1
|
Sima H, Shao W. Advancements in the design and function of bispecific CAR-T cells targeting B Cell-Associated tumor antigens. Int Immunopharmacol 2024; 142:113166. [PMID: 39298818 DOI: 10.1016/j.intimp.2024.113166] [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] [Received: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Single-targeted CAR-T has exhibited notable success in treating B-cell tumors, effectively improving patient outcomes. However, the recurrence rate among patients remains above fifty percent, primarily attributed to antigen escape and the diminished immune persistence of CAR-T cells. Over recent years, there has been a surge of interest in bispecific CAR-T cell therapies, marked by an increasing number of research articles and clinical applications annually. This paper undertakes a comprehensive review of influential studies on the design of bispecific CAR-T in recent years, examining their impact on bispecific CAR-T efficacy concerning disease classification, targeted antigens, and CAR design. Notable distinctions in antigen targeting within B-ALL, NHL, and MM are explored, along with an analysis of how CAR scFv, transmembrane region, hinge region, and co-stimulatory region design influence Bi-CAR-T efficacy across different tumors. The summary provided aims to serve as a reference for designing novel and improved CAR-Ts, facilitating more efficient treatment for B-cell malignant tumors.
Collapse
Affiliation(s)
- Helin Sima
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
2
|
Zhou S, Yang Y, Jing Y, Zhu X. Generating advanced CAR-based therapy for hematological malignancies in clinical practice: targets to cell sources to combinational strategies. Front Immunol 2024; 15:1435635. [PMID: 39372412 PMCID: PMC11449748 DOI: 10.3389/fimmu.2024.1435635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has been a milestone breakthrough in the treatment of hematological malignancies, offering an effective therapeutic option for multi-line therapy-refractory patients. So far, abundant CAR-T products have been approved by the United States Food and Drug Administration or China National Medical Products Administration to treat relapsed or refractory hematological malignancies and exhibited unprecedented clinical efficiency. However, there were still several significant unmet needs to be progressed, such as the life-threatening toxicities, the high cost, the labor-intensive manufacturing process and the poor long-term therapeutic efficacy. According to the demands, many researches, relating to notable technical progress and the replenishment of alternative targets or cells, have been performed with promising results. In this review, we will summarize the current research progress in CAR-T eras from the "targets" to "alternative cells", to "combinational drugs" in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Shu Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yuhang Yang
- The First Clinical Medical College, Wuhan University, Wuhan, China
| | - Yulu Jing
- The Second Clinical Medical College, Wuhan University, Wuhan, China
| | - Xiaoying Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
3
|
Zha C, Song J, Wan M, Lin X, He X, Wu M, Huang R. Recent advances in CAR-T therapy for the treatment of acute myeloid leukemia. Ther Adv Hematol 2024; 15:20406207241263489. [PMID: 39050113 PMCID: PMC11268017 DOI: 10.1177/20406207241263489] [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: 11/11/2023] [Accepted: 06/04/2024] [Indexed: 07/27/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy, which has demonstrated notable efficacy against B-cell malignancies and is approved by the US Food and Drug Administration for clinical use in this context, represents a significant milestone in cancer immunotherapy. However, the efficacy of CAR-T therapy for the treatment of acute myeloid leukemia (AML) is poor. The challenges associated with the application of CAR-T therapy for the clinical treatment of AML include, but are not limited to, nonspecific distribution of AML therapeutic targets, difficulties in the production of CAR-T cells, AML blast cell heterogeneity, the immunosuppressive microenvironment in AML, and treatment-related adverse events. In this review, we summarize the recent findings regarding various therapeutic targets for AML (CD33, CD123, CLL1, CD7, etc.) and the results of the latest clinical studies on these targets. Thereafter, we also discuss the challenges related to CAR-T therapy for AML and some promising strategies for overcoming these challenges, including novel approaches such as gene editing and advances in CAR design.
Collapse
Affiliation(s)
- Chenyu Zha
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Jialu Song
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Wan
- Department of Hematology, Zhujiang Hospital of Southern Medical University, No. 253 Gongyedadaozhong Road, Guangzhou, Guangdong 510282, China
| | - Xiao Lin
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaolin He
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Ming Wu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
- Department of Hematology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Huang
- Department of Hematology, Zhujiang Hospital of Southern Medical University, No. 253 Gongyedadaozhong Road, Guangzhou, Guangdong 510282, China
| |
Collapse
|
4
|
Martarelli N, Capurro M, Mansour G, Jahromi RV, Stella A, Rossi R, Longetti E, Bigerna B, Gentili M, Rosseto A, Rossi R, Cencini C, Emiliani C, Martino S, Beeg M, Gobbi M, Tiacci E, Falini B, Morena F, Perriello VM. Artificial Intelligence-Powered Molecular Docking and Steered Molecular Dynamics for Accurate scFv Selection of Anti-CD30 Chimeric Antigen Receptors. Int J Mol Sci 2024; 25:7231. [PMID: 39000338 PMCID: PMC11242627 DOI: 10.3390/ijms25137231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/16/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cells represent a revolutionary immunotherapy that allows specific tumor recognition by a unique single-chain fragment variable (scFv) derived from monoclonal antibodies (mAbs). scFv selection is consequently a fundamental step for CAR construction, to ensure accurate and effective CAR signaling toward tumor antigen binding. However, conventional in vitro and in vivo biological approaches to compare different scFv-derived CARs are expensive and labor-intensive. With the aim to predict the finest scFv binding before CAR-T cell engineering, we performed artificial intelligence (AI)-guided molecular docking and steered molecular dynamics analysis of different anti-CD30 mAb clones. Virtual computational scFv screening showed comparable results to surface plasmon resonance (SPR) and functional CAR-T cell in vitro and in vivo assays, respectively, in terms of binding capacity and anti-tumor efficacy. The proposed fast and low-cost in silico analysis has the potential to advance the development of novel CAR constructs, with a substantial impact on reducing time, costs, and the need for laboratory animal use.
Collapse
Affiliation(s)
- Nico Martarelli
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Michela Capurro
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Gizem Mansour
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (G.M.); (M.B.); (M.G.)
| | - Ramina Vossoughi Jahromi
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Arianna Stella
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Roberta Rossi
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Emanuele Longetti
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Barbara Bigerna
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Marco Gentili
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Ariele Rosseto
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Riccardo Rossi
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Chiara Cencini
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Carla Emiliani
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Marten Beeg
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (G.M.); (M.B.); (M.G.)
| | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156 Milano, Italy; (G.M.); (M.B.); (M.G.)
| | - Enrico Tiacci
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Brunangelo Falini
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| | - Francesco Morena
- Department of Chemistry, Biology, and Biotechnologies, Via del Giochetto, University of Perugia, 06122 Perugia, Italy; (C.C.); (C.E.); (S.M.)
| | - Vincenzo Maria Perriello
- Institute of Hematology and Center for Hemato-Oncology Research, University of Perugia and Santa Maria della Misericordia Hospital, 06132 Perugia, Italy; (N.M.); (M.C.); (R.V.J.); (A.S.); (R.R.); (E.L.); (B.B.); (M.G.); (A.R.); (R.R.); (E.T.); (B.F.)
| |
Collapse
|
5
|
Kennewick KT, Yamaguchi Y, Gibson J, Gerdts EA, Jeang B, Tilakawardane D, Murad JP, Chang WC, Wright SL, Thiel MS, Forman SJ, Stern LA, Priceman SJ. Nonsignaling extracellular spacer regulates tumor antigen selectivity of CAR T cells. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200789. [PMID: 38939825 PMCID: PMC11208731 DOI: 10.1016/j.omton.2024.200789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/13/2024] [Accepted: 02/27/2024] [Indexed: 06/29/2024]
Abstract
Advancing chimeric antigen receptor (CAR)-engineered T cells for the treatment of solid tumors is a major focus in the field of cellular immunotherapy. Several hurdles have hindered similar CAR T cell clinical responses in solid tumors as seen in hematological malignancies. These challenges include on-target off-tumor toxicities, which have inspired efforts to optimize CARs for improved tumor antigen selectivity and overall safety. We recently developed a CAR T cell therapy targeting prostate stem cell antigen (PSCA) for prostate and pancreatic cancers, showing improved preclinical antitumor activity and T cell persistence by optimizing the intracellular co-stimulatory domain. Similar studies were undertaken to optimize HER2-directed CAR T cells with modifications to the intracellular co-stimulatory domain for selective targeting of breast cancer brain metastasis. In the present study, we evaluate various nonsignaling extracellular spacers in these CARs to further improve tumor antigen selectivity. Our findings suggest that length and structure of the extracellular spacer can dictate the ability of CARs to selectively target tumor cells with high antigen density, while sparing cells with low antigen density. This study contributes to CAR construct design considerations and expands our knowledge of tuning solid tumor CAR T cell therapies for improved safety and efficacy.
Collapse
Affiliation(s)
- Kelly T. Kennewick
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Yukiko Yamaguchi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Jackson Gibson
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Ethan A. Gerdts
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Brook Jeang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Dileshni Tilakawardane
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - John P. Murad
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Wen-Chung Chang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Sarah L. Wright
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Michalina S. Thiel
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Stephen J. Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lawrence A. Stern
- Department of Chemical, Biological, and Materials Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Saul J. Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
6
|
Andreu-Saumell I, Rodriguez-Garcia A, Mühlgrabner V, Gimenez-Alejandre M, Marzal B, Castellsagué J, Brasó-Maristany F, Calderon H, Angelats L, Colell S, Nuding M, Soria-Castellano M, Barbao P, Prat A, Urbano-Ispizua A, Huppa JB, Guedan S. CAR affinity modulates the sensitivity of CAR-T cells to PD-1/PD-L1-mediated inhibition. Nat Commun 2024; 15:3552. [PMID: 38670972 PMCID: PMC11053011 DOI: 10.1038/s41467-024-47799-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy for solid tumors faces significant hurdles, including T-cell inhibition mediated by the PD-1/PD-L1 axis. The effects of disrupting this pathway on T-cells are being actively explored and controversial outcomes have been reported. Here, we hypothesize that CAR-antigen affinity may be a key factor modulating T-cell susceptibility towards the PD-1/PD-L1 axis. We systematically interrogate CAR-T cells targeting HER2 with either low (LA) or high affinity (HA) in various preclinical models. Our results reveal an increased sensitivity of LA CAR-T cells to PD-L1-mediated inhibition when compared to their HA counterparts by using in vitro models of tumor cell lines and supported lipid bilayers modified to display varying PD-L1 densities. CRISPR/Cas9-mediated knockout (KO) of PD-1 enhances LA CAR-T cell cytokine secretion and polyfunctionality in vitro and antitumor effect in vivo and results in the downregulation of gene signatures related to T-cell exhaustion. By contrast, HA CAR-T cell features remain unaffected following PD-1 KO. This behavior holds true for CD28 and ICOS but not 4-1BB co-stimulated CAR-T cells, which are less sensitive to PD-L1 inhibition albeit targeting the antigen with LA. Our findings may inform CAR-T therapies involving disruption of PD-1/PD-L1 pathway tailored in particular for effective treatment of solid tumors.
Collapse
Affiliation(s)
- Irene Andreu-Saumell
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Alba Rodriguez-Garcia
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain.
| | - Vanessa Mühlgrabner
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Marta Gimenez-Alejandre
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Berta Marzal
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Joan Castellsagué
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Fara Brasó-Maristany
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Hugo Calderon
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Laura Angelats
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Salut Colell
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Mara Nuding
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Marta Soria-Castellano
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Paula Barbao
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
| | - Aleix Prat
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
- Institute of Cancer and Blood Diseases, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Alvaro Urbano-Ispizua
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain
- Department of Medicine, University of Barcelona, Barcelona, Spain
| | - Johannes B Huppa
- Medical University of Vienna, Center for Pathophysiology, Infectiology and Immunology, Institute for Hygiene and Applied Immunology, Vienna, Austria
| | - Sonia Guedan
- Oncology and Hematology Department, Fundació Clínic Recerca Biomédica- IDIBAPS, Barcelona, Spain.
| |
Collapse
|
7
|
Liu Z, Lei W, Wang H, Liu X, Fu R. Challenges and strategies associated with CAR-T cell therapy in blood malignancies. Exp Hematol Oncol 2024; 13:22. [PMID: 38402232 PMCID: PMC10893672 DOI: 10.1186/s40164-024-00490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
Cellular immunotherapy, particularly CAR-T cells, has shown potential in the improvement of outcomes in patients with refractory and recurrent malignancies of the blood. However, achieving sustainable long-term complete remission for blood cancer remains a challenge, with resistance and relapse being expected outcomes for many patients. Although many studies have attempted to clarify the mechanisms of CAR-T cell therapy failure, the mechanism remains unclear. In this article, we discuss and describe the current state of knowledge regarding these factors, which include elements that influence the CAR-T cell, cancer cells as a whole, and the microenvironment surrounding the tumor. In addition, we propose prospective approaches to overcome these obstacles in an effort to decrease recurrence rates and extend patient survival subsequent to CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China.
| | - Wenhui Lei
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
- Department of Nephrology, Lishui Municipal Central Hospital, Lishui, Zhejiang, 323000, People's Republic of China
| | - Hao Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
| | - Xiaohan Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China.
| |
Collapse
|
8
|
Kalim M, Jing R, Li X, Jiang Z, Zheng N, Wang Z, Wei G, Lu Y. Essentials of CAR-T Therapy and Associated Microbial Challenges in Long Run Immunotherapy. JOURNAL OF CELLULAR IMMUNOLOGY 2024; 6:22-50. [PMID: 38883270 PMCID: PMC11172397 DOI: 10.33696/immunology.6.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has shown potential in improving outcomes for individuals with hematological malignancies. However, achieving long-term full remission for blood cancer remains challenging due to severe life-threatening toxicities such as limited anti-tumor efficacy, antigen escape, trafficking restrictions, and limited tumor invasion. Furthermore, the interactions between CAR-T cells and their host tumor microenvironments have a significant impact on CAR-T function. To overcome these considerable hurdles, fresh methodologies and approaches are needed to produce more powerful CAR-T cells with greater anti-tumor activity and less toxicity. Despite advances in CAR-T research, microbial resistance remains a significant obstacle. In this review, we discuss and describe the basics of CAR-T structures, generations, challenges, and potential risks of infections in CAR-T cell therapy.
Collapse
Affiliation(s)
- Muhammad Kalim
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Rui Jing
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Xin Li
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Zhiwu Jiang
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Ningbo Zheng
- Department of Microbiology & Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Ziyu Wang
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Guo Wei
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| | - Yong Lu
- Houston Methodist Cancer Center/Weill Cornell Medicine, Houston, TX 77030, USA
| |
Collapse
|
9
|
Wang XY, Bian MR, Lin GQ, Yu L, Zhang YM, Wu DP. Tandem bispecific CD123/CLL-1 CAR-T cells exhibit specific cytolytic effector functions against human acute myeloid leukaemia. Eur J Haematol 2024; 112:83-93. [PMID: 37712633 DOI: 10.1111/ejh.14104] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/16/2023]
Abstract
OBJECTIVES The treatment of refractory and recurrent acute myeloid leukaemia (AML) is still a challenge with poor response rates and short survival times. In an attempt to solve this problem, we constructed a tandem bispecific chimeric antigen receptor (CAR) targeting CD123 and C-type lectin-like molecule 1 (CLL-1), two different AML antigens, and verified its cytotoxic effects in vitro. METHODS We established and cultured K562 cell lines expressing both CD123 and CLL1 antigens. Single-target CAR-T cells specific to CD123 and CLL1 were engineered, alongside tandem CD123/CLL1 bispecific CAR-T cells. Flow cytometry was used to determine cell phenotypes, transfection efficiencies, cytokine release, and CAR-T-cell proliferation, and an lactate dehydrogenase assay was used to detect the cytotoxicity of CD123/CLL-1 bispecific tandem CAR-T cells in vitro. RESULTS Two types of tandem CAR-T cells exhibited significant killing effects on CLL-1 + CD123+ leukaemia cell lines and primary AML tumour cells. The killing efficiency of tandem CAR-T cells in the case of single antigen expression is comparable to that of single target CAR-T cells. When faced with dual target tumour cells, dual target CAR-T cells significantly surpass single target CAR-T cells. CD123/CLL-1 CAR-T cells in tandem targeted and killed CD123- and CLL-1-positive leukaemia cell lines and released a large number of cytokines. CONCLUSIONS CD123/CLL-1 CAR-T cells in tandem can simultaneously target CD123 and CLL-1 on AML cells, demonstrating a significant ability to kill single antigens and multi-target tumour cells. This suggests that CD123/CLL-1 CAR-T cells exhibit significant advantages in the expression of multiple antigens in a wide range of target cells, which may help overcome the challenges posed by tumour heterogeneity and evasion mechanisms.
Collapse
MESH Headings
- Humans
- Cell Line, Tumor
- Cytokines/metabolism
- Immunotherapy, Adoptive
- Interleukin-3 Receptor alpha Subunit/genetics
- Interleukin-3 Receptor alpha Subunit/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/metabolism
- Neoplasm Recurrence, Local
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes
Collapse
Affiliation(s)
- Xiang-Yu Wang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| | - Mei-Ru Bian
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| | - Guo-Qiang Lin
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
- Key Laboratory of Bone Marrow Stem Cell, Xuzhou, China
| | - Lei Yu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, Institute of Biomedical Engineering and Technology, East China Normal University, Shanghai, China
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, Shanghai, China
| | - Yan-Ming Zhang
- Department of Hematology, Huai'an Hospital Affiliated to Xuzhou Medical University, Huai'an Second People's Hospital, Huai'an, China
| | - De-Pei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
10
|
Kang JJ, Ohoka A, Sarkar CA. Designing Multivalent and Multispecific Biologics. Annu Rev Chem Biomol Eng 2023; 15:293-314. [PMID: 38064501 DOI: 10.1146/annurev-chembioeng-100722-112440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2024]
Abstract
In the era of precision medicine, multivalent and multispecific therapeutics present a promising approach for targeted disease intervention. These therapeutics are designed to interact with multiple targets simultaneously, promising enhanced efficacy, reduced side effects, and resilience against drug resistance. We dissect the principles guiding the design of multivalent biologics, highlighting challenges and strategies that must be considered to maximize therapeutic effect. Engineerable elements in multivalent and multispecific biologic design-domain affinities, valency, and spatial presentation-must be considered in the context of the molecular targets as well as the balance of important properties such as target avidity and specificity. We illuminate recent applications of these principles in designing protein and cell therapies and identify exciting future directions in this field, underscored by advances in biomolecular and cellular engineering and computational approaches. Expected final online publication date for the Annual Review of Chemical and Biomolecular Engineering , Volume 15 is June 2024. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Jennifer J Kang
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA; , ,
| | - Ayako Ohoka
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA; , ,
- Present affiliation: AbbVie Inc., North Chicago, Illinois, USA
| | - Casim A Sarkar
- Department of Biomedical Engineering, University of Minnesota, Minneapolis, Minnesota, USA; , ,
| |
Collapse
|
11
|
Nasiri F, Farrokhi K, Safarzadeh Kozani P, Mahboubi Kancha M, Dashti Shokoohi S, Safarzadeh Kozani P. CAR-T cell immunotherapy for ovarian cancer: hushing the silent killer. Front Immunol 2023; 14:1302307. [PMID: 38146364 PMCID: PMC10749368 DOI: 10.3389/fimmu.2023.1302307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/13/2023] [Indexed: 12/27/2023] Open
Abstract
As the most lethal gynecologic oncological indication, carcinoma of the ovary has been ranked as the 5th cause of cancer-related mortality in women, with a high percentage of the patients being diagnosed at late stages of the disease and a five-year survival of ~ 30%. Ovarian cancer patients conventionally undergo surgery for tumor removal followed by platinum- or taxane-based chemotherapy; however, a high percentage of patients experience tumor relapse. Cancer immunotherapy has been regarded as a silver lining in the treatment of patients with various immunological or oncological indications; however, mirvetuximab soravtansine (a folate receptor α-specific mAb) and bevacizumab (a VEGF-A-specific mAb) are the only immunotherapeutics approved for the treatment of ovarian cancer patients. Chimeric antigen receptor T-cell (CAR-T) therapy has achieved tremendous clinical success in the treatment of patients with certain B-cell lymphomas and leukemias, as well as multiple myeloma. In the context of solid tumors, CAR-T therapies face serious obstacles that limit their therapeutic benefit. Such hindrances include the immunosuppressive nature of solid tumors, impaired tumor infiltration, lack of qualified tumor-associated antigens, and compromised stimulation and persistence of CAR-Ts following administration. Over the past years, researchers have made arduous attempts to apply CAR-T therapy to ovarian cancer. In this review, we outline the principles of CAR-T therapy and then highlight its limitations in the context of solid tumors. Ultimately, we focus on preclinical and clinical findings achieved in CAR-T-mediated targeting of different ovarian cancer-associated target antigens.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Production Platforms & Analytics, Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | - Khadijeh Farrokhi
- Department of Microbial Biotechnology, Faculty of Biotechnology, Amol University of Special Modern Technologies, Amol, Iran
| | - Pouya Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Maral Mahboubi Kancha
- Department of Medical Nanotechnology, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Setareh Dashti Shokoohi
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pooria Safarzadeh Kozani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
12
|
Kausar MA, Anwar S, El-Horany HES, Khan FH, Tyagi N, Najm MZ, Sadaf, Eisa AA, Dhara C, Gantayat S. Journey of CAR T‑cells: Emphasising the concepts and advancements in breast cancer (Review). Int J Oncol 2023; 63:130. [PMID: 37830150 PMCID: PMC10622179 DOI: 10.3892/ijo.2023.5578] [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] [Received: 07/04/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
Cancer is the primary and one of the most prominent causes of the rising global mortality rate, accounting for nearly 10 million deaths annually. Specific methods have been devised to cure cancerous tumours. Effective therapeutic approaches must be developed, both at the cellular and genetic level. Immunotherapy offers promising results by providing sustained remission to patients with refractory malignancies. Genetically modified T‑lymphocytic cells have emerged as a novel therapeutic approach for the treatment of solid tumours, haematological malignancies, and relapsed/refractory B‑lymphocyte malignancies as a result of recent clinical trial findings; the treatment is referred to as chimeric antigen receptor T‑cell therapy (CAR T‑cell therapy). Leukapheresis is used to remove T‑lymphocytes from the leukocytes, and CARs are created through genetic engineering. Without the aid of a major histocompatibility complex, these genetically modified receptors lyse malignant tissues by interacting directly with the carcinogen. Additionally, the outcomes of preclinical and clinical studies reveal that CAR T‑cell therapy has proven to be a potential therapeutic contender against metastatic breast cancer (BCa), triple‑negative, and HER 2+ve BCa. Nevertheless, unique toxicities, including (cytokine release syndrome, on/off‑target tumour recognition, neurotoxicities, anaphylaxis, antigen escape in BCa, and the immunosuppressive tumour microenvironment in solid tumours, negatively impact the mechanism of action of these receptors. In this review, the potential of CAR T‑cell immunotherapy and its method of destroying tumour cells is explored using data from preclinical and clinical trials, as well as providing an update on the approaches used to reduce toxicities, which may improve or broaden the effectiveness of the therapies used in BCa.
Collapse
Affiliation(s)
- Mohd Adnan Kausar
- Department of Biochemistry, College of Medicine, University of Ha'il, Ha'il 81411, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia
| | - Sadaf Anwar
- Department of Biochemistry, College of Medicine, University of Ha'il, Ha'il 81411, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia
| | - Hemat El-Sayed El-Horany
- Department of Biochemistry, College of Medicine, University of Ha'il, Ha'il 81411, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Egypt
| | - Farida Habib Khan
- Medical and Diagnostic Research Centre, University of Ha'il, Hail 55473, Saudi Arabia
- Department of Community and Family Medicine, College of Medicine, University of Ha'il, Ha'il 81411, Saudi Arabia
| | - Neetu Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40202, USA
| | | | - Sadaf
- Department of Biotechnology, Jamia Millia Islamia, Okhla, New Delhi 110025, India
| | - Alaa Abdulaziz Eisa
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Taibah University, Medina 30002, Saudi Arabia
| | - Chandrajeet Dhara
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122003, Haryana
| | - Saumyatika Gantayat
- School of Biosciences, Apeejay Stya University, Sohna, Gurugram 122003, Haryana
| |
Collapse
|
13
|
Pérez-Amill L, Bataller À, Delgado J, Esteve J, Juan M, Klein-González N. Advancing CART therapy for acute myeloid leukemia: recent breakthroughs and strategies for future development. Front Immunol 2023; 14:1260470. [PMID: 38098489 PMCID: PMC10720337 DOI: 10.3389/fimmu.2023.1260470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/30/2023] [Indexed: 12/17/2023] Open
Abstract
Chimeric antigen receptor (CAR) T therapies are being developed for acute myeloid leukemia (AML) on the basis of the results obtained for other haematological malignancies and the need of new treatments for relapsed and refractory AML. The biggest challenge of CART therapy for AML is to identify a specific target antigen, since antigens expressed in AML cells are usually shared with healthy haematopoietic stem cells (HSC). The concomitant expression of the target antigen on both tumour and HSC may lead to on-target/off-tumour toxicity. In this review, we guide researchers to design, develop, and translate to the clinic CART therapies for the treatment of AML. Specifically, we describe what issues have to be considered to design these therapies; what in vitro and in vivo assays can be used to prove their efficacy and safety; and what expertise and facilities are needed to treat and manage patients at the hospital.
Collapse
Affiliation(s)
- Lorena Pérez-Amill
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Gyala Therapeutics S.L, Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
| | - Àlex Bataller
- Department of Haematology, Institut Clínic de Malalties Hematològiques i Oncològiques (ICHMO), Hospital Clínic de Barcelona, Barcelona, Spain
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Julio Delgado
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Haematology, Institut Clínic de Malalties Hematològiques i Oncològiques (ICHMO), Hospital Clínic de Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Jordi Esteve
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Haematology, Institut Clínic de Malalties Hematològiques i Oncològiques (ICHMO), Hospital Clínic de Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Manel Juan
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
- Hospital Sant Joan de Déu, Universidad de Barcelona, Barcelona, Spain
| | - Nela Klein-González
- Fundació de Recerca Clínic Barcelona-Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- Gyala Therapeutics S.L, Barcelona, Spain
- Department of Immunology, Centre de Diagnòstic Biomèdic (CDB), Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
14
|
Hur J, Kim H, Kim U, Kim GB, Kim J, Joo B, Cho D, Lee DS, Chung AJ. Genetically Stable and Scalable Nanoengineering of Human Primary T Cells via Cell Mechanoporation. NANO LETTERS 2023; 23:7341-7349. [PMID: 37506062 DOI: 10.1021/acs.nanolett.3c01720] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Effective tumor regression has been observed with chimeric antigen receptor (CAR) T cells; however, the development of an affordable, safe, and effective CAR-T cell treatment remains a challenge. One of the major obstacles is that the suboptimal genetic modification of T cells reduces their yield and antitumor activity, necessitating the development of a next-generation T cell engineering approach. In this study, we developed a nonviral T cell nanoengineering system that allows highly efficient delivery of diverse functional nanomaterials into primary human T cells in a genetically stable and scalable manner. Our platform leverages the unique cell deformation and restoration process induced by the intrinsic inertial flow in a microchannel to create nanopores in the cellular membrane for macromolecule internalization, leading to effective transfection with high scalability and viability. The proposed approach demonstrates considerable potential as a practical alternative technique for improving the current CAR-T cell manufacturing process.
Collapse
Affiliation(s)
- Jeongsoo Hur
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea
| | - Hyelee Kim
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, Seoul 02841, Republic of Korea
| | - Uijin Kim
- Department of Life Sciences, University of Seoul, Seoul 02504, Republic of Korea
| | - Gi-Beom Kim
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea
- MxT Biotech, Seoul 04785, Republic of Korea
| | - Jinho Kim
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Republic of Korea
| | | | - Duck Cho
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06355, Republic of Korea
- Department of Laboratory Medicine and Genetics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 03063, Republic of Korea
| | - Dong-Sung Lee
- Department of Life Sciences, University of Seoul, Seoul 02504, Republic of Korea
| | - Aram J Chung
- Department of Bioengineering, Korea University, Seoul 02841, Republic of Korea
- Interdisciplinary Program in Precision Public Health (PPH), Korea University, Seoul 02841, Republic of Korea
- MxT Biotech, Seoul 04785, Republic of Korea
- School of Biomedical Engineering, Korea University, Seoul 02841, Republic of Korea
| |
Collapse
|
15
|
Kokalaki E, Ma B, Ferrari M, Grothier T, Hazelton W, Manzoor S, Costu E, Taylor J, Bulek A, Srivastava S, Gannon I, Jha R, Gealy R, Stanczuk L, Rizou T, Robson M, El-Kholy M, Baldan V, Righi M, Sillibourne J, Thomas S, Onuoha S, Cordoba S, Pule M. Dual targeting of CD19 and CD22 against B-ALL using a novel high-sensitivity aCD22 CAR. Mol Ther 2023; 31:2089-2104. [PMID: 36945773 PMCID: PMC10362402 DOI: 10.1016/j.ymthe.2023.03.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/05/2023] [Accepted: 03/16/2023] [Indexed: 03/23/2023] Open
Abstract
CAR T cells recognizing CD19 effectively treat relapsed and refractory B-ALL and DLBCL. However, CD19 loss is a frequent cause of relapse. Simultaneously targeting a second antigen, CD22, may decrease antigen escape, but is challenging: its density is approximately 10-fold less than CD19, and its large structure may hamper immune synapse formation. The characteristics of the optimal CD22 CAR are underexplored. We generated 12 distinct CD22 antibodies and tested CARs derived from them to identify a CAR based on the novel 9A8 antibody, which was sensitive to low CD22 density and lacked tonic signaling. We found no correlation between affinity or membrane proximity of recognition epitope within Ig domains 3-6 of CD22 with CART function. The optimal strategy for CD19/CD22 CART co-targeting is undetermined. Co-administration of CD19 and CD22 CARs is costly; single CARs targeting CD19 and CD22 are challenging to construct. The co-expression of two CARs has previously been achieved using bicistronic vectors. Here, we generated a dual CART product by co-transduction with 9A8-41BBζ and CAT-41BBζ (obe-cel), the previously described CD19 CAR. CAT/9A8 CART eliminated single- and double-positive target cells in vitro and eliminated CD19- tumors in vivo. CAT/9A8 CART is being tested in a phase I clinical study (NCT02443831).
Collapse
Affiliation(s)
| | - Biao Ma
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Mathieu Ferrari
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Thomas Grothier
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Warren Hazelton
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Somayya Manzoor
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Eren Costu
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Julia Taylor
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Anna Bulek
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | | | - Isaac Gannon
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Ram Jha
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Rosalind Gealy
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Lukas Stanczuk
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Tatiana Rizou
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Mathew Robson
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | | | - Vania Baldan
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Matteo Righi
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | | | - Simon Thomas
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Shimobi Onuoha
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Shaun Cordoba
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK
| | - Martin Pule
- Autolus Ltd, The MediaWorks, 191 Wood Ln, London W12 7FP, UK; Department of Haematology, University College London, 72 Huntley Street, London WC1E 6BT, UK.
| |
Collapse
|
16
|
Ma XC, Lv X, Li Y. Development of CD30 CAR-T cells in refractory or relapsed Hodgkin's lymphoma. Expert Rev Hematol 2023; 16:1017-1023. [PMID: 37888882 DOI: 10.1080/17474086.2023.2276210] [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] [Received: 06/21/2023] [Accepted: 10/24/2023] [Indexed: 10/28/2023]
Abstract
INTRODUCTION After therapy, approximately 15% of individuals with Hodgkin's lymphoma (HL) develop relapsed or drug-resistant Hodgkin's lymphoma (r/rHL). r/rHL has a high fatality rate and poor therapeutic prognosis. CD30 CAR-T-cell therapy has emerged as a new way to treat r/rHL in recent years. However, CD30CAR-T cells are still being explored in clinical trials. To help more patients, this review focuses on current CD30CAR-T-cell advancements as well as clinical breakthroughs in treatment of r/rHL. AREAS COVERED This research examines the mechanism of action of CD30 CAR-T cells, their function in the real-world therapy of r/rHL, and the influence of different treatment regimens on treatment results. EXPERT OPINION There has been much research into CD30 CAR-T cells as a result of their successful use in treatment of r/rHL. This research has helped us to understand CD30 CAR-T-cell safety as well as the management options available before and after its administration to increase patient survival and reduce side effects.
Collapse
Affiliation(s)
- Xiao Chen Ma
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Xiao Lv
- Department of Haematology, Shan dong Provincial Hospital Affiliated to Shan dong First Medical University; Shan dong Provincial Hospital, Affiliated to Shandong University, Jinan, Shandong Province, China
| | - Ying Li
- Department of Haematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| |
Collapse
|
17
|
Zhang C, Wang L, Zhang Q, Shen J, Huang X, Wang M, Huang Y, Chen J, Xu Y, Zhao W, Qi Y, Li Y, Ou Y, Yang Z, Qian C. Screening and characterization of the scFv for chimeric antigen receptor T cells targeting CEA-positive carcinoma. Front Immunol 2023; 14:1182409. [PMID: 37304295 PMCID: PMC10248079 DOI: 10.3389/fimmu.2023.1182409] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/09/2023] [Indexed: 06/13/2023] Open
Abstract
Introduction Chimeric antigen receptor T (CAR-T) cell therapy presents a promising treatment option for various cancers, including solid tumors. Carcinoembryonic antigen (CEA) is an attractive target due to its high expression in many tumors, particularly gastrointestinal cancers, while limited expression in normal adult tissues. In our previous clinical study, we reported a 70% disease control rate with no severe side effects using a humanized CEA-targeting CAR-T cell. However, the selection of the appropriate single-chain variable fragment (scFv) significantly affects the therapeutic efficacy of CAR-T cells by defining their specific behavior towards the target antigen. Therefore, this study aimed to identify the optimal scFv and investigate its biological functions to further optimize the therapeutic potential of CAR-T cells targeting CEA-positive carcinoma. Methods We screened four reported humanized or fully human anti-CEA antibodies (M5A, hMN-14, BW431/26, and C2-45), and inserted them into a 3rd-generation CAR structure. We purified the scFvs and measured the affinity. We monitored CAR-T cell phenotype and scFv binding stability to CEA antigen through flow cytometry. We performed repeated CEA antigen stimulation assays to compare the proliferation potential and response of the four CAR-T cells, then further evaluated the anti-tumor efficacy of CAR-T cells ex vivo and in vivo. Results M5A and hMN-14 CARs displayed higher affinity and more stable CEA binding ability than BW431/26 and C2-45 CARs. During CAR-T cell production culture, hMN-14 CAR-T cells exhibit a larger proportion of memory-like T cells, while M5A CAR-T cells showed a more differentiated phenotype, suggesting a greater tonic signal of M5A scFv. M5A, hMN-14, and BW431/26 CAR-T cells exhibited effective tumor cell lysis and IFN-γ release when cocultured with CEA-positive tumor cells in vitro, correlating with the abundance of CEA expression in target cells. While C2-45 resulted in almost no tumor lysis or IFN-γ release. In a repeat CEA antigen stimulation assay, M5A showed the best cell proliferation and cytokine secretion levels. In a mouse xenograft model, M5A CAR-T cells displayed better antitumor efficacy without preconditioning. Discussion Our findings suggest that scFvs derived from different antibodies have distinctive characteristics, and stable expression and appropriate affinity are critical for robust antitumor efficacy. This study highlights the importance of selecting an optimal scFv in CAR-T cell design for effective CEA-targeted therapy. The identified optimal scFv, M5A, could be potentially applied in future clinical trials of CAR-T cell therapy targeting CEA-positive carcinoma.
Collapse
Affiliation(s)
- Chengcheng Zhang
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Linling Wang
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Qianzhen Zhang
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Junjie Shen
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Xia Huang
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Meiling Wang
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Yi Huang
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Jun Chen
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Yanmin Xu
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Wenxu Zhao
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Yanan Qi
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Yunyan Li
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Yanjiao Ou
- Department of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zhi Yang
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| | - Cheng Qian
- Chongqing Key Laboratory of Gene and Cell Therapy, Institute of Precision Medicine and Biotechnology, Chongqing Precision Biotech Co. Ltd., Chongqing, China
| |
Collapse
|
18
|
Cheng J, Ge T, Zhu X, Wang J, Zeng Y, Mu W, Cai H, Dai Z, Jin J, Yang Y, Hu G, Mao X, Zhou J, Zhu L, Huang L. Preclinical development and evaluation of nanobody-based CD70-specific CAR T cells for the treatment of acute myeloid leukemia. Cancer Immunol Immunother 2023:10.1007/s00262-023-03422-6. [PMID: 36932256 DOI: 10.1007/s00262-023-03422-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 03/05/2023] [Indexed: 03/19/2023]
Abstract
BACKGROUND Acute myeloid leukemia (AML) treatment remains challenging. CD70 was reported as a promising AML-specific antigen. Preclinically, CAR T-cell with single-chain-variable fragment (scFv) or truncated CD27 targeting CD70 has been reported to treat AML. However, various disadvantages including spontaneous exhaustion, proteinase-mediated loss of functional receptors, and high immunogenicity, limited its further application to clinical settings. Alternatively, the single-variable domain on heavy chain (VHH), also known as nanobodies, with comparable binding ability and specificity, provides an optional solution. METHOD We generated CD70 knocked-out novel nanobody-based anti-CD70-CAR T-cells (nb70CAR-T) with two different VHHs for antigen detection. Next, we detected the CD70 expression on primary AML blasts by flow cytometry and associated the efficacy of nb70CAR-T with the target antigen density. Finally, epigenetic modulators were investigated to regulate the CD70 expression on AML cells to promote the functionality of nb70CAR-T. RESULTS Our nb70CAR-T exhibited expected tumoricidal functionality against CD70-expressed cell lines and primary AML blasts. However, CD70 expression in primary AML blasts was not consistently high and nb70CAR-T potently respond to an estimated 40.4% of AML patients when the CD70 expression level was over a threshold of 1.6 (MFI ratio). Epigenetic modulators, Decitabine and Chidamide can up-regulate CD70 expression on AML cells, enhancing the treatment efficacy of nb70CAR-T. CONCLUSION CD70 expression in AML blasts was not fully supportive of its role in AML targeted therapy as reported. The combinational use of Chidamide and Decitabine with nb70CAR-T could provide a new potential for the treatment of AML.
Collapse
Affiliation(s)
- Jiali Cheng
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Tong Ge
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Jue Wang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Yuhao Zeng
- Department of Internal Medicine, Cleveland Clinic, Akron General, Akron, OH, USA
| | - Wei Mu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Haodong Cai
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Zhenyu Dai
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Jin Jin
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | | | - Guang Hu
- IASO Biotherapeutics, Nanjing, China
| | - Xia Mao
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China
| | - Li Zhu
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China.
| | - Liang Huang
- Department of Hematology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Qiaokou District, 1095 Jiefang Avenue, Wuhan, China.
| |
Collapse
|
19
|
Liu Q, Luo L, Gao X, Zhang D, Feng X, Yang P, Li H, Mao S. Co-Delivery of Daunorubicin and Homoharringtonine in Folic Acid Modified-Liposomes for Enhancing Therapeutic Effect on Acute Myeloid Leukemia. J Pharm Sci 2023; 112:123-131. [PMID: 35469834 DOI: 10.1016/j.xphs.2022.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/19/2022] [Accepted: 04/19/2022] [Indexed: 11/25/2022]
Abstract
Acute myeloid leukemia (AML) remains a threatening disease due to severe complications, drug resistance, and high recurrence rates. Many drug combinations have demonstrated enhanced therapeutic effects in clinical practice. However, it requires complicated dosing regimens and is accompanied by increased toxicity. This study explored the combined effect of two therapeutic agents, daunorubicin (DNR) and homoharringtonine (HHT) in cell viability, apoptosis, and cell cycle in vitro and verified their synergistic effect. We encapsulated the two drugs into liposomes to construct a folic acid-modified co-delivery system (FA-DH-LP) to achieve an effective and safe therapeutic strategy. The FA-DH-LP was prepared by film hydration method. The resultant FA-DH-LP was homogeneously spherical and showed good blood compatibility with high encapsulation efficiency for DNR and HHT. The FA-DH-LP exhibited higher cellular uptake in HL60 and K562 cells and enhanced cytotoxicity than DNR/HHT co-delivery liposomes without folic acid modification (DH-LP) in vitro. In the HL60 subcutaneous xenotransplantation model, FA-DH-LP showed improved tumor targeting ability, anti-leukemia activity and safety profile superior to free combinational drugs and DH-LP after 18-day treatment. The results demonstrated that FA-DH-LP might present a promising delivery strategy to improve the efficacy of the two combinational chemotherapeutics while reducing toxicity.
Collapse
Affiliation(s)
- Qi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Lijun Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xiaofeng Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Di Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Xinqian Feng
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Peng Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China
| | - Hui Li
- Department of Hematology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu 610072, China.
| | - Shengjun Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
20
|
Schorr C, Perna F. Targets for chimeric antigen receptor T-cell therapy of acute myeloid leukemia. Front Immunol 2022; 13:1085978. [PMID: 36605213 PMCID: PMC9809466 DOI: 10.3389/fimmu.2022.1085978] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 11/30/2022] [Indexed: 12/24/2022] Open
Abstract
Acute Myeloid Leukemia (AML) is an aggressive myeloid malignancy associated with high mortality rates (less than 30% 5-year survival). Despite advances in our understanding of the molecular mechanisms underpinning leukemogenesis, standard-of-care therapeutic approaches have not changed over the last couple of decades. Chimeric Antigen Receptor (CAR) T-cell therapy targeting CD19 has shown remarkable clinical outcomes for patients with acute lymphoblastic leukemia (ALL) and is now an FDA-approved therapy. Targeting of myeloid malignancies that are CD19-negative with this promising technology remains challenging largely due to lack of alternate target antigens, complex clonal heterogeneity, and the increased recognition of an immunosuppressive bone marrow. We carefully reviewed a comprehensive list of AML targets currently being used in both proof-of-concept pre-clinical and experimental clinical settings. We analyzed the expression profile of these molecules in leukemic as well normal tissues using reliable protein databases and data reported in the literature and we provide an updated overview of the current clinical trials with CAR T-cells in AML. Our study represents a state-of-art review of the field and serves as a potential guide for selecting known AML-associated targets for adoptive cellular therapies.
Collapse
Affiliation(s)
- Christopher Schorr
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States,Department of Biomedical Engineering, Purdue University Weldon School of Biomedical Engineering, West Lafayette, IN, United States
| | - Fabiana Perna
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, United States,*Correspondence: Fabiana Perna,
| |
Collapse
|
21
|
Mao R, Kong W, He Y. The affinity of antigen-binding domain on the antitumor efficacy of CAR T cells: Moderate is better. Front Immunol 2022; 13:1032403. [PMID: 36325345 PMCID: PMC9618871 DOI: 10.3389/fimmu.2022.1032403] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
The overall efficacy of chimeric antigen receptor modified T cells (CARTs) remain limited in solid tumors despite intensive studies that aim at targeting multiple antigens, enhancing migration, reducing tonic signaling, and improving tumor microenvironment. On the other hand, how the affinity and engaging kinetics of antigen-binding domain (ABD) affects the CART's efficacy has not been carefully investigated. In this article, we first analyzed 38 published solid tumor CART trials and correlated the response rate to their ABD affinity. Not surprisingly, majority (25 trials) of the CARTs utilized high-affinity ABDs, but generated merely 5.7% response rate. In contrast, 35% of the patients treated with the CARTs built from moderate-affinity ABDs had clinical responses. Thus, CARTs with moderate-affinity ABDs not only have less off-target toxicity, but also are more effective. We then reviewed the effects of ABD affinity on the biology and function of CARTs, providing further evidence that moderate-affinity ABDs may be better in CART development. In the end, we propose that a fast-on/fast-off (high Kon and Koff ) kinetics of CART-target engagement in solid tumor allow CARTs to generate sufficient signaling to kill tumor cells without being driven to exhaustion. We believe that studying the ABD affinity and the kinetics of CART-tumor interaction may hold a key to designing effective CARTs for solid tumors.
Collapse
Affiliation(s)
- Rui Mao
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Wanqing Kong
- South Carolina Governors School for Science and Math, Hartsville, SC, United States
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
22
|
To V, Evtimov VJ, Jenkin G, Pupovac A, Trounson AO, Boyd RL. CAR-T cell development for Cutaneous T cell Lymphoma: current limitations and potential treatment strategies. Front Immunol 2022; 13:968395. [PMID: 36059451 PMCID: PMC9433932 DOI: 10.3389/fimmu.2022.968395] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/27/2022] [Indexed: 11/21/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T therapy has demonstrated remarkable outcomes for B cell malignancies, however, its application for T cell lymphoma, particularly cutaneous T cell lymphoma (CTCL), has been limited. Barriers to effective CAR-T cell therapy in treating CTCL include T cell aplasia in autologous transplants, CAR-T product contamination with leukemic T cells, CAR-T fratricide (when the target antigen is present on normal T cells), and tumor heterogeneity. To address these critical challenges, innovative CAR engineering by targeting multiple antigens to strike a balance between efficacy and safety of the therapy is necessary. In this review, we discuss the current obstacles to CAR-T cell therapy and highlight potential targets in treating CTCL. Looking forward, we propose strategies to develop more powerful dual CARs that are advancing towards the clinic in CTCL therapy.
Collapse
Affiliation(s)
- Van To
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | | | - Graham Jenkin
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- The Ritchie Centre, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | | | - Alan O. Trounson
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- Department of Obstetrics and Gynaecology, Monash University, Clayton, VIC, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC, Australia
| | - Richard L. Boyd
- Cartherics Pty Ltd, Notting Hill, VIC, Australia
- *Correspondence: Richard L. Boyd,
| |
Collapse
|
23
|
Caraballo Galva LD, Jiang X, Hussein MS, Zhang H, Mao R, Brody P, Peng Y, He AR, Kehinde-Ige M, Sadek R, Qiu X, Shi H, He Y. Novel low-avidity glypican-3 specific CARTs resist exhaustion and mediate durable antitumor effects against HCC. Hepatology 2022; 76:330-344. [PMID: 34897774 PMCID: PMC10568540 DOI: 10.1002/hep.32279] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND AND AIMS Chimeric antigen receptor engineered T cells (CARTs) for HCC and other solid tumors are not as effective as they are for blood cancers. CARTs may lose function inside tumors due to persistent antigen engagement. The aims of this study are to develop low-affinity monoclonal antibodies (mAbs) and low-avidity CARTs for HCC and to test the hypothesis that low-avidity CARTs can resist exhaustion and maintain functions in solid tumors, generating durable antitumor effects. METHODS AND RESULTS New human glypican-3 (hGPC3) mAbs were developed from immunized mice. We obtained three hGPC3-specific mAbs that stained HCC tumors, but not the adjacent normal liver tissues. One of them, 8F8, bound an epitope close to that of GC33, the frequently used high-affinity mAb, but with approximately 17-fold lower affinity. We then compared the 8F8 CARTs to GC33 CARTs for their in vitro function and in vivo antitumor effects. In vitro, low-avidity 8F8 CARTs killed both hGPC3high and hGPC3low HCC tumor cells to the same extent as high-avidity GC33 CARTs. 8F8 CARTs expanded and persisted to a greater extent than GC33 CARTs, resulting in durable responses against HCC xenografts. Importantly, compared with GC33 CARTs, there were 5-fold more of 8F8-BBz CARTs in the tumor mass for a longer period of time. Remarkably, the tumor-infiltrating 8F8 CARTs were less exhausted and apoptotic, and more functional than GC33 CARTs. CONCLUSION The low-avidity 8F8-BBz CART resists exhaustion and apoptosis inside tumor lesions, demonstrating a greater therapeutic potential than high-avidity CARTs.
Collapse
Affiliation(s)
| | - Xiaotao Jiang
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Mohamed S. Hussein
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Huajun Zhang
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Rui Mao
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Pierce Brody
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yibing Peng
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Aiwu Ruth He
- Lombardi Cancer Center, Georgetown University, Washington, District of Columbia, USA
| | - Mercy Kehinde-Ige
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Ramses Sadek
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Xiangguo Qiu
- Department of Medical Microbiology and Infectious Diseases, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Huidong Shi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Yukai He
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
24
|
Development of Cancer Immunotherapies. Cancer Treat Res 2022; 183:1-48. [PMID: 35551655 DOI: 10.1007/978-3-030-96376-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cancer immunotherapy, or the utilization of components of the immune system to target and eliminate cancer, has become a highly active area of research in the past several decades and a common treatment strategy for several cancer types. The concept of harnessing the immune system for this purpose originated over 100 years ago when a physician by the name of William Coley successfully treated several of his cancer patients with a combination of live and attenuated bacteria, later known as "Coley's Toxins", after observing a subset of prior patients enter remission following their diagnosis with the common bacterial infection, erysipelas. However, it was not until late in the twentieth century that cancer immunotherapies were developed for widespread use, thereby transforming the treatment landscape of numerous cancer types. Pivotal studies elucidating molecular and cellular functions of immune cells, such as the discovery of IL-2 and production of monoclonal antibodies, fostered the development of novel techniques for studying the immune system and ultimately the development and approval of several cancer immunotherapies by the United States Food and Drug Association in the 1980s and 1990s, including the tuberculosis vaccine-Bacillus Calmette-Guérin, IL-2, and the CD20-targeting monoclonal antibody. Approval of the first therapeutic cancer vaccine, Sipuleucel-T, for the treatment of metastatic castration-resistant prostate cancer and the groundbreaking success and approval of immune checkpoint inhibitors and chimeric antigen receptor T cell therapy in the last decade, have driven an explosion of interest in and pursuit of novel cancer immunotherapy strategies. A broad range of modalities ranging from antibodies to adoptive T cell therapies is under investigation for the generalized treatment of a broad spectrum of cancers as well as personalized medicine. This chapter will focus on the recent advances, current strategies, and future outlook of immunotherapy development for the treatment of cancer.
Collapse
|
25
|
[Preparation of CD52-targeted chimeric antigen receptor-modified T cells and their anti-leukemia effects]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2022; 43:279-286. [PMID: 35680625 PMCID: PMC9189477 DOI: 10.3760/cma.j.issn.0253-2727.2022.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Objective: To construct chimeric antigen receptor (CAR) T cells targeting CD52 (CD52 CAR-T) and validate the effect of CD52 CAR-T cells on CD52-positive leukemia. Methods: A second-generation CD52-targeting CAR bearing 4-1BB costimulatory domain was ligated into a lentiviral vector through molecular cloning. Lentivirus was prepared and packaged by 293 T cells with a four-plasmid system. Fluorescein was used to label cell surface antigens to evaluate the phenotype of CD52 CAR-T cells after infection. Flow cytometry and ELISA were used to evaluate the specific cytotoxicity of CD52 CAR-T cells to CD52-positive cell lines in vitro. Results: ①A pCDH-CD52scFv-CD8α-4-1BB-CD3ζ-GFP expressing plasmid was successfully constructed and used to transduce T cells expressing a novel CD52-targeting CAR. ②On day 6, CD52-positive T cells were almost killed by CD52-targeted CAR-T post lentivirus transduction [CD52 CAR-T (4.48 ± 4.99) %, vs Vector-T (56.58±19.8) %, P=0.011]. ③T cells transduced with the CAR targeting CD52 showed low levels of apoptosis and could be expanded long-term ex vivo. ④The CD52 CAR could promote T cell differentiation into central and effector memory T cells, whereas the proportion of T cells with a CD45RA(+) effector memory phenotype were reduced. ⑤CD52 CAR-T cells could specifically kill CD52-positive HuT78-19t cells but had no killing effect on CD52-negative MOLT4-19t cells. For CD52 CAR-T cells, the percentage of residual of HuT78-19t cells was (2.66±1.60) % at an the E:T ratio of 1∶1 for 24 h, while (56.66±5.74) % of MOLT4-19t cells survived (P<0.001) . ⑥The results of a degranulation experiment confirmed that HuT78-19t cells significantly activated CD52 CAR-T cells but not MOLT4-19t cells[ (57.34±11.25) % vs (13.06± 4.23) %, P<0.001]. ⑦CD52 CAR-T cells released more cytokines when co-cultured with HuT78-19t cells than that of vector-T cells [IFN-γ: (3706±226) pg/ml, P<0.001; TNF-α: (1732±560) pg/ml, P<0.01]. Conclusions: We successfully prepared CD52 CAR-T cells with anti-leukemia effects, which might provide the foundation for further immunotherapy.
Collapse
|
26
|
Nguyen A, Johanning G, Shi Y. Emerging Novel Combined CAR-T Cell Therapies. Cancers (Basel) 2022; 14:cancers14061403. [PMID: 35326556 PMCID: PMC8945996 DOI: 10.3390/cancers14061403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 12/08/2022] Open
Abstract
Simple Summary As a result of FDA approval of CAR-T cell treatments in the last few years, this immunotherapy has provided further direction to precision medicine through its combination with other therapeutic approaches. In the past year, several review articles have been published focusing on advances in this fast-developing field, especially with respect to efforts to overcome hurdles associated with applying CAR-T cells in solid tumors. This review paper focuses on combining CAR-T cell therapy with small molecule drugs, up-to-date progress in CAR-T cell therapy research, and advances in combined CAR-T immunotherapy with other treatments targeting solid tumors. Abstract Chimeric antigen receptors (CAR) T cells are T cells engineered to express membrane receptors with high specificity to recognize specific target antigens presented by cancer cells and are co-stimulated with intracellular signals to increase the T cell response. CAR-T cell therapy is emerging as a novel therapeutic approach to improve T cell specificity that will lead to advances in precision medicine. CAR-T cells have had impressive outcomes in hematological malignancies. However, there continue to be significant limitations of these therapeutic responses in targeting solid malignancies such as heterogeneous antigens in solid tumors, tumor immunosuppressive microenvironment, risk of on-target/off-tumor, infiltrating CAR-T cells, immunosuppressive checkpoint molecules, and cytokines. This review paper summarizes recent approaches and innovations through combination therapies of CAR-T cells and other immunotherapy or small molecule drugs to counter the above disadvantages to potentiate the activity of CAR-T cells.
Collapse
Affiliation(s)
- Anh Nguyen
- College of Graduate Studies, California Northstate University, Elk Grove, CA 95757, USA;
| | | | - Yihui Shi
- College of Medicine, California Northstate University, Elk Grove, CA 95757, USA
- Correspondence:
| |
Collapse
|
27
|
Koedam J, Wermke M, Ehninger A, Cartellieri M, Ehninger G. Chimeric antigen receptor T-cell therapy in acute myeloid leukemia. Curr Opin Hematol 2022; 29:74-83. [PMID: 35013048 PMCID: PMC8815830 DOI: 10.1097/moh.0000000000000703] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
PURPOSE OF REVIEW Treatment outcome of relapsed or refractory AML patients remains dismal and new treatment options are needed. Adoptive cell therapy using CAR-T cells is a potentially interesting approach in this. RECENT FINDINGS Several potentially interesting AML targets are being investigated with CAR-T therapy with over 60 clinical trials listed on clinicaltrials.gov. The first clinical data are only just emerging with mixed results, once more proving that further research is needed. SUMMARY Adoptive cell therapy using chimeric antigen receptor T cells is being investigated in AML through many clinical trials. So far, no AML-specific antigen has been identified, requiring additional strategies to mitigate on-target off-tumor toxicity and to increase efficacy. Focus point is to acquire control over the CAR T cells once administered. Strategies to do so include biodegradable CARs, inducible CARs, suicide-switch containing CARs and two-component modular CARs. Limited and mixed results are available, confirming the risk of lasting toxicity for nonswitchable CARs. Initial results of modular CARs suggest toxicity can be mitigated whilst maintaining CAR activity by the use of modular CAR concepts that allows for 'ON' and 'OFF' switching.
Collapse
Affiliation(s)
| | - Martin Wermke
- Division of Hematology, Oncology and Stem Cell Transplantation, Medical Clinic I, Department of Medicine I, University Hospital Carl Gustav Carus
- National Center for Tumor Diseases
| | | | | | | |
Collapse
|
28
|
Mao R, Hussein MS, He Y. Chimeric antigen receptor engineered T cells and their application in the immunotherapy of solid tumours. Expert Rev Mol Med 2022; 24:e7. [PMID: 35086597 PMCID: PMC9617572 DOI: 10.1017/erm.2021.32] [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] [Indexed: 02/06/2023]
Abstract
In this article, we reviewed the current literature studies and our understanding of the parameters that affect the chimeric antigen receptor T cells (CAR-T's) activation, effector function, in vivo persistence, and antitumour effects. These factors include T cell subsets and their differentiation stages, the components of chimeric antigen receptors (CAR) design, the expression promoters and delivery vectors, and the CAR-T production process. The CAR signalling and CAR-T activation were also studied in comparison to TCR. The last section of the review gave special consideration of CAR design for solid tumours, focusing on strategies to improve CAR-T tumour infiltration and survival in the hostile tumour microenvironment. With several hundred clinical trials undergoing worldwide, the pace of CAR-T immunotherapy moves from bench to bedside is unprecedented. We hope that the article will provide readers a clear and comprehensive view of this rapidly evolving field and will help scientists and physician to design effective CAR-Ts immunotherapy for solid tumours.
Collapse
Affiliation(s)
- Rui Mao
- Georgia Cancer Center, Augusta, USA
| | | | - Yukai He
- Georgia Cancer Center, Augusta, USA
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, USA
| |
Collapse
|
29
|
Challenges and Advances in Chimeric Antigen Receptor Therapy for Acute Myeloid Leukemia. Cancers (Basel) 2022; 14:cancers14030497. [PMID: 35158765 PMCID: PMC8833567 DOI: 10.3390/cancers14030497] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/11/2022] [Accepted: 01/17/2022] [Indexed: 02/06/2023] Open
Abstract
The advent of chimeric antigen receptor (CAR) T-cell therapy has led to dramatic remission rates in multiple relapsed/refractory hematologic malignancies. While CAR T-cell therapy has been particularly successful as a treatment for B-cell malignancies, effectively treating acute myeloid leukemia (AML) with CARs has posed a larger challenge. AML not only creates an immunosuppressive tumor microenvironment that dampens CAR T-cell responses, but it also lacks many unique tumor-associated antigens, making leukemic-specific targeting difficult. One advantage of CAR T-cell therapy compared to alternative treatment options is the ability to provide prolonged antigen-specific immune effector and surveillance functions. Since many AML CAR targets under investigation including CD33, CD117, and CD123 are also expressed on hematopoietic stem cells, CAR T-cell therapy can lead to severe and potentially lethal myeloablation. Novel strategies to combat these issues include creation of bispecific CARs, CAR T-cell "safety switches", TCR-like CARs, NK CARs, and universal CARs, but all vary in their ability to provide a sustained remission, and consolidation with an allogeneic hematopoietic cell transplantation (allo-HCT) will be necessary in most cases This review highlights the delicate balance between effectively eliminating AML blasts and leukemic stem cells, while preserving the ability for bone marrow to regenerate. The impact of CAR therapy on treatment landscape of AML and changing scope of allo-HCT is discussed. Continued advances in AML CAR therapy would be of great benefit to a disease that still has high morbidity and mortality.
Collapse
|
30
|
Hanssens H, Meeus F, De Veirman K, Breckpot K, Devoogdt N. The antigen-binding moiety in the driver's seat of CARs. Med Res Rev 2022; 42:306-342. [PMID: 34028069 PMCID: PMC9292017 DOI: 10.1002/med.21818] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 02/17/2021] [Accepted: 04/21/2021] [Indexed: 12/16/2022]
Abstract
Immuno-oncology has been at the forefront of cancer treatment in recent decades. In particular immune checkpoint and chimeric antigen receptor (CAR)-T cell therapy have achieved spectacular results. Over the years, CAR-T cell development has followed a steady evolutionary path, focusing on increasing T cell potency and sustainability, which has given rise to different CAR generations. However, there was less focus on the mode of interaction between the CAR-T cell and the cancer cell; more specifically on the targeting moiety used in the CAR and its specific properties. Recently, the importance of optimizing this domain has been recognized and the possibilities have been exploited. Over the last 10 years-in addition to the classical scFv-based CARs-single domain CARs, natural receptor-ligand CARs, universal CARs and CARs targeting more than one antigen have emerged. In addition, the specific parameters of the targeting domain and their influence on T cell activation are being examined. In this review, we concisely present the history of CAR-T cell therapy, and then expand on various developments in the CAR ectodomain. We discuss different formats, each with their own advantages and disadvantages, as well as the developments in affinity tuning, avidity effects, epitope location, and influence of the extracellular spacer.
Collapse
Affiliation(s)
- Heleen Hanssens
- In Vivo Cellular and Molecular Imaging LaboratoryVrije Universiteit BrusselBrusselsBelgium
- Laboratory of Hematology and ImmunologyVrije Universiteit BrusselBrusselsBelgium
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical SciencesVrije Universiteit BrusselBrusselsBelgium
| | - Fien Meeus
- In Vivo Cellular and Molecular Imaging LaboratoryVrije Universiteit BrusselBrusselsBelgium
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical SciencesVrije Universiteit BrusselBrusselsBelgium
| | - Kim De Veirman
- Laboratory of Hematology and ImmunologyVrije Universiteit BrusselBrusselsBelgium
| | - Karine Breckpot
- Laboratory for Molecular and Cellular Therapy, Department of Biomedical SciencesVrije Universiteit BrusselBrusselsBelgium
| | - Nick Devoogdt
- In Vivo Cellular and Molecular Imaging LaboratoryVrije Universiteit BrusselBrusselsBelgium
| |
Collapse
|
31
|
Current Limitations and Perspectives of Chimeric Antigen Receptor-T-Cells in Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:cancers13246157. [PMID: 34944782 PMCID: PMC8699597 DOI: 10.3390/cancers13246157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/22/2021] [Accepted: 11/25/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Acute myeloid leukemia (AML) is the most frequent type of acute leukemia in adults. Allogeneic hematopoietic cell transplantation (allo-HCT) has been the only potentially curative treatment for the majority of patients. The ability of chimeric antigen receptor (CAR)-modified T-cell therapy directed against the CD19 antigen to induce durable remissions in patients with acute lymphoblastic leukemia (ALL) has provided optimism that this novel treatment paradigm can be extrapolated to AML. In this review, we provide an overview of candidate target antigens for CAR-T-cells in AML, an update on recent progress in preclinical and clinical development of investigational CAR-T-cell products, and discuss challenges for the clinical implementation of CAR-T-cell therapy in AML. Abstract Adoptive transfer of gene-engineered chimeric antigen receptor (CAR)-T-cells has emerged as a powerful immunotherapy for combating hematologic cancers. Several target antigens that are prevalently expressed on AML cells have undergone evaluation in preclinical CAR-T-cell testing. Attributes of an ‘ideal’ target antigen for CAR-T-cell therapy in AML include high-level expression on leukemic blasts and leukemic stem cells (LSCs), and absence on healthy tissues, normal hematopoietic stem and progenitor cells (HSPCs). In contrast to other blood cancer types, where CAR-T therapies are being similarly studied, only a rather small number of AML patients has received CAR-T-cell treatment in clinical trials, resulting in limited clinical experience for this therapeutic approach in AML. For curative AML treatment, abrogation of bulk blasts and LSCs is mandatory with the need for hematopoietic recovery after CAR-T administration. Herein, we provide a critical review of the current pipeline of candidate target antigens and corresponding CAR-T-cell products in AML, assess challenges for clinical translation and implementation in routine clinical practice, as well as perspectives for overcoming them.
Collapse
|
32
|
Ghamari A, Pakzad P, Majd A, Ebrahimi M, Hamidieh AA. Design and Production An Effective Bispecific Tandem Chimeric Antigen Receptor on T Cells against CD123 and Folate Receptor ß towards B-Acute Myeloid Leukaemia Blasts. CELL JOURNAL 2021; 23:650-657. [PMID: 34939758 PMCID: PMC8665988 DOI: 10.22074/cellj.2021.7314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 01/21/2020] [Indexed: 11/21/2022]
Abstract
Objective The clinical studies of acute myeloid leukaemia (AML) revealed that antigen escaping variants cause cancer
recurrence even after treatment with chimeric antigen receptor (CAR)-T cells that target a single tumour antigen. Due to
the heterogeneous expression of antigens on leukaemia blasts, we hypothesized that a novel bispecific CAR, directed
to the folate receptor beta (FRβ)-binding single-chain variable fragment (scFv) and an IL3α-binding receptor (CD123)
that has more expression in AML blasts, can decrease CAR-T cell exhaustion and increase the efficacy of CAR-T cells
to prevent antigen escaping and consequent recurrence of AML.
Materials and Methods In this experimental study, the survival, proliferation, and cytolysis of CAR-T cells remains
suboptimal even with a costimulatory endodomain. Hence, we designed and constructed a tandem CAR that joins an
FRβ and CD123 in the second generation retroviral vector to generate a bispecific tandem CAR (TanCAR-T cell).
Results TanCAR FRβ-CD123 T cells showed distinct binding to FRβ or CD123 expressing cells. They could lyse the
leukaemia cell lines (66.1 ± 11%) comparable to the single CAR-T cells against these determinants. TanCAR FRβ-
CD123 T cells simultaneously engaged FRβ and CD123, which promoted T cell activation in targeting and lysis of the
examined leukaemia cell lines. TanCAR-T cell significantly induced interferon gamma (IFNγ) and interleukin 2 (IL-2)
production more than single CAR-T cells, which produced a synergistic enhancement of TanCAR FRβ-CD123 T cell
function when dual antigens faced simultaneously.
Conclusion Dual-specific TanCAR FRβ-CD123 T cells showed therapeutic potential to improve AML control by co-
engaging FRβ and CD123 molecules in a robust, divalent immune system. This strategy may be a useful therapeutic
approach in patients with relapsed B-cell malignancies.
Collapse
Affiliation(s)
- Ali Ghamari
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Parviz Pakzad
- Department of Microbiology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ahmad Majd
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Marzieh Ebrahimi
- Department of Stem Cells and Developmental Biology, Cell Science Research Centre, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Amir Ali Hamidieh
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell and Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
33
|
Maiorova V, Mollaev MD, Vikhreva P, Kulakovskaya E, Pershin D, Chudakov DM, Kibardin A, Maschan MA, Larin S. Natural Flt3Lg-Based Chimeric Antigen Receptor (Flt3-CAR) T Cells Successfully Target Flt3 on AML Cell Lines. Vaccines (Basel) 2021; 9:vaccines9111238. [PMID: 34835169 PMCID: PMC8621097 DOI: 10.3390/vaccines9111238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 10/01/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Relapsed/refractory acute myeloid leukemia (AML) cannot be cured with chemotherapy alone, as the blasts survive the treatment. Chimeric antigen receptor (CAR) approaches for AML are being actively developed. CARs promote immune reactions through recognition of the target molecular epitopes at the surface of cancer cells. The recognition involves the extracellular portion of the CAR protein, which corresponds to either the antibody or the physiological binding partner of the targeted antigen. Here, we design a chimeric receptor with a full-length natural Flt3-ligand recognition module that targets Flt3 tyrosine kinase, known as an adverse marker in AML. We demonstrate specific killing of Flt3-positive THP-1 cells by Flt3-CAR T cells and the lack of cytotoxicity towards Flt3-negative U937 cells. We prove that the inherent cytolytic capacity of T cells is essential for the killing. Finally, we confirm the authenticity of targeting by its competitive dose-dependent inhibition with a soluble Flt3-ligand. The developed system can be viewed as a non-immunogenic functional equivalent of scFv-mediated targeting. The robust in vitro antitumor effects of Flt3-CAR T cells, combined with their low off-target cytotoxicity, hold promise for AML treatment.
Collapse
Affiliation(s)
- Varvara Maiorova
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia;
- Correspondence:
| | - Murad D. Mollaev
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| | - Polina Vikhreva
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| | - Elena Kulakovskaya
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| | - Dmitry Pershin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| | - Dmitriy M. Chudakov
- Center of Life Sciences, Skolkovo Institute of Science and Technology, 121205 Moscow, Russia;
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia
| | - Alexey Kibardin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| | - Michael A. Maschan
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| | - Sergey Larin
- Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, 117997 Moscow, Russia; (M.D.M.); (P.V.); (E.K.); (D.P.); (A.K.); (M.A.M.); (S.L.)
| |
Collapse
|
34
|
Shademan B, Karamad V, Nourazarian A, Avcı CB. CAR T Cells: Cancer Cell Surface Receptors Are the Target for Cancer Therapy. Adv Pharm Bull 2021; 12:476-489. [PMID: 35935042 PMCID: PMC9348524 DOI: 10.34172/apb.2022.051] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 06/12/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022] Open
Abstract
Immunotherapy has become a prominent strategy for the treatment of cancer. A method that improves the immune system's ability to attack a tumor (Enhances antigen binding). Targeted killing of malignant cells by adoptive transfer of chimeric antigen receptor (CAR) T cells is a promising immunotherapy technique in the treatment of cancers. For this purpose, the patient's immune cells, with genetic engineering aid, are loaded with chimeric receptors that have particular antigen binding and activate cytotoxic T lymphocytes. That increases the effectiveness of immune cells and destroying cancer cells. This review discusses the basic structure and function of CAR-T cells and how antigenic targets are identified to treat different cancers and address the disadvantages of this treatment for cancer.
Collapse
Affiliation(s)
- Behrouz Shademan
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Vahidreza Karamad
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| | - Alireza Nourazarian
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Cigir Biray Avcı
- Department of Medical Biology, Faculty of Medicine, EGE University, Izmir, Turkey
| |
Collapse
|
35
|
Watanabe K, Nishikawa H. Engineering strategies for broad application of TCR-T and CAR-T cell therapies. Int Immunol 2021; 33:551-562. [PMID: 34374779 DOI: 10.1093/intimm/dxab052] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/09/2021] [Indexed: 12/19/2022] Open
Abstract
Adoptive cell therapy, including the transfer of tumor-infiltrating T lymphocytes after in vitro expansion or T cells redirected to tumor antigens using antigen-specific transgenic T cell receptor T cells (TCR-T cells) or chimeric antigen receptor T cells (CAR-T cells), has shown a significant clinical impact. Particularly, several types of CAR-T cell therapies have been approved for the treatment of hematological malignancies. The striking success of CAR-T cell therapies in hematological malignancies motivates their further expansion to a wide range of solid tumors, yet multiple obstacles, including the lack of proper target antigens exhibiting a tumor-specific expression pattern and the immunosuppressive tumor microenvironment (TME) impairing the effector functions of adoptively transferred T cells, have prevented clinical application. Gene engineering technologies such as the CRISPR/Cas9 system have enabled flexible reprograming of TCR/CAR-T cell signaling or loading genes that are targets of the tumor immunosuppression as a payload to overcome the difficulties. Here, we discuss recent advances in TCR/CAR-T cell engineering: various promising approaches to enhance the antitumor activity of adoptively transferred T cells in the TME for maximizing the efficacy and the safety of adoptive cell therapy are now being tested in the clinic, especially targeting solid tumors.
Collapse
Affiliation(s)
- Keisuke Watanabe
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan
| | - Hiroyoshi Nishikawa
- Division of Cancer Immunology, Research Institute, National Cancer Center, Tokyo 104-0045, Japan.,Department of Immunology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| |
Collapse
|
36
|
Sauer T, Parikh K, Sharma S, Omer B, Sedloev D, Chen Q, Angenendt L, Schliemann C, Schmitt M, Müller-Tidow C, Gottschalk S, Rooney CM. CD70-specific CAR T cells have potent activity against acute myeloid leukemia without HSC toxicity. Blood 2021; 138:318-330. [PMID: 34323938 PMCID: PMC8323977 DOI: 10.1182/blood.2020008221] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/05/2021] [Indexed: 02/06/2023] Open
Abstract
The prognosis of patients with acute myeloid leukemia (AML) remains dismal, highlighting the need for novel innovative treatment strategies. The application of chimeric antigen receptor (CAR) T-cell therapy to patients with AML has been limited, in particular by the lack of a tumor-specific target antigen. CD70 is a promising antigen to target AML, as it is expressed on most leukemic blasts, whereas little or no expression is detectable in normal bone marrow samples. To target CD70 on AML cells, we generated a panel of CD70-CAR T cells that contained a common single-chain variable fragment (scFv) for antigen detection, but differed in size and flexibility of the extracellular spacer and in the transmembrane and the costimulatory domains. These CD70scFv CAR T cells were compared with a CAR construct that contained human CD27, the ligand of CD70 fused to the CD3ζ chain (CD27z). The structural composition of the CAR strongly influenced expression levels, viability, expansion, and cytotoxic capacities of CD70scFv-based CAR T cells, but CD27z-CAR T cells demonstrated superior proliferation and antitumor activity in vitro and in vivo, compared with all CD70scFv-CAR T cells. Although CD70-CAR T cells recognized activated virus-specific T cells (VSTs) that expressed CD70, they did not prevent colony formation by normal hematopoietic stem cells. Thus, CD70-targeted immunotherapy is a promising new treatment strategy for patients with CD70-positive AML that does not affect normal hematopoiesis but will require monitoring of virus-specific T-cell responses.
Collapse
Affiliation(s)
- Tim Sauer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Kathan Parikh
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| | - Bilal Omer
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| | - David Sedloev
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Qian Chen
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Linus Angenendt
- Department of Internal Medicine A, University Hospital of Muenster, Muenster, Germany; and
| | - Christoph Schliemann
- Department of Internal Medicine A, University Hospital of Muenster, Muenster, Germany; and
| | - Michael Schmitt
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephen Gottschalk
- Department of Bone Marrow Transplantation and Cellular Therapy, St Jude Children's Research Hospital, Memphis, TN
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston Methodist Hospital-Texas Children's Hospital, Houston, TX
| |
Collapse
|
37
|
Siglec-6 is a novel target for CAR T-cell therapy in acute myeloid leukemia (AML). Blood 2021; 138:1830-1842. [PMID: 34289026 PMCID: PMC9642786 DOI: 10.1182/blood.2020009192] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 06/26/2021] [Indexed: 11/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is attractive for the development of CAR T-cell immunotherapy because AML blasts are susceptible to T-cell-mediated elimination. Here, we introduce sialic-acid-binding immunoglobulin-like lectin (Siglec)-6 as a novel target for CAR T-cells in AML. We designed a Siglec-6-specific CAR with a targeting-domain derived from a human monoclonal antibody JML‑1. We found that Siglec-6 is prevalently expressed on AML cell lines and primary AML blasts, including the subpopulation of AML stem cells. Treatment with Siglec-6-CAR T-cells confers specific anti-leukemia reactivity that correlates with Siglec-6-expression in pre-clinical models, including induction of complete remission in a xenograft AML model in immunodeficient mice (NSG/U937). In addition, we confirmed Siglec-6-expression on transformed B-cells in chronic lymphocytic leukemia (CLL) and show specific anti-CLL-reactivity of Siglec-6-CAR T-cells in vitro. Of particular interest, we found that Siglec-6 is not detectable on normal hematopoietic stem and progenitor cells (HSC/P) and that treatment with Siglec-6-CAR T-cells does not affect their viability and lineage differentiation in colony-formation assays. These data suggest that Siglec-6-CAR T-cell therapy may be used to effectively treat AML without a need for subsequent allogeneic hematopoietic stem cell transplantation. In mature normal hematopoietic cells, we detected Siglec-6 in a proportion of memory (and naïve) B-cells and basophilic granulocytes, suggesting the potential for limited on-target/off-tumor reactivity. The lacking expression of Siglec-6 on normal HSC/P is a key differentiator from other Siglec-family members (e.g. Siglec-3=CD33) and other CAR target antigens, e.g. CD123, that are under investigation in AML and warrants the clinical investigation of Siglec-6-CAR T-cell therapy.
Collapse
|
38
|
Improved Activity against Acute Myeloid Leukemia with Chimeric Antigen Receptor (CAR)-NK-92 Cells Designed to Target CD123. Viruses 2021; 13:v13071365. [PMID: 34372571 PMCID: PMC8310147 DOI: 10.3390/v13071365] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 01/08/2023] Open
Abstract
Anti-cancer activity can be improved by engineering immune cells to express chimeric antigen receptors (CARs) that recognize tumor-associated antigens. Retroviral vector gene transfer strategies allow stable and durable transgene expression. Here, we used alpharetroviral vectors to modify NK-92 cells, a natural killer cell line, with a third-generation CAR designed to target the IL-3 receptor subunit alpha (CD123), which is strongly expressed on the surface of acute myeloid leukemia (AML) cells. Alpharetroviral vectors also contained a transgene cassette to allow constitutive expression of human IL-15 for increased NK cell persistence in vivo. The anti-AML activity of CAR-NK-92 cells was tested via in vitro cytotoxicity assays with the CD123+ AML cell line KG-1a and in vivo in a patient-derived xenotransplantation CD123+ AML model. Unmodified NK-92 cells or NK-92 cells modified with a truncated version of the CAR that lacked the signaling domain served as controls. Alpharetroviral vector-modified NK-92 cells stably expressed the transgenes and secreted IL-15. Anti-CD123-CAR-NK-92 cells exhibited enhanced anti-AML activity in vitro and in vivo as compared to control NK-92 cells. Our data (1) shows the importance of IL-15 expression for in vivo persistence of NK-92 cells, (2) supports continued investigation of anti-CD123-CAR-NK cells to target AML, and (3) points towards potential strategies to further improve CAR-NK anti-AML activity.
Collapse
|
39
|
Masoumi J, Jafarzadeh A, Abdolalizadeh J, Khan H, Philippe J, Mirzaei H, Mirzaei HR. Cancer stem cell-targeted chimeric antigen receptor (CAR)-T cell therapy: Challenges and prospects. Acta Pharm Sin B 2021; 11:1721-1739. [PMID: 34386318 PMCID: PMC8343118 DOI: 10.1016/j.apsb.2020.12.015] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/03/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023] Open
Abstract
Cancer stem cells (CSCs) with their self-renewal ability are accepted as cells which initiate tumors. CSCs are regarded as interesting targets for novel anticancer therapeutic agents because of their association with tumor recurrence and resistance to conventional therapies, including radiotherapy and chemotherapy. Chimeric antigen receptor (CAR)-T cells are engineered T cells which express an artificial receptor specific for tumor associated antigens (TAAs) by which they accurately target and kill cancer cells. In recent years, CAR-T cell therapy has shown more efficiency in cancer treatment, particularly regarding blood cancers. The expression of specific markers such as TAAs on CSCs in varied cancer types makes them as potent tools for CAR-T cell therapy. Here we review the CSC markers that have been previously targeted with CAR-T cells, as well as the CSC markers that may be used as possible targets for CAR-T cell therapy in the future. Furthermore, we will detail the most important obstacles against CAR-T cell therapy and suggest solutions.
Collapse
Affiliation(s)
- Javad Masoumi
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan 77181759111, Iran
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran
| | - Jalal Abdolalizadeh
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Jeandet Philippe
- Research Unit “Induced Resistance and Plant Bioprotection”, EA 4707, SFR Condorcet FR CNRS 3417, Faculty of Sciences University of Reims Champagne-Ardenne, BP 1039, 51687, Reims Cedex 2, France
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan 8713781147, Iran
- Corresponding authors. Tel./fax: +98 31 55540022; Tel./fax: +98 21 66419536.
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Corresponding authors. Tel./fax: +98 31 55540022; Tel./fax: +98 21 66419536.
| |
Collapse
|
40
|
Michelozzi IM, Kirtsios E, Giustacchini A. Driving CAR T Stem Cell Targeting in Acute Myeloid Leukemia: The Roads to Success. Cancers (Basel) 2021; 13:2816. [PMID: 34198742 PMCID: PMC8201025 DOI: 10.3390/cancers13112816] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 12/14/2022] Open
Abstract
Current treatment outcome for acute myeloid leukemia (AML) patients is unsatisfactory and characterized by high rates of relapse and poor overall survival. Increasing evidence points to a crucial role of leukemic stem cells (LSC) and the bone marrow (BM) leukemic niche, in which they reside, in AML evolution and chemoresistance. Thus, future strategies aiming at improving AML therapeutic protocols are likely to be directed against LSC and their niche. Chimeric antigen receptor (CAR) T-cells have been extremely successful in the treatment of relapsed/refractory acute lymphoblastic leukemia and B-cell non-Hodgkin lymphoma and comparable results in AML are highly desirable. At present, we are at the dawn of CAR T-cell application in AML, with several preclinical studies and few early phase clinical trials. However, the lack of leukemia-specific targets and the genetic and phenotypic heterogeneity of the disease combined with the leukemia-induced remodeling of the BM microenvironment are limiting CAR T-cell exploitation in AML. Here, we reviewed AML-LSC and AML-BM niche features in the context of their therapeutic targeting using CAR T-cells. We summarized recent progress in CAR T-cell application to the treatment of AML, and we discussed the remaining therapeutic challenges and promising novel strategies to overcome them.
Collapse
Affiliation(s)
- Ilaria M. Michelozzi
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London WC1N 1DZ, UK;
| | | | - Alice Giustacchini
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, Zayed Centre for Research into Rare Disease in Children, London WC1N 1DZ, UK;
| |
Collapse
|
41
|
Roy AG, Robinson JM, Sharma P, Rodriguez-Garcia A, Poussin MA, Nickerson-Nutter C, Powell DJ. Folate Receptor Beta as a Direct and Indirect Target for Antibody-Based Cancer Immunotherapy. Int J Mol Sci 2021; 22:5572. [PMID: 34070369 PMCID: PMC8197521 DOI: 10.3390/ijms22115572] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/12/2021] [Accepted: 05/15/2021] [Indexed: 01/08/2023] Open
Abstract
Folate receptor beta (FRβ) is a folate binding receptor expressed on myeloid lineage hematopoietic cells. FRβ is commonly expressed at high levels on malignant blasts in patients with acute myeloid leukemia (AML), as well as on M2 polarized tumor-associated macrophages (TAMs) in the tumor microenvironment of many solid tumors. Therefore, FRβ is a potential target for both direct and indirect cancer therapy. We demonstrate that FRβ is expressed in both AML cell lines and patient-derived AML samples and that a high-affinity monoclonal antibody against FRβ (m909) has the ability to cause dose- and expression-dependent ADCC against these cells in vitro. Importantly, we find that administration of m909 has a significant impact on tumor growth in a humanized mouse model of AML. Surprisingly, m909 functions in vivo with and without the infusion of human NK cells as mediators of ADCC, suggesting potential involvement of mouse macrophages as effector cells. We also found that TAMs from primary ovarian ascites samples expressed appreciable levels of FRβ and that m909 has the ability to cause ADCC in these samples. These results indicate that the targeting of FRβ using m909 has the potential to limit the outgrowth of AML in vitro and in vivo. Additionally, m909 causes cytotoxicity to TAMs in the tumor microenvironment of ovarian cancer warranting further investigation of m909 and its derivatives as therapeutic agents in patients with FRβ-expressing cancers.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/pharmacology
- CHO Cells
- Cricetulus
- Female
- Folate Receptor 2/antagonists & inhibitors
- Folate Receptor 2/immunology
- HL-60 Cells
- Humans
- Immunotherapy
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/therapy
- Mice
- Mice, Inbred NOD
- Mice, Knockout
- Mice, SCID
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/immunology
- Ovarian Neoplasms/immunology
- Ovarian Neoplasms/pathology
- Ovarian Neoplasms/therapy
- THP-1 Cells
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Allison G. Roy
- Ovarian Cancer Research Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (P.S.); (A.R.-G.); (M.A.P.)
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - J. Michael Robinson
- Department of Gynecologic Oncology, Zimmer Cancer Center, New Hanover Regional Medical Center, Wilmington, NC 28401, USA;
| | - Prannda Sharma
- Ovarian Cancer Research Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (P.S.); (A.R.-G.); (M.A.P.)
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alba Rodriguez-Garcia
- Ovarian Cancer Research Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (P.S.); (A.R.-G.); (M.A.P.)
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mathilde A. Poussin
- Ovarian Cancer Research Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (P.S.); (A.R.-G.); (M.A.P.)
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Daniel J. Powell
- Ovarian Cancer Research Center, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; (A.G.R.); (P.S.); (A.R.-G.); (M.A.P.)
- Center for Cellular Immunotherapies, Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
42
|
Lee JB, Vasic D, Kang H, Fang KKL, Zhang L. State-of-Art of Cellular Therapy for Acute Leukemia. Int J Mol Sci 2021; 22:ijms22094590. [PMID: 33925571 PMCID: PMC8123829 DOI: 10.3390/ijms22094590] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 12/13/2022] Open
Abstract
With recent clinical breakthroughs, immunotherapy has become the fourth pillar of cancer treatment. Particularly, immune cell-based therapies have been envisioned as a promising treatment option with curative potential for leukemia patients. Hence, an increasing number of preclinical and clinical studies focus on various approaches of immune cell-based therapy for treatment of acute leukemia (AL). However, the use of different immune cell lineages and subsets against different types of leukemia and patient disease statuses challenge the interpretation of the clinical applicability and outcome of immune cell-based therapies. This review aims to provide an overview on recent approaches using various immune cell-based therapies against acute B-, T-, and myeloid leukemias. Further, the apparent limitations observed and potential approaches to overcome these limitations are discussed.
Collapse
MESH Headings
- Acute Disease
- Cell- and Tissue-Based Therapy
- Humans
- Immunotherapy
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Killer Cells, Natural/immunology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, T-Cell/metabolism
- Leukemia, T-Cell/therapy
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Jong-Bok Lee
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (J.-B.L.); (D.V.); (H.K.); (K.K.-L.F.)
| | - Daniel Vasic
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (J.-B.L.); (D.V.); (H.K.); (K.K.-L.F.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Hyeonjeong Kang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (J.-B.L.); (D.V.); (H.K.); (K.K.-L.F.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Karen Kai-Lin Fang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (J.-B.L.); (D.V.); (H.K.); (K.K.-L.F.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Li Zhang
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada; (J.-B.L.); (D.V.); (H.K.); (K.K.-L.F.)
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Department of Immunology, University of Toronto, Toronto, ON M5S 1A8, Canada
- Correspondence:
| |
Collapse
|
43
|
CAR-T cell-mediated depletion of immunosuppressive tumor-associated macrophages promotes endogenous antitumor immunity and augments adoptive immunotherapy. Nat Commun 2021; 12:877. [PMID: 33563975 PMCID: PMC7873057 DOI: 10.1038/s41467-021-20893-2] [Citation(s) in RCA: 155] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
The immunosuppressive tumor microenvironment (TME) represents a major barrier for effective immunotherapy. Tumor-associated macrophages (TAMs) are highly heterogeneous and plastic cell components of the TME which can either promote tumor progression (M2-like) or boost antitumor immunity (M1-like). Here, we demonstrate that a subset of TAMs that express folate receptor β (FRβ) possess an immunosuppressive M2-like profile. In syngeneic tumor mouse models, chimeric antigen receptor (CAR)-T cell-mediated selective elimination of FRβ+ TAMs in the TME results in an enrichment of pro-inflammatory monocytes, an influx of endogenous tumor-specific CD8+ T cells, delayed tumor progression, and prolonged survival. Preconditioning of the TME with FRβ-specific CAR-T cells also improves the effectiveness of tumor-directed anti-mesothelin CAR-T cells, while simultaneous co-administration of both CAR products does not. These results highlight the pro-tumor role of FRβ+ TAMs in the TME and the therapeutic implications of TAM-depleting agents as preparative adjuncts to conventional immunotherapies that directly target tumor antigens.
Collapse
|
44
|
Acharya UH, Walter RB. Chimeric Antigen Receptor (CAR)-Modified Immune Effector Cell Therapy for Acute Myeloid Leukemia (AML). Cancers (Basel) 2020; 12:E3617. [PMID: 33287224 PMCID: PMC7761730 DOI: 10.3390/cancers12123617] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 11/28/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Despite the availability of an increasing number of targeted therapeutics and wider use of allogeneic hematopoietic stem cell transplantation, many patients with acute myeloid leukemia (AML) ultimately succumb to this disease. Given their remarkable efficacy in B-acute lymphoblastic leukemia and other CD19-expressing B cell malignancies, there is hope adoptive cellular transfer, particularly chimeric antigen receptor (CAR)-modified immune effector cell (IEC) therapies, may afford a novel, potent immune-based approach for the treatment of AML that complements or replaces existing ones and improves cure rates. However, it is unclear how best to translate the success of these therapies from B cell malignancies, where use of highly potent immunotherapies is facilitated by identified target antigens with near ubiquitous expression on malignant cells and non-fatal consequences from "on-target, off-tumor cell" toxicities. Herein, we review the current status of CAR-modified IEC therapies for AML, with considerations regarding suitable, relatively leukemia-restricted target antigens, expected toxicities, and interactions of the engineered cells with a profoundly immunosuppressive tumor microenvironment that restricts their therapeutic efficacy. With these challenges in mind, we will discuss possible strategies to improve the cells' potency as well as their therapeutic window for optimal clinical use in AML.
Collapse
Affiliation(s)
- Utkarsh H. Acharya
- Divisions of Hematologic Malignancies & Immune Effector Cell Therapy, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02215, USA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA;
- Department of Medicine, Division of Hematology, University of Washington, Seattle, WA 98195, USA
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA 98195, USA
- Department of Epidemiology, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
45
|
Chimeric antigen receptor T cell therapies for acute myeloid leukemia. Front Med 2020; 14:701-710. [PMID: 33263835 DOI: 10.1007/s11684-020-0763-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 02/16/2020] [Indexed: 12/12/2022]
Abstract
Chimeric antigen receptor T cell (CAR T) therapies have achieved unprecedented efficacy in B-cell tumors, prompting scientists and doctors to exploit this strategy to treat other tumor types. Acute myeloid leukemia (AML) is a group of heterogeneous myeloid malignancies. Relapse remains the main cause of treatment failure, especially for patients with intermediate or high risk stratification. Allogeneic hematopoietic stem cell transplantation could be an effective therapy because of the graft-versus-leukemia effect, which unfortunately puts the patient at risk of serious complications, such as graft-versus-host disease. Although the identification of an ideal target antigen for AML is challenging, CAR T therapy remains a highly promising strategy for AML patients, particularly for those who are ineligible to receive a transplantation or have positive minimal residual disease. In this review, we focus on the most recent and promising advances in CAR T therapies for AML.
Collapse
|
46
|
Shi W, Jin W, Xia L, Hu Y. Novel agents targeting leukemia cells and immune microenvironment for prevention and treatment of relapse of acute myeloid leukemia after allogeneic hematopoietic stem cell transplantation. Acta Pharm Sin B 2020; 10:2125-2139. [PMID: 32837873 PMCID: PMC7326461 DOI: 10.1016/j.apsb.2020.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/27/2020] [Accepted: 06/05/2020] [Indexed: 12/20/2022] Open
Abstract
Relapse remains the worst life-threatening complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with acute myeloid leukemia (AML), whose prognosis has been historically dismal. Given the rapid development of genomics and immunotherapies, the interference strategies for AML recurrence have been changing these years. More and more novel targeting agents that have received the U.S. Food and Drug Administration (FDA) approval for de novo AML treatment have been administrated in the salvage or maintenance therapy of post-HSCT relapse. Targeted strategies that regulate the immune microenvironment of and optimize the graft versus leukemia (GVL) effect of immune cells are gradually improved. Such agents not only have been proven to achieve clinical benefits from a single drug, but if combined with classic therapies, can significantly improve the poor prognosis of AML patients who relapse after allo-HSCT. This review will focus on currently available and promising upcoming agents and also discuss the challenges and limitations of targeted therapies in the allogeneic hematopoietic stem cell transplantation community.
Collapse
Affiliation(s)
- Wei Shi
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109-1024, USA
| | - Weiwei Jin
- Department of Cardiovascular, Optical Valley School District, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan 430061, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan 430074, China
| | - Linghui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
47
|
Watanabe N, McKenna MK, Rosewell Shaw A, Suzuki M. Clinical CAR-T Cell and Oncolytic Virotherapy for Cancer Treatment. Mol Ther 2020; 29:505-520. [PMID: 33130314 DOI: 10.1016/j.ymthe.2020.10.023] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 09/30/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023] Open
Abstract
Immunotherapy has recently garnered success with the induction of clinical responses in tumors, which are traditionally associated with poor outcomes. Chimeric antigen receptor T (CAR-T) cells and oncolytic viruses (OVs) have emerged as promising cancer immunotherapy agents. Herein, we provide an overview of the current clinical status of CAR-T cell and OV therapies. While preclinical studies have demonstrated curative potential, the benefit of CAR-T cells and OVs as single-agent treatments remains limited to a subset of patients. Combinations of different targeted therapies may be required to achieve efficient, durable responses against heterogeneous tumors, as well as the microenvironment. Using a combinatorial approach to take advantage of the unique features of CAR-T cells and OVs with other treatments can produce additive therapeutic effects. This review also discusses ongoing clinical evaluations of these combination strategies for improved outcomes in treatment of resistant malignancies.
Collapse
Affiliation(s)
- Norihiro Watanabe
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Mary Kathryn McKenna
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Amanda Rosewell Shaw
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Masataka Suzuki
- Department of Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Texas Children's Hospital, Houston Methodist Hospital, Houston, TX 77030, USA.
| |
Collapse
|
48
|
Jayaraman J, Mellody MP, Hou AJ, Desai RP, Fung AW, Pham AHT, Chen YY, Zhao W. CAR-T design: Elements and their synergistic function. EBioMedicine 2020; 58:102931. [PMID: 32739874 PMCID: PMC7393540 DOI: 10.1016/j.ebiom.2020.102931] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/30/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells use re-engineered cell surface receptors to specifically bind to and lyse oncogenic cells. Two clinically approved CAR-T–cell therapies have significant clinical efficacy in treating CD19-positive B cell cancers. With widespread interest to deploy this immunotherapy to other cancers, there has been great research activity to design new CAR structures to increase the range of targeted cancers and anti-tumor efficacy. However, several obstacles must be addressed before CAR-T–cell therapies can be more widely deployed. These include limiting the frequency of lethal cytokine storms, enhancing T-cell persistence and signaling, and improving target antigen specificity. We provide a comprehensive review of recent research on CAR design and systematically evaluate design aspects of the four major modules of CAR structure: the ligand-binding, spacer, transmembrane, and cytoplasmic domains, elucidating design strategies and principles to guide future immunotherapeutic discovery.
Collapse
Affiliation(s)
- Jayapriya Jayaraman
- Department of Biomedical Engineering, University of California, Irvine, Irvine,CA,92697, United States
| | - Michael P Mellody
- Department of Biomedical Engineering, University of California, Irvine, Irvine,CA,92697, United States
| | - Andrew J Hou
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095
| | - Ruchi P Desai
- School of Medicine, University of California, Irvine, Irvine, CA, 92697
| | - Audrey W Fung
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, 92697
| | - An Huynh Thuy Pham
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA, 92697
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, Los Angeles, CA, 90095; Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, 90095; Parker Institute for Cancer Immunotherapy Center, University of California, Los Angeles, Los Angeles, Los Angeles, 90095
| | - Weian Zhao
- Department of Biomedical Engineering, University of California, Irvine, Irvine,CA,92697, United States; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, United States; Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, United States; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, CA 92697, United States; Department of Biological Chemistry, University of California, Irvine, Irvine, CA 92697, United States.
| |
Collapse
|
49
|
Zhylko A, Winiarska M, Graczyk-Jarzynka A. The Great War of Today: Modifications of CAR-T Cells to Effectively Combat Malignancies. Cancers (Basel) 2020; 12:E2030. [PMID: 32722109 PMCID: PMC7466082 DOI: 10.3390/cancers12082030] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/17/2020] [Accepted: 07/21/2020] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy of cancer had its early beginnings in the times when the elements of the immune system were still poorly characterized. However, with the progress in molecular biology, it has become feasible to re-engineer T cells in order to eradicate tumour cells. The use of synthetic chimeric antigen receptors (CARs) helped to re-target and simultaneously unleash the cytotoxic potential of T cells. CAR-T therapy proved to be remarkably effective in cases of haematological malignancies, often refractory and relapsed. The success of this approach yielded two Food and Drug Administration (FDA) approvals for the first "living drug" modalities. However, CAR-T therapy is not without flaws. Apart from the side effects associated with the treatment, it became apparent that CAR introduction alters T cell biology and the possible therapeutic outcomes. Additionally, it was shown that CAR-T approaches in solid tumours do not recapitulate the success in the haemato-oncology. Therefore, in this review, we aim to discuss the recent concerns of CAR-T therapy for both haematological and solid tumours. We also summarise the general strategies that are implemented to enhance the efficacy and safety of the CAR-T regimens in blood and solid malignancies.
Collapse
|
50
|
Sharma P, Marada VVVR, Cai Q, Kizerwetter M, He Y, Wolf SP, Schreiber K, Clausen H, Schreiber H, Kranz DM. Structure-guided engineering of the affinity and specificity of CARs against Tn-glycopeptides. Proc Natl Acad Sci U S A 2020; 117:15148-15159. [PMID: 32541028 PMCID: PMC7334454 DOI: 10.1073/pnas.1920662117] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The potency of adoptive T cell therapies targeting the cell surface antigen CD19 has been demonstrated in hematopoietic cancers. It has been difficult to identify appropriate targets in nonhematopoietic tumors, but one class of antigens that have shown promise is aberrant O-glycoprotein epitopes. It has long been known that dysregulated synthesis of O-linked (threonine or serine) sugars occurs in many cancers, and that this can lead to the expression of cell surface proteins containing O-glycans comprised of a single N-acetylgalactosamine (GalNAc, known as Tn antigen) rather than the normally extended carbohydrate. Previously, we used the scFv fragment of antibody 237 as a chimeric antigen receptor (CAR) to mediate recognition of mouse tumor cells that bear its cognate Tn-glycopeptide epitope in podoplanin, also called OTS8. Guided by the structure of the 237 Fab:Tn-OTS8-glycopeptide complex, here we conducted a deep mutational scan showing that residues flanking the Tn-glycan contributed significant binding energy to the interaction. Design of 237-scFv libraries in the yeast display system allowed us to isolate scFv variants with higher affinity for Tn-OTS8. Selection with a noncognate human antigen, Tn-MUC1, yielded scFv variants that were broadly reactive with multiple Tn-glycoproteins. When configured as CARs, engineered T cells expressing these scFv variants showed improved activity against mouse and human cancer cell lines defective in O-linked glycosylation. This strategy provides CARs with Tn-peptide specificities, all based on a single scFv scaffold, that allows the same CAR to be tested for toxicity in mice and efficacy against mouse and human tumors.
Collapse
Affiliation(s)
- Preeti Sharma
- Department of Biochemistry, Cancer Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801;
| | - Venkata V V R Marada
- Department of Biochemistry, Cancer Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Qi Cai
- Department of Biochemistry, Cancer Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Monika Kizerwetter
- Department of Biochemistry, Cancer Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801
| | - Yanran He
- Department of Pathology, Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Steven P Wolf
- Department of Pathology, Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Karin Schreiber
- Department of Pathology, Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - Henrik Clausen
- Copenhagen Center for Glycomics, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Hans Schreiber
- Department of Pathology, Committee on Immunology, University of Chicago, Chicago, IL 60637
| | - David M Kranz
- Department of Biochemistry, Cancer Center, University of Illinois at Urbana-Champaign, Urbana, IL 61801;
| |
Collapse
|