac004. [PMID: 35692443 PMCID: PMC9026204 DOI: 10.1093/pcmedi/pbac004] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/23/2022] [Indexed: 11/17/2022] Open
Abstract
In 2006, Takahashi and Yamanaka first created induced pluripotent stem cells from mouse fibroblasts via the retroviral introduction of genes encoding the transcription factors Oct3/4, Sox2, Klf44, and c-Myc. Since then, the future clinical application of somatic cell reprogramming technology has become an attractive research topic in the field of regenerative medicine. Of note, considerable interest has been placed in circumventing ethical issues linked to embryonic stem cell research. However, tumorigenicity, immunogenicity, and heterogeneity may hamper attempts to deploy this technology therapeutically. This review highlights the progress aimed at reducing induced pluripotent stem cells tumorigenicity risk and how to assess the safety of induced pluripotent stem cells cell therapy products.
Collapse
Affiliation(s)
- Chaoliang Zhong
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Miao Liu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, and Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
- Shenzhen Bay Laboratory, Shenzhen 518032, Guangdong, China
| | - Haiying Zhu
- Department of Cell Biology, Naval Medical University, Shanghai, China
| |
Collapse
|
14
|
Advances in Allogeneic Cancer Cell Therapy and Future Perspectives on “Off-the-Shelf” T Cell Therapy Using iPSC Technology and Gene Editing. Cells 2022; 11:cells11020269. [PMID: 35053386 PMCID: PMC8773622 DOI: 10.3390/cells11020269] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/07/2022] [Accepted: 01/10/2022] [Indexed: 12/03/2022] Open
Abstract
The concept of allogeneic cell therapy was first presented over 60 years ago with hematopoietic stem cell transplantation. However, complications such as graft versus host disease (GVHD) and regimen-related toxicities remained as major obstacles. To maximize the effect of graft versus leukemia, while minimizing the effect of GVHD, donor lymphocyte infusion was utilized. This idea, which was used against viral infections, postulated that adoptive transfer of virus-specific cytotoxic T lymphocytes could reconstitute specific immunity and eliminate virus infected cells and led to the idea of banking third party cytotoxic T cells (CTLs). T cell exhaustion sometimes became a problem and difficulty arose in creating robust CTLs. However, the introduction of induced pluripotent stem cells (iPSCs) lessens such problems, and by using iPSC technology, unlimited numbers of allogeneic rejuvenated CTLs with robust and proliferative cytotoxic activity can be created. Despite this revolutionary concept, several concerns still exist, such as immunorejection by recipient cells and safety issues of gene editing. In this review, we describe approaches to a feasible “off-the-shelf” therapy that can be distributed rapidly worldwide. We also offer perspectives on the future of allogeneic cell cancer immunotherapy.
Collapse
|
15
|
Miao L, Zhang Z, Ren Z, Li Y. Application of Immunotherapy in Hepatocellular Carcinoma. Front Oncol 2021; 11:699060. [PMID: 34513678 PMCID: PMC8426571 DOI: 10.3389/fonc.2021.699060] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/09/2021] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma is one of the most common malignancies globally. It not only has a hidden onset but also progresses rapidly. Most HCC patients are already in the advanced stage of cancer when they are diagnosed, and have even lost the opportunity for surgical treatment. As an inflammation-related tumor, the immunosuppressive microenvironment of HCC can promote immune tolerance through a variety of mechanisms. Immunotherapy can activate tumor-specific immune responses, which brings a new hope for the treatment of HCC. At the present time, main immunotherapy strategies of HCC include immune checkpoint inhibitors, tumor vaccines, adoptive cell therapy, and so on. This article reviews the application and research progress of immune checkpoint inhibitors, tumor vaccines, and adoptive cell therapy in the treatment of HCC.
Collapse
Affiliation(s)
- Lele Miao
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhengchao Zhang
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Zhijian Ren
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| | - Yumin Li
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China.,Key Laboratory of the Digestive System Tumors of Gansu Province, Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
16
|
Ueda N, Cahen M, Danger Y, Moreaux J, Sirac C, Cogné M. Immunotherapy perspectives in the new era of B-cell editing. Blood Adv 2021; 5:1770-1779. [PMID: 33755093 PMCID: PMC7993091 DOI: 10.1182/bloodadvances.2020003792] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/09/2021] [Indexed: 12/27/2022] Open
Abstract
Since the early days of vaccination, targeted immunotherapy has gone through multiple conceptual changes and challenges. It now provides the most efficient and up-to-date strategies for either preventing or treating infections and cancer. Its most recent and successful weapons are autologous T cells carrying chimeric antigen receptors, engineered purposely for binding cancer-specific antigens and therefore used for so-called adoptive immunotherapy. We now face the merger of such achievements in cell therapy: using lymphocytes redirected on purpose to bind specific antigens and the clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated protein 9 (Cas9) revolution, which conferred genome-editing methodologies with both safety and efficacy. This unique affiliation will soon and considerably expand the scope of diseases susceptible to adoptive immunotherapy and of immune cells available for being reshaped as therapeutic tools, including B cells. Following the monumental success story of passive immunotherapy with monoclonal antibodies (mAbs), we are thus entering into a new era, where a combination of gene therapy/cell therapy will enable reprogramming of the patient's immune system and notably endow his B cells with the ability to produce therapeutic mAbs on their own.
Collapse
Affiliation(s)
- Natsuko Ueda
- INSERM U1236, University of Rennes 1, Etablissement Français du Sang, Rennes, France
| | - Marine Cahen
- INSERM U1262, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7276, Limoges University, Limoges, France; and
| | - Yannic Danger
- INSERM U1236, University of Rennes 1, Etablissement Français du Sang, Rennes, France
| | - Jérôme Moreaux
- CNRS UMR 9002, Institute of Human Genetics, Montpellier, France
| | - Christophe Sirac
- INSERM U1262, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7276, Limoges University, Limoges, France; and
| | - Michel Cogné
- INSERM U1236, University of Rennes 1, Etablissement Français du Sang, Rennes, France
- INSERM U1262, Centre National de la Recherche Scientifique (CNRS) Unité Mixte de Recherche (UMR) 7276, Limoges University, Limoges, France; and
| |
Collapse
|
17
|
Moghimi B, Muthugounder S, Jambon S, Tibbetts R, Hung L, Bassiri H, Hogarty MD, Barrett DM, Shimada H, Asgharzadeh S. Preclinical assessment of the efficacy and specificity of GD2-B7H3 SynNotch CAR-T in metastatic neuroblastoma. Nat Commun 2021; 12:511. [PMID: 33479234 PMCID: PMC7820416 DOI: 10.1038/s41467-020-20785-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
The ability to utilize preclinical models to predict the clinical toxicity of chimeric antigen receptor (CAR) T cells in solid tumors is tenuous, thereby necessitating the development and evaluation of gated systems. Here we found that murine GD2 CAR-T cells, specific for the tumor-associated antigen GD2, induce fatal neurotoxicity in a costimulatory domain-dependent manner. Meanwhile, human B7H3 CAR-T cells exhibit efficacy in preclinical models of neuroblastoma. Seeking a better CAR, we generated a SynNotch gated CAR-T, GD2-B7H3, recognizing GD2 as the gate and B7H3 as the target. GD2-B7H3 CAR-T cells control the growth of neuroblastoma in vitro and in metastatic xenograft mouse models, with high specificity and efficacy. These improvements come partly from the better metabolic fitness of GD2-B7H3 CAR-T cells, as evidenced by their naïve T-like post-cytotoxicity oxidative metabolism and lower exhaustion profile.
Collapse
MESH Headings
- Animals
- Cell Line, Tumor
- Cell Survival/immunology
- Cytotoxicity, Immunologic/immunology
- Gangliosides/immunology
- Gangliosides/metabolism
- Humans
- Immunotherapy, Adoptive/methods
- Mice, 129 Strain
- Mice, Inbred C57BL
- Neoplasm Metastasis
- Neuroblastoma/immunology
- Neuroblastoma/pathology
- Neuroblastoma/therapy
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Tumor Burden/immunology
- Xenograft Model Antitumor Assays/methods
- Mice
Collapse
Affiliation(s)
- Babak Moghimi
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Sakunthala Muthugounder
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA
| | - Samy Jambon
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA
| | - Rachelle Tibbetts
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA
| | - Long Hung
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA
| | - Hamid Bassiri
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Michael D Hogarty
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - David M Barrett
- Children's Hospital of Philadelphia and University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Hiroyuki Shimada
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Shahab Asgharzadeh
- Children's Hospital Los Angeles, Children's Center for Cancer and Blood Diseases, Division of Hematology, Oncology and Blood & Marrow Transplantation, and The Saban Research Institute, Los Angeles, CA, USA.
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Montaño-Samaniego M, Bravo-Estupiñan DM, Méndez-Guerrero O, Alarcón-Hernández E, Ibáñez-Hernández M. Strategies for Targeting Gene Therapy in Cancer Cells With Tumor-Specific Promoters. Front Oncol 2020; 10:605380. [PMID: 33381459 PMCID: PMC7768042 DOI: 10.3389/fonc.2020.605380] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 10/30/2020] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second cause of death worldwide, surpassed only by cardiovascular diseases, due to the lack of early diagnosis, and high relapse rate after conventional therapies. Chemotherapy inhibits the rapid growth of cancer cells, but it also affects normal cells with fast proliferation rate. Therefore, it is imperative to develop other safe and more effective treatment strategies, such as gene therapy, in order to significantly improve the survival rate and life expectancy of patients with cancer. The aim of gene therapy is to transfect a therapeutic gene into the host cells to express itself and cause a beneficial biological effect. However, the efficacy of the proposed strategies has been insufficient for delivering the full potential of gene therapy in the clinic. The type of delivery vehicle (viral or non viral) chosen depends on the desired specificity of the gene therapy. The first gene therapy trials were performed with therapeutic genes driven by viral promoters such as the CMV promoter, which induces non-specific toxicity in normal cells and tissues, in addition to cancer cells. The use of tumor-specific promoters over-expressed in the tumor, induces specific expression of therapeutic genes in a given tumor, increasing their localized activity. Several cancer- and/or tumor-specific promoters systems have been developed to target cancer cells. This review aims to provide up-to-date information concerning targeting gene therapy with cancer- and/or tumor-specific promoters including cancer suppressor genes, suicide genes, anti-tumor angiogenesis, gene silencing, and gene-editing technology, as well as the type of delivery vehicle employed. Gene therapy can be used to complement traditional therapies to provide more effective treatments.
Collapse
Affiliation(s)
- Mariela Montaño-Samaniego
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Diana M. Bravo-Estupiñan
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Oscar Méndez-Guerrero
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Ernesto Alarcón-Hernández
- Laboratorio de Genética Molecular, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| | - Miguel Ibáñez-Hernández
- Laboratorio de Terapia Génica, Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas del Instituto Politécnico Nacional, Ciudad de México, México
| |
Collapse
|
19
|
Jin Y, Dong Y, Zhang J, Sun J, Liu Y, Chen Y. The toxicity of cell therapy: Mechanism, manifestations, and challenges. J Appl Toxicol 2020; 41:659-667. [DOI: 10.1002/jat.4100] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/09/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Yongjia Jin
- Shanghai Electric Power Hospital Shanghai China
| | - Yan Dong
- Department of Musculoskeletal Oncology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology Shanghai Medical College, Fudan University Shanghai China
| | | | | | | | - Yong Chen
- Department of Musculoskeletal Oncology Fudan University Shanghai Cancer Center Shanghai China
- Department of Oncology Shanghai Medical College, Fudan University Shanghai China
| |
Collapse
|
20
|
Anti-tumour effect of the fourth-generation chimeric antigen receptor T cells targeting CD133 against cholangiocarcinoma cells. Int Immunopharmacol 2020; 89:107069. [PMID: 33242709 DOI: 10.1016/j.intimp.2020.107069] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/29/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022]
Abstract
Current treatment of cholangiocarcinoma (CCA) - a lethal bile duct cancer - is ineffective because the disease is usually diagnosed at late and advanced stage. Thus, a novel therapeutic modality is urgently required. Fourth-generation chimeric antigen receptor (CAR4) T cells was created to target CD133, a well-known cancer stem cell marker, that is highly expressed and associates with cancer progression. The anti-CD133-CAR4 T cells showed high efficacy against CD133-expressing CCA cells. Tumour cell lysis occurred in a dose- and CD133 antigen-dependent manner, and significantly higher, up to 57.59% ± 9.62 at effector to target ratio of 5:1 in a CCA cell line - KKU-213A cells, compared to mock control (p = 0.008). Similarly, significant IFN-γ (p = 0.011) and TNF-α (p = 0.002) upregulation was observed upon tumour treatment. The effectiveness of our anti-CD133-CAR4 T cells will be beneficial not only for CD133-expressing CCA, but also for other CD133-expressing tumours. This study may guide future in vivo study and clinical trials.
Collapse
|
21
|
Han F, Dellacecca ER, Barse LW, Cosgrove C, Henning SW, Ankney CM, Jaishankar D, Yemelyanov A, Krymskaya VP, Dilling DF, Le Poole IC. Adoptive T-Cell Transfer to Treat Lymphangioleiomyomatosis. Am J Respir Cell Mol Biol 2020; 62:793-804. [PMID: 32078336 DOI: 10.1165/rcmb.2019-0117oc] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Patients with lymphangioleiomyomatosis (LAM) develop pulmonary cysts associated with neoplastic, smooth muscle-like cells that feature neuroendocrine cell markers. The disease preferentially affects premenopausal women. Existing therapeutics do not cure LAM. As gp100 is a diagnostic marker expressed by LAM lesions, we proposed to target this immunogenic glycoprotein using TCR transgenic T cells. To reproduce the genetic mutations underlying LAM, we cultured Tsc2-/- kidney tumor cells from aged Tsc2 heterozygous mice and generated a stable gp100-expressing cell line by lentiviral transduction. T cells were isolated from major histocompatibility complex-matched TCR transgenic pmel-1 mice to measure cytotoxicity in vitro, and 80% cytotoxicity was observed within 48 hours. Antigen-specific cytotoxicity was likewise observed using pmel-1 TCR-transduced mouse T cells, suggesting that transgenic T cells may likewise be useful to treat LAM in vivo. On intravenous injection, slow-growing gp100+ LAM-like cells formed lung nodules that were readily detectable in severe combined immunodeficient/beige mice. Adoptive transfer of gp100-reactive but not wild-type T cells into mice significantly shrunk established lung tumors, even in the absence of anti-PD-1 therapy. These results demonstrate the treatment potential of adoptively transferred T cells to eliminate pulmonary lesions in LAM.
Collapse
Affiliation(s)
- Fei Han
- Lurie Comprehensive Cancer Center
| | | | | | | | | | - Christian M Ankney
- Department of Microbiology and Immunology, Loyola University Chicago, Maywood, Illinois
| | | | - Alexander Yemelyanov
- Division of Pulmonary and Critical Care, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Vera P Krymskaya
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; and
| | - Daniel F Dilling
- Department of Medicine, Loyola University Medical Center, Maywood, Illinois
| | - I Caroline Le Poole
- Lurie Comprehensive Cancer Center.,Department of Dermatology, Microbiology, and Immunology, Northwestern University, Chicago, Illinois
| |
Collapse
|
22
|
Wang X, Wu Z, Qiu W, Chen P, Xu X, Han W. Programming CAR T cells to enhance anti-tumor efficacy through remodeling of the immune system. Front Med 2020; 14:726-745. [PMID: 32794014 DOI: 10.1007/s11684-020-0746-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have been indicated effective in treating B cell acute lymphoblastic leukemia and non-Hodgkin lymphoma and have shown encouraging results in preclinical and clinical studies. However, CAR T cells have achieved minimal success against solid malignancies because of the additional obstacles of their insufficient migration into tumors and poor amplification and persistence, in addition to antigen-negative relapse and an immunosuppressive microenvironment. Various preclinical studies are exploring strategies to overcome the above challenges. Mobilization of endogenous immune cells is also necessary for CAR T cells to obtain their optimal therapeutic effect given the importance of the innate immune responses in the elimination of malignant tumors. In this review, we focus on the recent advances in the engineering of CAR T cell therapies to restore the immune response in solid malignancies, especially with CAR T cells acting as cellular carriers to deliver immunomodulators to tumors to mobilize the endogenous immune response. We also explored the sensitizing effects of conventional treatment approaches, such as chemotherapy and radiotherapy, on CAR T cell therapy. Finally, we discuss the combination of CAR T cells with biomaterials or oncolytic viruses to enhance the anti-tumor outcomes of CAR T cell therapies in solid tumors.
Collapse
Affiliation(s)
- Xiaohui Wang
- College of Biotechnology, Southwest University, Chongqing, 400715, China.,State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China.,Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiqiang Wu
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China
| | - Wei Qiu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China
| | - Ping Chen
- College of Biotechnology, Southwest University, Chongqing, 400715, China
| | - Xiang Xu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Stem Cell & Regenerative Medicine, Daping Hospital and Research Institute of Surgery, Chongqing, 400042, China.
| | - Weidong Han
- Molecular & Immunological Department, Bio-therapeutic Department, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
23
|
Khazal S, Kebriaei P. Debate: Transplant Is Still Necessary in the Era of Targeted Cellular Therapy for Acute Lymphoblastic Leukemia. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:713-719. [PMID: 32694050 DOI: 10.1016/j.clml.2020.06.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 06/18/2020] [Indexed: 12/19/2022]
Abstract
Despite high complete remission (CR) rates, relapse remains a significant problem among subsets of patients with B acute lymphoblastic leukemia (ALL), and is associated with poor prognosis. The recent Food and Drug Administration approval of highly effective immunotherapies for B-lineage ALL (B-ALL), blinatumomab, inotuzumab ozogamicin, and tisagenlecleucel, a chimeric antigen receptor (CAR) modified T-cell therapy, targeting CD19, or CD22, have dramatically changed the therapeutic landscape for the treatment of B-ALL, resulting in high rates of deep and durable remissions. Therefore, there is now debate regarding the role of allogeneic hematopoietic cell transplantation (HCT) in this new landscape. Herein, we review these novel agents, and discuss the sequence of therapy, including allogeneic HCT in B-ALL.
Collapse
Affiliation(s)
- Sajad Khazal
- Division of Pediatrics, Pediatric Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Children's Cancer Hospital, Houston, TX.
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
24
|
Rath JA, Arber C. Engineering Strategies to Enhance TCR-Based Adoptive T Cell Therapy. Cells 2020; 9:E1485. [PMID: 32570906 PMCID: PMC7349724 DOI: 10.3390/cells9061485] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/13/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
T cell receptor (TCR)-based adoptive T cell therapies (ACT) hold great promise for the treatment of cancer, as TCRs can cover a broad range of target antigens. Here we summarize basic, translational and clinical results that provide insight into the challenges and opportunities of TCR-based ACT. We review the characteristics of target antigens and conventional αβ-TCRs, and provide a summary of published clinical trials with TCR-transgenic T cell therapies. We discuss how synthetic biology and innovative engineering strategies are poised to provide solutions for overcoming current limitations, that include functional avidity, MHC restriction, and most importantly, the tumor microenvironment. We also highlight the impact of precision genome editing on the next iteration of TCR-transgenic T cell therapies, and the discovery of novel immune engineering targets. We are convinced that some of these innovations will enable the field to move TCR gene therapy to the next level.
Collapse
MESH Headings
- Biomedical Engineering
- Cell Engineering
- Cell- and Tissue-Based Therapy/adverse effects
- Cell- and Tissue-Based Therapy/methods
- Cell- and Tissue-Based Therapy/trends
- Gene Editing
- Genetic Therapy
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Lymphocyte Activation
- Molecular Targeted Therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Safety
- Synthetic Biology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Translational Research, Biomedical
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
| | - Caroline Arber
- Department of oncology UNIL CHUV, Ludwig Institute for Cancer Research Lausanne, Lausanne University Hospital and University of Lausanne, 1015 Lausanne, Switzerland;
| |
Collapse
|
25
|
Lipus A, Janosz E, Ackermann M, Hetzel M, Dahlke J, Buchegger T, Wunderlich S, Martin U, Cathomen T, Schambach A, Moritz T, Lachmann N. Targeted Integration of Inducible Caspase-9 in Human iPSCs Allows Efficient in vitro Clearance of iPSCs and iPSC-Macrophages. Int J Mol Sci 2020; 21:E2481. [PMID: 32260086 PMCID: PMC7177583 DOI: 10.3390/ijms21072481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 02/06/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) offer great promise for the field of regenerative medicine, and iPSC-derived cells have already been applied in clinical practice. However, potential contamination of effector cells with residual pluripotent cells (e.g., teratoma-initiating cells) or effector cell-associated side effects may limit this approach. This also holds true for iPSC-derived hematopoietic cells. Given the therapeutic benefit of macrophages in different disease entities and the feasibility to derive macrophages from human iPSCs, we established human iPSCs harboring the inducible Caspase-9 (iCasp9) suicide safety switch utilizing transcription activator-like effector nuclease (TALEN)-based designer nuclease technology. Mono- or bi-allelic integration of the iCasp9 gene cassette into the AAVS1 locus showed no effect on the pluripotency of human iPSCs and did not interfere with their differentiation towards macrophages. In both, iCasp9-mono and iCasp9-bi-allelic clones, concentrations of 0.1 nM AP20187 were sufficient to induce apoptosis in more than 98% of iPSCs and their progeny-macrophages. Thus, here we provide evidence that the introduction of the iCasp9 suicide gene into the AAVS1 locus enables the effective clearance of human iPSCs and thereof derived macrophages.
Collapse
Affiliation(s)
- Alexandra Lipus
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Ewa Janosz
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Mania Ackermann
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| | - Miriam Hetzel
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Julia Dahlke
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Theresa Buchegger
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| | - Stephanie Wunderlich
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover 30625, Germany; (S.W.); (U.M.)
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), Member of the German Center for Lung Research (DZL), Hannover Medical School, Hannover 30625, Germany; (S.W.); (U.M.)
| | - Toni Cathomen
- Institute for Transfusion Medicine and Gene Therapy, Medical Center-University of Freiburg, Freiburg 79106, Germany;
- Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg 79095, Germany
| | - Axel Schambach
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Thomas Moritz
- RG Reprogramming and Gene Therapy, Hannover Medical School, Hannover 30625, Germany; (A.L.); (E.J.); (M.H.); (T.M.)
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
| | - Nico Lachmann
- Institute of Experimental Hematology, REBIRTH, Hannover Medical School, Hannover 30625, Germany; (M.A.); (J.D.); (T.B.); (A.S.)
- RG Translational Hematology of Congenital Diseases, Hannover Medical School, Hannover 30625, Germany
| |
Collapse
|
26
|
Rovatti PE, Gambacorta V, Lorentino F, Ciceri F, Vago L. Mechanisms of Leukemia Immune Evasion and Their Role in Relapse After Haploidentical Hematopoietic Cell Transplantation. Front Immunol 2020; 11:147. [PMID: 32158444 PMCID: PMC7052328 DOI: 10.3389/fimmu.2020.00147] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Accepted: 01/20/2020] [Indexed: 01/05/2023] Open
Abstract
Over the last decade, the development of multiple strategies to allow the safe transfer from the donor to the patient of high numbers of partially HLA-incompatible T cells has dramatically reduced the toxicities of haploidentical hematopoietic cell transplantation (haplo-HCT), but this was not accompanied by a similar positive impact on the incidence of post-transplantation relapse. In the present review, we will elaborate on how the unique interplay between HLA-mismatched immune system and malignancy that characterizes haplo-HCT may impact relapse biology, shaping the selection of disease variants that are resistant to the “graft-vs.-leukemia” effect. In particular, we will present current knowledge on genomic loss of HLA, a relapse modality first described in haplo-HCT and accounting for a significant proportion of relapses in this setting, and discuss other more recently identified mechanisms of post-transplantation immune evasion and relapse, including the transcriptional downregulation of HLA class II molecules and the enforcement of inhibitory checkpoints between T cells and leukemia. Ultimately, we will review the available treatment options for patients who relapse after haplo-HCT and discuss on how a deeper insight into relapse immunobiology might inform the rational and personalized selection of therapies to improve the largely unsatisfactory clinical outcome of relapsing patients.
Collapse
Affiliation(s)
- Pier Edoardo Rovatti
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valentina Gambacorta
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Unit of Senescence in Stem Cell Aging, Differentiation and Cancer, San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Lorentino
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Vago
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
27
|
Liu D, Zhao J, Song Y. Engineering switchable and programmable universal CARs for CAR T therapy. J Hematol Oncol 2019; 12:69. [PMID: 31272471 PMCID: PMC6610960 DOI: 10.1186/s13045-019-0763-0] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 06/25/2019] [Indexed: 12/27/2022] Open
Abstract
A traditional chimeric antigen receptor (CAR) has a fixed design, and one type of CAR T cells can only target one antigen epitope. This rigid design limits clinical application and leads to exceptionally high manufacturing cost. New CARs are being engineered with a modular approach so that the antigen recognition domain is split from the signaling domain of a conventional CAR, hence the target antigen can be switched or re-directed more readily without the requirement of re-engineering the CAR T cells. This CAR system can therefore serve as a universal CAR (UniCAR). The UniCAR platform has a modular design that splits the conventional CAR to 2 separate components: (1) a signaling module that binds to a specific epitope on the switching molecule and (2) a switching module with an antigen-binding domain and a switching epitope specifically recognized by the signaling module. A variety of switchable CARs have been engineered. The switchable modular designs include the dimerizing platforms using leucine zippers and biotin-avidin system, and the neo-epitope tagging platform using FITC, 5B9, and PNE. The switch molecule serves as a synapse between the CAR T cells and the target tumor cells. The universal CAR platforms are highly versatile, are easily re-programmable, and therefore have a vast potential for broad application and may significantly lower the cost of CAR T cell therapy. However, the current modular design of the switching molecules relies on adding exogenous sequences/epitopes. These unnatural epitopes can potentially lead to new antigenicity which may lead to generation of blocking antibodies. Furthermore, the generation, preparation, and clinical applications of the switching modules per se may involve additional clinical trials and regulatory examination for safety and efficacy, since repeated administrations of these molecules/"drugs" are anticipated. Thus, these switching molecules and UniCAR CAR T cells may require separate clinical trials and invoke different regulatory processes. This whole field is medically appealing and could present new challenges in the development of novel immunotherapeutic agents.
Collapse
Affiliation(s)
- Delong Liu
- Department of Oncology, The First affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY USA
| | - Juanjuan Zhao
- The affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| | - Yongping Song
- The affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, 127 Dongming Road, Zhengzhou, 450008 China
| |
Collapse
|
28
|
Chu Y, Gardenswartz A, Termuhlen AM, Cairo MS. Advances in cellular and humoral immunotherapy - implications for the treatment of poor risk childhood, adolescent, and young adult B-cell non-Hodgkin lymphoma. Br J Haematol 2019; 185:1055-1070. [PMID: 30613939 PMCID: PMC6555680 DOI: 10.1111/bjh.15753] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Patients with relapsed, refractory or advanced stage B non-Hodgkin lymphoma (NHL) continue to have a dismal prognosis. This review summarises current and novel cellular and immunotherapy for these high-risk populations, including haematopoietic stem cell transplant, bispecific antibodies, viral-derived cytotoxic T cells, chimeric antigen receptor (CAR) T cells, and natural killer (NK) cell therapy, as discussed at the 6th International Symposium on Childhood, Adolescent and Young Adult Non-Hodgkin Lymphoma on September 26th-29th 2018 in Rotterdam, the Netherlands, and explores the future of NK/CAR NK therapies.
Collapse
Affiliation(s)
- Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
| | | | - Amanda M. Termuhlen
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Mitchell S. Cairo
- Department of Pediatrics, New York Medical College, Valhalla, NY, USA
- Department of Medicine, New York Medical College, Valhalla, NY, USA
- Department of Pathology, New York Medical College, Valhalla, NY, USA
- Department of Microbiology & Immunology, New York Medical College, Valhalla, NY, USA
- Department of Cell Biology & Anatomy, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
29
|
Bougioukli S, Vakhshori V, Ortega B, Sugiyama O, Lieberman J. Regulated ex vivo regional gene therapy for bone repair using an inducible caspase-9 suicide gene system. Gene Ther 2019; 26:230-239. [PMID: 30962534 DOI: 10.1038/s41434-019-0069-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/04/2019] [Accepted: 03/01/2019] [Indexed: 12/25/2022]
Abstract
In order to adapt ex vivo regional gene therapy for clinical applications in orthopaedic surgery, safety issues must be considered. In this study we developed a suicide approach using a dual gene expression two step transcriptional amplification lentiviral vector (LV-TSTA) encoding BMP-2 and an inducible caspase 9 (iC9) system that selectively induces apoptosis upon activation with a chemical inducer of dimerization (CID). Transduction of rat bone marrow stromal cells (RBMSCs) with LV-TSTA-iC9/BMP-2 led to abundant BMP-2 production (90.3 ± 7.9 ng/24 h/106 cells) in vitro and stimulated bone formation in a mouse muscle pouch in the absence of CID. Moreover it was shown that CID could be used to selectively induce apoptosis in iC9-transduced cells both in vitro and in vivo. Double exposure to serial dilutions of CID decreased in vitro production of BMP-2 by 85-87% and Luc activity by 97-99% in iC9/BMP-2 or iC9/Luc-transduced cells respectively. Early administration of CID (Days 0-1 post-op) in mice implanted with iC9/BMP-2-transduced RBMSCs was effective in blocking bone formation, indicating that CID was toxic to the transduced cells. In iC9/Luc-implanted mice, late administration of two doses of CID (Days 27-28 post-op) significantly reduced the luciferase signal. The current study provides proof of concept for the potential clinical application of regulated gene therapy to promote bone repair.
Collapse
Affiliation(s)
- Sofia Bougioukli
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Venus Vakhshori
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brandon Ortega
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Osamu Sugiyama
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jay Lieberman
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
30
|
Dholaria BR, Bachmeier CA, Locke F. Mechanisms and Management of Chimeric Antigen Receptor T-Cell Therapy-Related Toxicities. BioDrugs 2019; 33:45-60. [PMID: 30560413 PMCID: PMC6733400 DOI: 10.1007/s40259-018-0324-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has proven to be a very effective cancer immunotherapy. Axicabtagene ciloleucel and tisagenlecleucel are the first-in-class anti-CD19 CAR-T currently available for relapsed/refractory adult large B-cell lymphoma. Tisagenlecleucel is also available for pediatric and young adult (up to age 25 years) patients with relapsed/refractory B-acute lymphoblastic leukemia. Cytokine release syndrome (CRS) and CAR-T-associated encephalopathy syndrome (neurotoxicity) are the most common adverse effects associated with CAR-T therapy. They can lead to significant morbidity and preclude widespread use of this treatment modality. Treatment-related deaths from severe CRS and cerebral edema have been reported. There is a significant heterogeneity in the side-effect profile of different CAR-T products under investigation and there is a need to develop standardized guidelines for toxicity grading and management. Here, we summarize the current literature on pathogenesis, clinical presentation, and management of CRS and neurotoxicity. The different grading systems of CRS and management protocols used in different trials have made it difficult to compare the outcomes of different CAR-T therapies. Several prevention strategies such as predictive biomarkers of CRS and neurotoxicity and modified CAR-T with 'built-in' safety mechanisms are being studied, with the potential to greatly expand the safety and applicability of CAR-T treatment across various malignancies.
Collapse
Affiliation(s)
- Bhagirathbhai R Dholaria
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, 12902 Magnolia Drive, FOB-3, Tampa, FL, 33612, USA
| | | | - Frederick Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, 12902 Magnolia Drive, FOB-3, Tampa, FL, 33612, USA.
| |
Collapse
|
31
|
Liu Y, Bewersdorf JP, Stahl M, Zeidan AM. Immunotherapy in acute myeloid leukemia and myelodysplastic syndromes: The dawn of a new era? Blood Rev 2018; 34:67-83. [PMID: 30553527 DOI: 10.1016/j.blre.2018.12.001] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/02/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
Immunotherapy has revolutionized therapy in both solid and liquid malignancies. The ability to cure acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) with an allogeneic hematopoietic stem cell transplant (HSCT) is proof of concept for the application of immunotherapy in AML and MDS. However, outside of HSCT, only the anti-CD33 antibody drug conjugate gemtuzumab ozogamicin is currently approved as an antibody-targeted therapy for AML. Several avenues of immunotherapeutic drugs are currently in different stages of clinical development. Here, we review recent advances in antibody-based therapy, immune checkpoint inhibitors, vaccines and adoptive cell-based therapy for patients with AML and MDS. First, we discuss different antibody constructs. Immune checkpoint inhibitors targeting cytotoxic T-lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein-1 (PD-1) and CD47 as well as peptide, dendritic cell and dendritic/AML cell-based vaccines are reviewed next. Lastly, adoptive cell-based therapy including chimeric antigen receptor (CAR)-T cell and NK cell therapy is discussed.
Collapse
Affiliation(s)
- Yuxin Liu
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Medicine, Section of Hematologic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
32
|
Kolos JM, Voll AM, Bauder M, Hausch F. FKBP Ligands-Where We Are and Where to Go? Front Pharmacol 2018; 9:1425. [PMID: 30568592 PMCID: PMC6290070 DOI: 10.3389/fphar.2018.01425] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 11/19/2018] [Indexed: 12/24/2022] Open
Abstract
In recent years, many members of the FK506-binding protein (FKBP) family were increasingly linked to various diseases. The binding domain of FKBPs differs only in a few amino acid residues, but their biological roles are versatile. High-affinity ligands with selectivity between close homologs are scarce. This review will give an overview of the most prominent ligands developed for FKBPs and highlight a perspective for future developments. More precisely, human FKBPs and correlated diseases will be discussed as well as microbial FKBPs in the context of anti-bacterial and anti-fungal therapeutics. The last section gives insights into high-affinity ligands as chemical tools and dimerizers.
Collapse
Affiliation(s)
| | | | | | - Felix Hausch
- Department of Chemistry, Institute of Chemistry and Biochemistry, Darmstadt University of Technology, Darmstadt, Germany
| |
Collapse
|
33
|
Rein LA, Yang H, Chao NJ. Applications of Gene Editing Technologies to Cellular Therapies. Biol Blood Marrow Transplant 2018; 24:1537-1545. [DOI: 10.1016/j.bbmt.2018.03.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 03/23/2018] [Indexed: 12/26/2022]
|
34
|
Ireland SM, Southan C, Dominguez-Monedero A, Harding SD, Sharman JL, Davies JA. SynPharm: A Guide to PHARMACOLOGY Database Tool for Designing Drug Control into Engineered Proteins. ACS OMEGA 2018; 3:7993-8002. [PMID: 30087931 PMCID: PMC6068591 DOI: 10.1021/acsomega.8b00659] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 06/26/2018] [Indexed: 05/03/2023]
Abstract
A major challenge in synthetic biology, particularly for mammalian systems, is the inclusion of adequate external control for the synthetic system activities. Control at the transcriptional level can be achieved by adaptation of bacterial repressor-operator systems (e.g., TetR), but altering the activity of a protein by controlling transcription is indirect and for longer half-life mRNAs, decreasing activity this way can be inconveniently slow. Where possible, direct modulation of protein activity by soluble ligands has many advantages, including rapid action. Decades of drug discovery and pharmacological research have uncovered detailed information on the interactions between large numbers of small molecules and their primary protein targets (as well as off-target secondary interactions), many of which have been well studied in mammals, including humans. In principle, this accumulated knowledge would be a powerful resource for synthetic biology. Here, we present SynPharm, a tool that draws together information from the pharmacological database GtoPdb and the structural database, PDB, to help synthetic biologists identify ligand-binding domains of natural proteins. Consequently, as sequence cassettes, these may be suitable for building into engineered proteins to confer small-molecule modulation on them. The tool has ancillary utilities which include assessing contact changes among different ligands in the same protein, predicting possible effects of genetic variants on binding residues, and insights into ligand cross-reactivity among species.
Collapse
|
35
|
Kean LS. Defining success with cellular therapeutics: the current landscape for clinical end point and toxicity analysis. Blood 2018; 131:2630-2639. [PMID: 29728399 PMCID: PMC6032897 DOI: 10.1182/blood-2018-02-785881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 04/11/2018] [Indexed: 12/19/2022] Open
Abstract
Cellular therapies play a major and expanding role in the treatment of hematologic diseases. For each of these therapies, a narrow therapeutic window exists, where efficacy is maximized and toxicities minimized. This review focuses on one of the most established cellular therapies, hematopoietic stem cell transplant, and one of the newest cellular therapies, chimeric antigen receptor-T cells. In this review, I will discuss the current state of the field for clinical end point analysis with each of these therapeutics, including their critical toxicities, and focus on the major elements of success for each of these complex treatments for hematologic disease.
Collapse
Affiliation(s)
- Leslie S Kean
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, WA; Clinical Research Division, The Fred Hutchinson Cancer Research Center, Seattle, WA; and Department of Pediatrics, University of Washington, Seattle, WA
| |
Collapse
|
36
|
Perales MA, Kebriaei P, Kean LS, Sadelain M. Reprint of: Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR. Biol Blood Marrow Transplant 2018; 24:S15-S19. [PMID: 29425516 DOI: 10.1016/j.bbmt.2017.12.789] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 01/14/2023]
Abstract
Therapeutic T cell engineering has recently garnered widespread interest because of the success of CD19 chimeric antigen receptor (CAR) therapy. CARs are synthetic receptors for antigen that redirect the specificity and reprogram the function of the T cells in which they are genetically introduced. CARs targeting CD19, a cell surface molecule found in most leukemias and lymphomas, have yielded high remission rates in patients with chemorefractory, relapsed disease, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. The toxicities of this treatment include B cell aplasia, cytokine release syndrome (CRS), and neurotoxicity. Although reversible in most instances, these toxicities may require specific medical interventions, including transfer to intensive care to treat severe CRS. Guidelines for managing these toxicities are emerging. The recent report of a nonhuman primate model for CRS is poised to help advance the management of this syndrome. Finally, new engineering modalities, based on the use of targeted nucleases like CRISPR, may further enhance the efficacy and safety of CAR T cells.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Leslie S Kean
- The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Center, Seattle, Washington; The Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
37
|
Engineering chimeric antigen receptor-T cells for cancer treatment. Mol Cancer 2018; 17:32. [PMID: 29448937 PMCID: PMC5815249 DOI: 10.1186/s12943-018-0814-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 02/09/2018] [Indexed: 02/07/2023] Open
Abstract
Intratumor heterogeneity of tumor clones and an immunosuppressive microenvironment in cancer ecosystems contribute to inherent difficulties for tumor treatment. Recently, chimeric antigen receptor (CAR) T-cell therapy has been successfully applied in the treatment of B-cell malignancies, underscoring its great potential in antitumor therapy. However, functional challenges of CAR-T cell therapy, especially in solid tumors, remain. Here, we describe cancer-immunity phenotypes from a clonal-stromal-immune perspective and elucidate mechanisms of T-cell exhaustion that contribute to tumor immune evasion. Then we assess the functional challenges of CAR-T cell therapy, including cell trafficking and infiltration, targeted-recognition and killing of tumor cells, T-cell proliferation and persistence, immunosuppressive microenvironment and self-control regulation. Finally, we delineate tumor precision informatics and advancements in engineered CAR-T cells to counteract inherent challenges of the CAR-T cell therapy, either alone or in combination with traditional therapeutics, and highlight the therapeutic potential of this approach in future tumor precision treatment.
Collapse
|
38
|
Neelapu SS, Tummala S, Kebriaei P, Wierda W, Gutierrez C, Locke FL, Komanduri KV, Lin Y, Jain N, Daver N, Westin J, Gulbis AM, Loghin ME, de Groot JF, Adkins S, Davis SE, Rezvani K, Hwu P, Shpall EJ. Chimeric antigen receptor T-cell therapy - assessment and management of toxicities. Nat Rev Clin Oncol 2018; 15:47-62. [PMID: 28925994 PMCID: PMC6733403 DOI: 10.1038/nrclinonc.2017.148] [Citation(s) in RCA: 1650] [Impact Index Per Article: 235.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Immunotherapy using T cells genetically engineered to express a chimeric antigen receptor (CAR) is rapidly emerging as a promising new treatment for haematological and non-haematological malignancies. CAR-T-cell therapy can induce rapid and durable clinical responses, but is associated with unique acute toxicities, which can be severe or even fatal. Cytokine-release syndrome (CRS), the most commonly observed toxicity, can range in severity from low-grade constitutional symptoms to a high-grade syndrome associated with life-threatening multiorgan dysfunction; rarely, severe CRS can evolve into fulminant haemophagocytic lymphohistiocytosis (HLH). Neurotoxicity, termed CAR-T-cell-related encephalopathy syndrome (CRES), is the second most-common adverse event, and can occur concurrently with or after CRS. Intensive monitoring and prompt management of toxicities is essential to minimize the morbidity and mortality associated with this potentially curative therapeutic approach; however, algorithms for accurate and consistent grading and management of the toxicities are lacking. To address this unmet need, we formed a CAR-T-cell-therapy-associated TOXicity (CARTOX) Working Group, comprising investigators from multiple institutions and medical disciplines who have experience in treating patients with various CAR-T-cell therapy products. Herein, we describe the multidisciplinary approach adopted at our institutions, and provide recommendations for monitoring, grading, and managing the acute toxicities that can occur in patients treated with CAR-T-cell therapy.
Collapse
Affiliation(s)
- Sattva S Neelapu
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Sudhakar Tummala
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - William Wierda
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Cristina Gutierrez
- Department of Critical Care, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Frederick L Locke
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, Moffitt Cancer Center, 12902 USF Magnolia Drive, Tampa, Florida 33613, USA
| | - Krishna V Komanduri
- Adult Stem Cell Transplant Program, Sylvester Comprehensive Cancer Center, University of Miami, 1475 Northwest 12 th Avenue, Miami, Florida 33136, USA
| | - Yi Lin
- Division of Hematology, Mayo Clinic, 200 First Street South West, Rochester, Minnesota 55905, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Jason Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Alison M Gulbis
- Division of Pharmacy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Monica E Loghin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - John F de Groot
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Sherry Adkins
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Suzanne E Davis
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Patrick Hwu
- Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| | - Elizabeth J Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, Texas 77030, USA
| |
Collapse
|
39
|
Perales MA, Kebriaei P, Kean LS, Sadelain M. Building a Safer and Faster CAR: Seatbelts, Airbags, and CRISPR. Biol Blood Marrow Transplant 2018; 24:27-31. [PMID: 29032264 PMCID: PMC5901894 DOI: 10.1016/j.bbmt.2017.10.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 10/09/2017] [Indexed: 12/01/2022]
Abstract
Therapeutic T cell engineering has recently garnered widespread interest because of the success of CD19 chimeric antigen receptor (CAR) therapy. CARs are synthetic receptors for antigen that redirect the specificity and reprogram the function of the T cells in which they are genetically introduced. CARs targeting CD19, a cell surface molecule found in most leukemias and lymphomas, have yielded high remission rates in patients with chemorefractory, relapsed disease, including acute lymphoblastic leukemia, chronic lymphocytic leukemia, and non-Hodgkin lymphoma. The toxicities of this treatment include B cell aplasia, cytokine release syndrome (CRS), and neurotoxicity. Although reversible in most instances, these toxicities may require specific medical interventions, including transfer to intensive care to treat severe CRS. Guidelines for managing these toxicities are emerging. The recent report of a nonhuman primate model for CRS is poised to help advance the management of this syndrome. Finally, new engineering modalities, based on the use of targeted nucleases like CRISPR, may further enhance the efficacy and safety of CAR T cells.
Collapse
Affiliation(s)
- Miguel-Angel Perales
- Department of Medicine, Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, MD Anderson Cancer Center, Houston, Texas
| | - Leslie S Kean
- The Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Center, Seattle, Washington; The Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Pediatrics, University of Washington, Seattle, Washington
| | - Michel Sadelain
- Center for Cell Engineering and Immunology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
40
|
Pay SL, Qi X, Willard JF, Godoy J, Sankhavaram K, Horton R, Mitter SK, Quigley JL, Chang LJ, Grant MB, Boulton ME. Improving the Transduction of Bone Marrow-Derived Cells with an Integrase-Defective Lentiviral Vector. Hum Gene Ther Methods 2017; 29:44-59. [PMID: 29160102 PMCID: PMC5806075 DOI: 10.1089/hgtb.2017.082] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In lentiviral vector (LV) applications where transient transgene expression is sufficient, integrase-defective lentiviral vectors (IDLVs) are beneficial for reducing the potential for off-target effects associated with insertional mutagenesis. It was previously demonstrated that human RPE65 mRNA expression from an integrating lentiviral vector (ILV) induces endogenous Rpe65 and Cralbp mRNA expression in murine bone marrow–derived cells (BMDCs), initiating programming of the cells to retinal pigment epithelium (RPE)-like cells. These cells regenerate RPE in retinal degeneration models when injected systemically. As transient expression of RPE65 is sufficient to activate endogenous RPE-associated genes for programming BMDCs, use of an ILV is an unnecessary risk. In this study, an IDLV expressing RPE65 (IDLV3-RPE65) was generated. Transduction with IDLV3-RPE65 is less efficient than the integrating vector (ILV3-RPE65). Therefore, IDLV3-RPE65 transduction was enhanced with a combination of preloading 20 × -concentrated viral supernatant on RetroNectin at a multiplicity of infection of 50 and transduction of BMDCs by low-speed centrifugation. RPE65 mRNA levels increased from ∼12-fold to ∼25-fold (p < 0.05) after modification of the IDLV3-RPE65 transduction protocol, achieving expression similar to the ∼27-fold (p < 0.05) increase observed with ILV3-RPE65. Additionally, the study shows that the same preparation of RetroNectin can be used to coat up to three wells with no reduction in transduction. Critically, IDLV3-RPE65 transduction initiates endogenous Rpe65 mRNA expression in murine BMDCs and Cralbp/CRALBP mRNA in both murine and human BMDCs, similar to expression observed in ILV3-RPE65-transduced cells. Systemic administration of ILV3-RPE65 or IDLV3-RPE65 programmed BMDCs in a mouse model of retinal degeneration is sufficient to retain visual function and reduce retinal degeneration compared to mice receiving no treatment or naïve BMDC. It is concluded that IDLV3-RPE65 is appropriate for programming BMDCs to RPE-like cells.
Collapse
Affiliation(s)
- S Louise Pay
- 1 Department of Medical and Molecular Genetics, Indiana University School of Medicine , Indianapolis, Indiana.,2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Xiaoping Qi
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Jeffrey F Willard
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Juliana Godoy
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Kavya Sankhavaram
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Ranier Horton
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Sayak K Mitter
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Judith L Quigley
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana
| | - Lung-Ji Chang
- 4 Department of Molecular Genetics and Microbiology, University of Florida , Gainesville, Florida
| | - Maria B Grant
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| | - Michael E Boulton
- 2 Department of Ophthalmology, Eugene and Marilyn Glick Eye Institute, Indiana University School of Medicine , Indianapolis, Indiana.,3 Department of Ophthalmology, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
41
|
Audehm S, Krackhardt AM. Specific Adoptive Cellular Immunotherapy in Allogeneic Stem Cell Transplantation. Oncol Res Treat 2017; 40:691-696. [PMID: 29069663 DOI: 10.1159/000484051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) represents a treatment option for a diversity of advanced hematopoietic malignancies providing hope for long-term responses especially due to immunogenic effects associated with the treatment modality. Despite respectable progress in the field, relapses and/or opportunistic infections are major reasons for the high treatment-related mortality. However, a number of novel immunotherapeutic approaches using defined cell populations have been developed to directly target residual malignant cells as well as defined infectious diseases. We here provide an overview of current adoptive cellular immunotherapies in the context of allo-HSCT and close with an outlook on new directions within the field.
Collapse
Affiliation(s)
- Stefan Audehm
- Medizinische Klinik III, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | | |
Collapse
|
42
|
Maroun J, Muñoz-Alía M, Ammayappan A, Schulze A, Peng KW, Russell S. Designing and building oncolytic viruses. Future Virol 2017; 12:193-213. [PMID: 29387140 PMCID: PMC5779534 DOI: 10.2217/fvl-2016-0129] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/30/2017] [Indexed: 02/07/2023]
Abstract
Oncolytic viruses (OVs) are engineered and/or evolved to propagate selectively in cancerous tissues. They have a dual mechanism of action; direct killing of infected cancer cells cross-primes anticancer immunity to boost the killing of uninfected cancer cells. The goal of the field is to develop OVs that are easily manufactured, efficiently delivered to disseminated sites of cancer growth, undergo rapid intratumoral spread, selectively kill tumor cells, cause no collateral damage and pose no risk of transmission in the population. Here we discuss the many virus engineering strategies that are being pursued to optimize delivery, intratumoral spread and safety of OVs derived from different virus families. With continued progress, OVs have the potential to transform the paradigm of cancer care.
Collapse
Affiliation(s)
- Justin Maroun
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Miguel Muñoz-Alía
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Arun Ammayappan
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Autumn Schulze
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Kah-Whye Peng
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Stephen Russell
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| |
Collapse
|
43
|
Ando M, Nakauchi H. 'Off-the-shelf' immunotherapy with iPSC-derived rejuvenated cytotoxic T lymphocytes. Exp Hematol 2016; 47:2-12. [PMID: 27826124 DOI: 10.1016/j.exphem.2016.10.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/12/2016] [Accepted: 10/18/2016] [Indexed: 02/07/2023]
Abstract
Adoptive T-cell therapy to target and kill tumor cells shows promise and induces durable remissions in selected malignancies. However, for most cancers, clinical utility is limited. Cytotoxic T lymphocytes continuously exposed to viral or tumor antigens, with long-term expansion, may become unable to proliferate ("exhausted"). To exploit fully rejuvenated induced pluripotent stem cell (iPSC)-derived antigen-specific cytotoxic T lymphocytes is a potentially powerful approach. We review recent progress in engineering iPSC-derived T cells and prospects for clinical translation. We also describe the importance of introducing a suicide gene safeguard system into adoptive T-cell therapy, including iPSC-derived T-cell therapy, to protect from unexpected events in first-in-humans clinical trials.
Collapse
Affiliation(s)
- Miki Ando
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Department of Transfusion Medicine and Stem Cell Regulation, Juntendo University School of Medicine, Tokyo, Japan.
| | - Hiromitsu Nakauchi
- Division of Stem Cell Therapy, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|