1
|
Ceolin V, Spadea M, Apolito V, Saglio F, Fagioli F. Emerging CART Therapies for Pediatric Acute Myeloid Leukemia. J Pediatr Hematol Oncol 2024; 46:393-403. [PMID: 39469946 DOI: 10.1097/mph.0000000000002956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/16/2024] [Indexed: 10/30/2024]
Abstract
The prognosis of children with acute myeloid leukemia (AML) has improved incrementally over the last decades. However, at relapse, overall survival (OS) ∼40% to 50% and is even lower for patients with chemorefractory disease. Effective and less-toxic therapies are urgently needed for these children. In the last years, immune-directed therapies such as chimeric antigen receptor (CAR)-T cells were introduced, which showed outstanding clinical activity against B-cell malignancies. CART therapies are being developed for AML on the basis of the results obtained for other hematologic malignancies. 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 hematopoietic stem cells. An overview of prospects of CART in pediatric AML, focused on the common antigens targeted by CART in AML that have been tested or are currently under investigation, is provided in this manuscript.
Collapse
Affiliation(s)
- Valeria Ceolin
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
| | - Manuela Spadea
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
- Department of Pediatric Oncology/Hematology, University of Turin, Turin, Italy
| | - Vincenzo Apolito
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
| | - Francesco Saglio
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
| | - Franca Fagioli
- Department of Pediatric Oncology/Hematology, Regina Margherita Children's Hospital
- Department of Pediatric Oncology/Hematology, University of Turin, Turin, Italy
| |
Collapse
|
2
|
Colonne CK, Kimble EL, Turtle CJ. Evolving strategies to overcome barriers in CAR-T cell therapy for acute myeloid leukemia. Expert Rev Hematol 2024:1-22. [PMID: 39439295 DOI: 10.1080/17474086.2024.2420614] [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: 06/26/2024] [Revised: 09/05/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a complex and heterogeneous disease characterized by an aggressive clinical course and limited efficacious treatment options in the relapsed/refractory (R/R) setting. Chimeric antigen receptor (CAR)-modified T (CAR-T) cell immunotherapy is an investigational treatment strategy for R/R AML that has shown some promise. However, obstacles to successful CAR-T cell immunotherapy for AML remain. AREAS COVERED In analyses of clinical trials of CAR-T cell therapy for R/R AML, complete responses without measurable residual disease have been reported, but the durability of those responses remains unclear. Significant barriers to successful CAR-T cell therapy in AML include the scarcity of suitable tumor-target antigens (TTA), inherent T cell functional deficits, and the immunoinhibitory and hostile tumor microenvironment (TME). This review will focus on these barriers to successful CAR-T cell therapy in AML, and discuss scientific advancements and evolving strategies to overcome them. EXPERT OPINION Achieving durable remissions in R/R AML will likely require a multifaceted approach that integrates advancements in TTA selection, enhancement of the intrinsic quality of CAR-T cells, and development of strategies to overcome inhibitory mechanisms in the AML TME.
Collapse
Affiliation(s)
- Chanukya K Colonne
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
| | - Erik L Kimble
- Translational Science and Therapeutic Division, Fred Hutchinson Cancer Center, Seattle, USA
- Department of Medicine, Division of Hematology and Oncology, University of Washington, Seattle, USA
| | - Cameron J Turtle
- Faculty of Medicine and Health, The University of Sydney, Sydney, Australia
- Haematology and Transfusion Medicine, Royal North Shore Hospital, Sydney, Australia
| |
Collapse
|
3
|
Kheirkhah AH, Habibi S, Yousefi MH, Mehri S, Ma B, Saleh M, Kavianpour M. Finding potential targets in cell-based immunotherapy for handling the challenges of acute myeloid leukemia. Front Immunol 2024; 15:1460437. [PMID: 39411712 PMCID: PMC11474923 DOI: 10.3389/fimmu.2024.1460437] [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: 07/06/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Acute myeloid leukemia (AML) is a hostile hematological malignancy under great danger of relapse and poor long-term survival rates, despite recent therapeutic advancements. To deal with this unfulfilled clinical necessity, innovative cell-based immunotherapies have surfaced as promising approaches to improve anti-tumor immunity and enhance patient outcomes. In this comprehensive review, we provide a detailed examination of the latest developments in cell-based immunotherapies for AML, including chimeric antigen receptor (CAR) T-cell therapy, T-cell receptor (TCR)-engineered T-cell therapy, and natural killer (NK) cell-based therapies. We critically evaluate the unique mechanisms of action, current challenges, and evolving strategies to improve the efficacy and safety of these modalities. The review emphasizes how promising these cutting-edge immune-based strategies are in overcoming the inherent complexities and heterogeneity of AML. We discuss the identification of optimal target antigens, the importance of mitigating on-target/off-tumor toxicity, and the need to enhance the persistence and functionality of engineered immune effector cells. All things considered, this review offers a thorough overview of the rapidly evolving field of cell-based immunotherapy for AML, underscoring the significant progress made and the ongoing efforts to translate these innovative approaches into more effective and durable treatments for this devastating disease.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Killer Cells, Natural/immunology
- Immunotherapy/methods
- Antigens, Neoplasm/immunology
- T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Amir Hossein Kheirkhah
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sina Habibi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad Hasan Yousefi
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Sara Mehri
- Department of Biotechnology, School of Paramedical Sciences, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Bin Ma
- School of Biomedical Engineering, Med-X Research Institute, Shanghai Jiao Tong University, Shanghai, China
- Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mahshid Saleh
- Wisconsin National Primate Research Center, University of Wisconsin Graduate School, Madison, WI, United States
| | - Maria Kavianpour
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| |
Collapse
|
4
|
Wang Y, Chang YJ, Chen J, Han M, Hu J, Hu J, Huang H, Lai Y, Liu D, Liu Q, Luo Y, Jiang EL, Jiang M, Song Y, Tang XW, Wu D, Xia LH, Xu K, Zhang X, Zhang XH, Huang X. Consensus on the monitoring, treatment, and prevention of leukaemia relapse after allogeneic haematopoietic stem cell transplantation in China: 2024 update. Cancer Lett 2024; 605:217264. [PMID: 39332587 DOI: 10.1016/j.canlet.2024.217264] [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: 07/02/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
The consensus in 2018 from The Chinese Society of Haematology (CSH) on the monitoring, treatment, and prevention of leukaemia relapse after allogeneic haematopoietic stem cell transplantation (HSCT) facilitated the standardization of clinical practices in China and progressive integration with the world. To integrate recent developments and further improve the consensus, a panel of experts from the CSH recently updated the following consensus: (1) integrate risk-adapted, measurable residual disease (MRD)-guided strategy on modified donor lymphocyte infusion (DLI) and interferon-α into total therapy, which was pioneered and refined by Chinese researchers; (2) provide additional evidence of the superiority of haploidentical HSCT (the dominant donor source in China) to matched HSCT for high-risk populations, especially for pre-HSCT MRD-positive patients; (3) support the rapid progress of techniques for MRD detection, such as next-generation sequencing (NGS) and leukaemia stem cell-based MRD detection; and (4) address the role of new targeted options in transplant settings. In conclusion, the establishment of a "total therapy" strategy represents a great step forward. We hope that the consensus updated by Chinese scholars will include the latest cutting-edge developments and inspire progress in post-HSCT relapse management.
Collapse
Affiliation(s)
- Yu Wang
- Peking University People's Hospital & Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, PR China
| | - Ying-Jun Chang
- Peking University People's Hospital & Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, PR China
| | - Jing Chen
- Shanghai Children's Medical Center, Shanghai, PR China
| | - Mingzhe Han
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Hematology and Blood Disease Hospital, Tianjin, PR China
| | - JianDa Hu
- The Second Affiliated Hospital of Fujian Medical University, Quanzhou, PR China
| | - Jiong Hu
- Ruijin Hospital, Shanghai Jiao Tong University, Shanghai, PR China
| | - He Huang
- First Affiliated Hospital of Zhejiang University, Hangzhou, PR China
| | - Yongrong Lai
- The First Affiliated Hospital of Guangxi Medical University, Nanning, PR China
| | - Daihong Liu
- General Hospital of PLA(People's Liberation Army of China), Beijing, PR China
| | - Qifa Liu
- Nanfang Hospital of Southern Medical University, Guangzhou, PR China
| | - Yi Luo
- First Affiliated Hospital of Zhejiang University, Hangzhou, PR China
| | - Er-Lie Jiang
- Chinese Academy of Medical Sciences and Peking Union Medical College, Institute of Hematology and Blood Disease Hospital, Tianjin, PR China
| | - Ming Jiang
- The First Affiliated Hospital of Xinjiang Medical University, Urumqi, PR China
| | - Yongping Song
- Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, PR China
| | - Xiao-Wen Tang
- The First Affiliated Hospital of Soochow University, Soochow, PR China
| | - Depei Wu
- The First Affiliated Hospital of Soochow University, Soochow, PR China
| | - Ling-Hui Xia
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Kailin Xu
- The First Affiliated Hospital of Xuzhou Medical University, Xuzhou, PR China
| | - Xi Zhang
- Xinqiao Hospital, Army Medical University, Chongqing, PR China
| | - Xiao-Hui Zhang
- Peking University People's Hospital & Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, PR China
| | - Xiaojun Huang
- Peking University People's Hospital & Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, No. 11 Xizhimen South Street, Xicheng District, Beijing, 100044, PR China; Peking-Tsinghua Center for Life Sciences, Beijing, PR China; State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, PR China.
| |
Collapse
|
5
|
Sabile JMG, Swords R, Tyner JW. Evaluating targeted therapies in older patients with TP53-mutated AML. Leuk Lymphoma 2024; 65:1201-1218. [PMID: 38646877 DOI: 10.1080/10428194.2024.2344057] [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: 11/23/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
Mutation of thetumor suppressor gene, TP53 (tumor protein 53), occurs in up to 15% of all patients with acute myeloid leukemia (AML) and is enriched within specific clinical subsets, most notably in older adults, and including secondary AML cases arising from preceding myeloproliferative neoplasm (MPN), myelodysplastic syndrome (MDS), patients exposed to prior DNA-damaging, cytotoxic therapies. In all cases, these tumors have remained difficult to effectively treat with conventional therapeutic regimens. Newer approaches fortreatmentofTP53-mutated AML have shifted to interventions that maymodulateTP53 function, target downstream molecular vulnerabilities, target non-p53 dependent molecular pathways, and/or elicit immunogenic responses. This review will describe the basic biology of TP53, the clinical and biological patterns of TP53 within myeloid neoplasms with a focus on elderly AML patients and will summarize newer therapeutic strategies and current clinical trials.
Collapse
Affiliation(s)
- Jean M G Sabile
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ronan Swords
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
6
|
Martín-Sánchez E, Blanco L, Kim PS, Bisht K, Wang H, Van de Velde H, Jelinek T, Simoes C, Prosper F, San Miguel JF, Alfonso A, Bergua J, Rodríguez-Veiga R, Vives S, Martínez-Cuadrón D, Montesinos P, Paiva B, Zabaleta A. Targeting CD38 with isatuximab and a novel CD38/CD3×CD28 trispecific T-cell engager in older patients with acute myeloid leukemia. Blood Adv 2024; 8:3875-3879. [PMID: 38815228 PMCID: PMC11321322 DOI: 10.1182/bloodadvances.2024013212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 05/07/2024] [Accepted: 05/20/2024] [Indexed: 06/01/2024] Open
Affiliation(s)
- Esperanza Martín-Sánchez
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Laura Blanco
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | | | | | | | | | - Tomas Jelinek
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
- Department of Hematooncology, University Hospital of Ostrava, and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Catia Simoes
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Felipe Prosper
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Jesus F. San Miguel
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Ana Alfonso
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Juan Bergua
- Hospital San Pedro de Alcántara, Cáceres, Spain
| | | | - Susana Vives
- Institut Català d'Oncologia Badalona-Hospital Germans Trias I Pujol, Josep Carreras Research Institute, Badalona, Spain
| | | | | | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Aintzane Zabaleta
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada, Instituto de Investigación Sanitaria de Navarra, CIBER-ONC numbers CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| |
Collapse
|
7
|
Chomczyk M, Gazzola L, Dash S, Firmanty P, George BS, Mohanty V, Abbas HA, Baran N. Impact of p53-associated acute myeloid leukemia hallmarks on metabolism and the immune environment. Front Pharmacol 2024; 15:1409210. [PMID: 39161899 PMCID: PMC11330794 DOI: 10.3389/fphar.2024.1409210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Accepted: 07/08/2024] [Indexed: 08/21/2024] Open
Abstract
Acute myeloid leukemia (AML), an aggressive malignancy of hematopoietic stem cells, is characterized by the blockade of cell differentiation, uncontrolled proliferation, and cell expansion that impairs healthy hematopoiesis and results in pancytopenia and susceptibility to infections. Several genetic and chromosomal aberrations play a role in AML and influence patient outcomes. TP53 is a key tumor suppressor gene involved in a variety of cell features, such as cell-cycle regulation, genome stability, proliferation, differentiation, stem-cell homeostasis, apoptosis, metabolism, senescence, and the repair of DNA damage in response to cellular stress. In AML, TP53 alterations occur in 5%-12% of de novo AML cases. These mutations form an important molecular subgroup, and patients with these mutations have the worst prognosis and shortest overall survival among patients with AML, even when treated with aggressive chemotherapy and allogeneic stem cell transplant. The frequency of TP53-mutations increases in relapsed and recurrent AML and is associated with chemoresistance. Progress in AML genetics and biology has brought the novel therapies, however, the clinical benefit of these agents for patients whose disease is driven by TP53 mutations remains largely unexplored. This review focuses on the molecular characteristics of TP53-mutated disease; the impact of TP53 on selected hallmarks of leukemia, particularly metabolic rewiring and immune evasion, the clinical importance of TP53 mutations; and the current progress in the development of preclinical and clinical therapeutic strategies to treat TP53-mutated disease.
Collapse
Affiliation(s)
- Monika Chomczyk
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Luca Gazzola
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Shubhankar Dash
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Patryk Firmanty
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Binsah S. George
- Department of Hematology-oncology, The University of Texas Health Sciences, Houston, TX, United States
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Hussein A. Abbas
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Natalia Baran
- Department of Experimental Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
8
|
Chandra DJ, Alber B, Saultz JN. The Immune Resistance Signature of Acute Myeloid Leukemia and Current Immunotherapy Strategies. Cancers (Basel) 2024; 16:2615. [PMID: 39123343 PMCID: PMC11311077 DOI: 10.3390/cancers16152615] [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: 05/03/2024] [Revised: 06/29/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024] Open
Abstract
Acute myeloid leukemia (AML) is a complex hematopoietic clonal disorder with limited curative options beyond stem cell transplantation. The success of transplant is intimately linked with the graft versus leukemia effect from the alloreactive donor immune cells including, T and NK cells. The immune system plays a dynamic role in leukemia survival and resistance. Despite our growing understanding of the immune microenvironment, responses to immune-based therapies differ greatly between patients. Herein, we review the biology of immune evasion mechanisms in AML, discuss the current landscape of immunotherapeutic strategies, and discuss the implications of therapeutic targets. This review focuses on T and NK cell-based therapy, including modified and non-modified NK cells, CAR-T and CAR-NK cells, antibodies, and checkpoint blockades. Understanding the complex interchange between immune tolerance and the emergence of tumor resistance will improve patient outcomes.
Collapse
Affiliation(s)
- Daniel J. Chandra
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Bernhard Alber
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| | - Jennifer N. Saultz
- Division of Hematology/Medical Oncology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA;
| |
Collapse
|
9
|
Tameni A, Toffalori C, Vago L. Tricking the trickster: precision medicine approaches to counteract leukemia immune escape after transplant. Blood 2024; 143:2710-2721. [PMID: 38728431 DOI: 10.1182/blood.2023019962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 04/08/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024] Open
Abstract
ABSTRACT Over the last decades, significant improvements in reducing the toxicities of allogeneic hematopoietic cell transplantation (allo-HCT) have widened its use as consolidation or salvage therapy for high-risk hematological malignancies. Nevertheless, relapse of the original malignant disease remains an open issue with unsatisfactory salvage options and limited rationales to select among them. In the last years, several studies have highlighted that relapse is often associated with specific genomic and nongenomic mechanisms of immune escape. In this review we summarize the current knowledge about these modalities of immune evasion, focusing on the mechanisms that leverage antigen presentation and pathologic rewiring of the bone marrow microenvironment. We present examples of how this biologic information can be translated into specific approaches to treat relapse, discuss the status of the clinical trials for patients who relapsed after a transplant, and show how dissecting the complex immunobiology of allo-HCT represents a crucial step toward developing new personalized approaches to improve clinical outcomes.
Collapse
Affiliation(s)
- Annalisa Tameni
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Cristina Toffalori
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Luca Vago
- Unit of Immunogenetics, Leukemia Genomics and Immunobiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Hematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Dutta S, Mudaranthakam DP, Li Y, Sardiu ME. PerSEveML: a web-based tool to identify persistent biomarker structure for rare events using an integrative machine learning approach. Mol Omics 2024; 20:348-358. [PMID: 38690925 DOI: 10.1039/d4mo00008k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
Omics data sets often pose a computational challenge due to their high dimensionality, large size, and non-linear structures. Analyzing these data sets becomes especially daunting in the presence of rare events. Machine learning (ML) methods have gained traction for analyzing rare events, yet there has been limited exploration of bioinformatics tools that integrate ML techniques to comprehend the underlying biology. Expanding upon our previously developed computational framework of an integrative machine learning approach, we introduce PerSEveML, an interactive web-based tool that uses crowd-sourced intelligence to predict rare events and determine feature selection structures. PerSEveML provides a comprehensive overview of the integrative approach through evaluation metrics that help users understand the contribution of individual ML methods to the prediction process. Additionally, PerSEveML calculates entropy and rank scores, which visually organize input features into a persistent structure of selected, unselected, and fluctuating categories that help researchers uncover meaningful hypotheses regarding the underlying biology. We have evaluated PerSEveML on three diverse biologically complex data sets with extremely rare events from small to large scale and have demonstrated its ability to generate valid hypotheses. PerSEveML is available at https://biostats-shinyr.kumc.edu/PerSEveML/ and https://github.com/sreejatadutta/PerSEveML.
Collapse
Affiliation(s)
- Sreejata Dutta
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
| | - Dinesh Pal Mudaranthakam
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
- University of Kansas Cancer Center, Kansas City, USA
| | - Yanming Li
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
- University of Kansas Cancer Center, Kansas City, USA
| | - Mihaela E Sardiu
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA.
- University of Kansas Cancer Center, Kansas City, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
11
|
Damiani D, Tiribelli M. CAR-T Cells in Acute Myeloid Leukemia: Where Do We Stand? Biomedicines 2024; 12:1194. [PMID: 38927401 PMCID: PMC11200794 DOI: 10.3390/biomedicines12061194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 06/28/2024] Open
Abstract
Despite recent advances, the prognosis of acute myeloid leukemia (AML) remains unsatisfactory due to disease recurrence and the development of resistance to both conventional and novel therapies. Engineered T cells expressing chimeric antigen receptors (CARs) on their cellular surface represent one of the most promising anticancer agents. CAR-T cells are increasingly used in patients with B cell malignancies, with remarkable clinical results despite some immune-related toxicities. However, at present, the role of CAR-T cells in myeloid neoplasms, including AML, is extremely limited, as specific molecular targets for immune cells are generally lacking on AML blasts. Besides the paucity of dispensable targets, as myeloid antigens are often co-expressed on normal hematopoietic stem and progenitor cells with potentially intolerable myeloablation, the AML microenvironment is hostile to T cell proliferation due to inhibitory soluble factors. In addition, the rapidly progressive nature of the disease further complicates the use of CAR-T in AML. This review discusses the current state of CAR-T cell therapy in AML, including the still scanty clinical evidence and the potential approaches to overcome its limitations, including genetic modifications and combinatorial strategies, to make CAR-T cell therapy an effective option for AML patients.
Collapse
Affiliation(s)
- Daniela Damiani
- Division of Hematology and Stem Cell Transplantation, University Hospital, 33100 Udine, Italy;
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy
| | - Mario Tiribelli
- Division of Hematology and Stem Cell Transplantation, University Hospital, 33100 Udine, Italy;
- Department of Medicine (DMED), University of Udine, 33100 Udine, Italy
| |
Collapse
|
12
|
Vittayawacharin P, Kongtim P, Chu Y, June CH, Bollard CM, Ciurea SO. Adoptive cellular therapy after hematopoietic stem cell transplantation. Am J Hematol 2024; 99:910-921. [PMID: 38269484 DOI: 10.1002/ajh.27204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 12/13/2023] [Accepted: 01/01/2024] [Indexed: 01/26/2024]
Abstract
Effective cellular therapy using CD19 chimeric antigen receptor T-cells for the treatment of advanced B-cell malignancies raises the question of whether the administration of adoptive cellular therapy (ACT) posttransplant could reduce relapse and improve survival. Moreover, several early phase clinical studies have shown the potential beneficial effects of administration of tumor-associated antigen-specific T-cells and natural killer cells posttransplant for high-risk patients, aiming to decrease relapse and possibly improve survival. In this article, we present an in-depth review of ACT after transplantation, which has the potential to significantly improve the efficacy of this procedure and revolutionize this field.
Collapse
Affiliation(s)
- Pongthep Vittayawacharin
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Orange, California, USA
| | - Piyanuch Kongtim
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Orange, California, USA
| | - Yaya Chu
- Department of Pediatrics, New York Medical College, Valhalla, New York, USA
| | - Carl H June
- Department of Pathology and Laboratory Medicine, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Catherine M Bollard
- Center for Cancer and Immunology Research, Children's National Hospital and The George Washington University, Washington, DC, USA
| | - Stefan O Ciurea
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Department of Medicine, University of California, Irvine, Orange, California, USA
| |
Collapse
|
13
|
Szlasa W, Sztuder A, Kaczmar-Dybko A, Maciejczyk A, Dybko J. Efficient combination of radiotherapy and CAR-T - A systematic review. Biomed Pharmacother 2024; 174:116532. [PMID: 38574625 DOI: 10.1016/j.biopha.2024.116532] [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: 01/09/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/06/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy, a groundbreaking immunotherapy. However, it faces formidable challenges in treating solid tumors, grappling with issues like poor trafficking, limited penetration, and insufficient persistence within the tumor microenvironment (TME). CAR-T cells are engineered to express receptors that target specific cancer antigens, enhancing their ability to recognize and eliminate cancer cells. This review paper explores the intricate interplay between CAR-T therapy and radiotherapy (RT), investigating their synergistic potential. Radiotherapy, a standard cancer treatment, involves using high doses of radiation to target and damage cancer cells, disrupting their ability to grow and divide. We highlight that RT modulates the TME, augments antigen presentation, and promotes immune cell infiltration, bolstering CAR-T cell-mediated tumor eradication. Molecular insights shed light on RT-induced alterations in tumor stroma, T cell recruitment promotion, and induction of immunogenic cell death. Noteworthy, strategies, such as combining hypofractionated radiotherapy with myeloid-derived suppressor cell blockade, underscore innovative approaches to enhance CAR-T cell therapy in solid tumors. Bridging indications for RT and CAR-T cells in hematological malignancies are discussed, emphasizing scenarios where RT strategically enhances CAR-T cell efficacy. The paper critically evaluates the RT as a bridge compared to traditional chemotherapy, highlighting timing and dosage considerations crucial for optimizing CAR-T therapy outcomes. In summary, the paper provides valuable insights into the intricate molecular mechanisms activated by RT and innovative strategies to improve CAR-T cell therapy, fostering a deeper understanding of their combined potential in cancer treatment.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Medical University Hospital, Borowska 213, Wrocław 50-556, Poland.
| | - Aleksandra Sztuder
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | | | - Adam Maciejczyk
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology, Wroclaw Medical University, Wroclaw 50-367, Poland
| | - Jarosław Dybko
- Lower Silesian Centre of Oncology, Pulmonology and Hematology, Wroclaw 53-413, Poland; Department of Oncology and Hematology, Wroclaw University of Science and Technology, Wrocław 50-370, Poland
| |
Collapse
|
14
|
Gao C, Li X, Xu Y, Zhang T, Zhu H, Yao D. Recent advances in CAR-T cell therapy for acute myeloid leukaemia. J Cell Mol Med 2024; 28:e18369. [PMID: 38712978 PMCID: PMC11075639 DOI: 10.1111/jcmm.18369] [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: 09/12/2023] [Revised: 02/18/2024] [Accepted: 04/17/2024] [Indexed: 05/08/2024] Open
Abstract
Acute myeloid leukaemia (AML) is a fatal and refractory haematologic cancer that primarily affects adults. It interferes with bone marrow cell proliferation. Patients have a 5 years survival rate of less than 30% despite the availability of several treatments, including chemotherapy, allogeneic haematopoietic stem cell transplantation (Allo-HSCT), and receptor antagonist drugs. Allo-HSCT is the mainstay of acute myeloid leukaemia treatment. Although it does work, there are severe side effects, such as graft-versus-host disease (GVHD). In recent years, chimeric antigen receptor (CAR)-T cell therapies have made significant progress in the treatment of cancer. These engineered T cells can locate and recognize tumour cells in vivo and release a large number of effectors through immune action to effectively kill tumour cells. CAR-T cells are among the most effective cancer treatments because of this property. CAR-T cells have demonstrated positive therapeutic results in the treatment of acute myeloid leukaemia, according to numerous clinical investigations. This review highlights recent progress in new targets for AML immunotherapy, and the limitations, and difficulties of CAR-T therapy for AML.
Collapse
Affiliation(s)
- Chi Gao
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Xin Li
- College of BiotechnologyTianjin University of Science and TechnologyTianjinChina
| | - Yao Xu
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Tongcun Zhang
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
- Institute of Biology and MedicineWuhan University of Science and TechnologyWuhanChina
| | - Haichuan Zhu
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| | - Di Yao
- College of Life Science and HealthWuhan University of Science and TechnologyWuhanChina
| |
Collapse
|
15
|
Morsy MM, Azzam AY, Elamin O, Elswedy A, Nashwan AJ. Safety and efficacy of chimeric antigen receptor T-cell therapy for acute myeloid leukemia: A subgroup based meta-analysis. Leuk Res 2024; 140:107498. [PMID: 38582045 DOI: 10.1016/j.leukres.2024.107498] [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: 02/11/2024] [Revised: 03/22/2024] [Accepted: 03/30/2024] [Indexed: 04/08/2024]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a significant hematological malignancy in the United States, with a high mortality rate and limited treatment options. CAR T-cell therapy, a new and promising treatment, is being investigated for its efficacy and safety in AML. This meta-analysis aims to assess the safety and efficacy of CAR T-cell therapy in AML, considering various subgroups such as study location, study design, prior transplantation status, conditioning regimen, and CAR T-cell source. METHODS We conducted a comprehensive literature review across multiple databases, adhering to PRISMA guidelines and focusing on studies concerning CAR T-cell therapy in AML. We included original articles in English and excluded non-original reviews, abstracts, and non-English studies. The risk of bias was assessed using the Cochrane ROBINS-I tool. Statistical analysis involved meta-analysis with Cochrane's Q-test and I² statistic, using both fixed-effect and random-effects models, and assessed for publication bias. RESULTS Our search yielded studies encompassing 57 AML patients treated with CAR T-cell therapy. The meta-analysis revealed a 48% incidence of complete remission with CAR T-cell therapy, varying significantly across subgroups based on study design, location, prior transplantation, conditioning regimen, and CAR T-cell source. The highest complete remission rates were observed in patients from China, those who had undergone prior hematopoietic cell transplantation, and those treated with fludarabine and cyclophosphamide conditioning regimen. Adverse events included graft-versus-host disease (7%) and cytokine release syndrome (53%). CONCLUSIONS This meta-analysis highlights the potential of CAR T-cell therapy in AML treatment, especially when integrated with certain prior treatments and conditioning regimens. The findings suggest a higher efficacy in patients with previous hematopoietic cell transplantation and specific conditioning regimens. Further large-scale, randomized trials are essential to confirm these findings and establish CAR T-cell therapy as a standard treatment for AML.
Collapse
Affiliation(s)
| | - Ahmed Y Azzam
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Osman Elamin
- Department of Neurosurgery, Jordan Hospital, Amman, Jordan
| | - Adam Elswedy
- Faculty of Medicine, October 6 University, Giza, Egypt
| | - Abdulqadir J Nashwan
- Nursing & Midwifery Research Department (NMRD), Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
16
|
Jiang Y, Feng D, Zhu J, Wei D, Zhao C, Liu H, Shao S, Wang C. Case report: Preventive infusion of donor-derived CD7 chimeric antigen receptor T cells after allogeneic hematopoietic stem cell transplantation. Front Immunol 2024; 15:1381308. [PMID: 38745670 PMCID: PMC11091287 DOI: 10.3389/fimmu.2024.1381308] [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: 02/03/2024] [Accepted: 04/08/2024] [Indexed: 05/16/2024] Open
Abstract
Chimeric antigen receptor T cells (CAR T) targeting CD7 for T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL) showed promising efficacy and safety in some clinical trials. However, most of them were bridged with allogeneic hematopoietic stem cell transplantation (allo-HSCT). We described successful treatment with preventive donor-derived anti-CD7 CAR-T therapy in a case of refractory T lymphoblastic lymphoma following allo-HSCT, who could not receive autologous anti-CD7 CAR-T products due to the low-quality of T lymphocytes. To date, the patient's complete remission has persisted for 20 months after HSCT.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chun Wang
- Department of Hematology, Shanghai Zhaxin Traditional Chinese and Western Medicine Hospital, Shanghai, China
| |
Collapse
|
17
|
Murtadha M, Park M, Zhu Y, Caserta E, Napolitano O, Tandoh T, Moloudizargari M, Pozhitkov A, Singer M, Dona AA, Vahed H, Gonzalez A, Ly K, Ouyang C, Sanchez JF, Nigam L, Duplan A, Chowdhury A, Ghoda L, Li L, Zhang B, Krishnan A, Marcucci G, Williams JC, Pichiorri F. A CD38-directed, single-chain T-cell engager targets leukemia stem cells through IFN-γ-induced CD38 expression. Blood 2024; 143:1599-1615. [PMID: 38394668 PMCID: PMC11103097 DOI: 10.1182/blood.2023021570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 02/25/2024] Open
Abstract
ABSTRACT Treatment resistance of leukemia stem cells (LSCs) and suppression of the autologous immune system represent major challenges to achieve a cure in acute myeloid leukemia (AML). Although AML blasts generally retain high levels of surface CD38 (CD38pos), LSCs are frequently enriched in the CD34posCD38neg blast fraction. Here, we report that interferon gamma (IFN-γ) reduces LSCs clonogenic activity and induces CD38 upregulation in both CD38pos and CD38neg LSC-enriched blasts. IFN-γ-induced CD38 upregulation depends on interferon regulatory factor 1 transcriptional activation of the CD38 promoter. To leverage this observation, we created a novel compact, single-chain CD38-CD3 T-cell engager (BN-CD38) designed to promote an effective immunological synapse between CD38pos AML cells and both CD8pos and CD4pos T cells. We demonstrate that BN-CD38 engages autologous CD4pos and CD8pos T cells and CD38pos AML blasts, leading to T-cell activation and expansion and to the elimination of leukemia cells in an autologous setting. Importantly, BN-CD38 engagement induces the release of high levels of IFN-γ, driving the expression of CD38 on CD34posCD38neg LSC-enriched blasts and their subsequent elimination. Critically, although BN-CD38 showed significant in vivo efficacy across multiple disseminated AML cell lines and patient-derived xenograft models, it did not affect normal hematopoietic stem cell clonogenicity and the development of multilineage human immune cells in CD34pos humanized mice. Taken together, this study provides important insights to target and eliminate AML LSCs.
Collapse
Affiliation(s)
- Mariam Murtadha
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Miso Park
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Yinghui Zhu
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
- Research Center for Translational Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Enrico Caserta
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ottavio Napolitano
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Theophilus Tandoh
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Milad Moloudizargari
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Alex Pozhitkov
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Mahmoud Singer
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ada Alice Dona
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Hawa Vahed
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Asaul Gonzalez
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Kevin Ly
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ching Ouyang
- Integrative Genomics Core, City of Hope Comprehensive Cancer Center, City of Hope, Duarte, CA
| | - James F. Sanchez
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
| | - Lokesh Nigam
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Amanda Duplan
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Arnab Chowdhury
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Computational and Quantitative Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Lucy Ghoda
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Ling Li
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Bin Zhang
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - Amrita Krishnan
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
| | - Guido Marcucci
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| | - John C. Williams
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA
| | - Flavia Pichiorri
- Department of Hematology and Hematopoietic Cell Transplantation, Judy and Bernard Briskin Center for Multiple Myeloma Research, City of Hope, Duarte, CA
- Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, CA
| |
Collapse
|
18
|
Roders N, Nakid-Cordero C, Raineri F, Fayon M, Abecassis A, Choisy C, Nelson E, Maillard C, Garrick D, Talbot A, Fermand JP, Arnulf B, Bories JC. Dual Chimeric Antigen Receptor T Cells Targeting CD38 and SLAMF7 with Independent Signaling Demonstrate Preclinical Efficacy and Safety in Multiple Myeloma. Cancer Immunol Res 2024; 12:478-490. [PMID: 38289260 DOI: 10.1158/2326-6066.cir-23-0839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/29/2023] [Accepted: 01/26/2024] [Indexed: 04/04/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma targeting B-cell maturation antigen (BCMA) induces high overall response rates. However, relapse still occurs and novel strategies for targeting multiple myeloma cells using CAR T-cell therapy are needed. SLAMF7 (also known as CS1) and CD38 on tumor plasma cells represent potential alternative targets for CAR T-cell therapy in multiple myeloma, but their expression on activated T cells and other hematopoietic cells raises concerns about the efficacy and safety of such treatments. Here, we used CRISPR/Cas9 deletion of the CD38 gene in T cells and developed DCAR, a double CAR system targeting CD38 and CS1 through activation and costimulation receptors, respectively. Inactivation of CD38 enhanced the anti-multiple myeloma activity of DCAR T in vitro. Edited DCAR T cells showed strong in vitro and in vivo responses specifically against target cells expressing both CD38 and CS1. Furthermore, we provide evidence that, unlike anti-CD38 CAR T-cell therapy, which elicited a rapid immune reaction against hematopoietic cells in a humanized mouse model, DCAR T cells showed no signs of toxicity. Thus, DCAR T cells could provide a safe and efficient alternative to anti-BCMA CAR T-cell therapy to treat patients with multiple myeloma.
Collapse
Affiliation(s)
- Nathalie Roders
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Cecilia Nakid-Cordero
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Fabio Raineri
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Maxime Fayon
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Audrey Abecassis
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Caroline Choisy
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Elisabeth Nelson
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | | | - David Garrick
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| | - Alexis Talbot
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
- Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Jean-Paul Fermand
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
- Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Bertrand Arnulf
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
- Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Jean-Christophe Bories
- INSERM, Human Immunology, Pathophysiology, Immunotherapy (HIPI), Institut de Recherche Saint-Louis, Université de Paris-Cité, Paris, France
| |
Collapse
|
19
|
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
|
20
|
Chen X, Wong OK, Reiman L, Sherbenou DW, Post L. CD38 x ICAM-1 Bispecific Antibody Is a Novel Approach for Treating Multiple Myeloma and Lymphoma. Mol Cancer Ther 2024; 23:127-138. [PMID: 37816503 DOI: 10.1158/1535-7163.mct-23-0052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/18/2023] [Accepted: 10/05/2023] [Indexed: 10/12/2023]
Abstract
The cluster of differentiation 38 (CD38) is a well-validated target for treating multiple myeloma. Although anti-CD38 mAbs have demonstrated outstanding initial responses in patients with multiple myeloma, nearly all patients eventually develop resistance and relapse. In addition, currently approved CD38 targeting therapies have failed to show monotherapy efficacy in lymphomas, where CD38 expression is present but at lower levels. To effectively target CD38 on tumor cells, we generated an antibody-dependent cellular cytotoxicity (ADCC) enhanced bispecific CD38 x intercellular cell adhesion molecule 1 (ICAM-1) antibody, VP301. This bispecific antibody targets unique epitopes on CD38 and ICAM-1 on tumor cells with reduced red blood cell binding compared with the benchmark CD38 antibody daratumumab. VP301 demonstrated potent ADCC and antibody-dependent cellular phagocytosis activities on a selected set of myeloma and lymphoma cell lines even those with low CD38 expression. In an ex vivo drug sensitivity assay, we observed responses to VP301 in multiple myeloma primary samples from relapsed/refractory patients. Moreover, VP301 demonstrated potent tumor inhibition activities in in vivo myeloma and lymphoma models. Interestingly, combination of VP301 with the immunomodulatory drug, lenalidomide, led to synergistic antitumor growth activity in an in vivo efficacy study. In conclusion, the CD38 x ICAM-1 bispecific antibody VP301 demonstrated promising efficacy and specificity toward CD38+ and ICAM-1+ tumor cells and represents a novel approach for treating multiple myeloma and lymphoma.
Collapse
Affiliation(s)
| | - Oi Kwan Wong
- Virtuoso Therapeutics, Inc., San Mateo, California
| | - Lauren Reiman
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Daniel W Sherbenou
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Leonard Post
- Virtuoso Therapeutics, Inc., San Mateo, California
| |
Collapse
|
21
|
Edri A, Ben-Haim N, Hailu A, Brycman N, Berhani-Zipori O, Rifman J, Cohen S, Yackoubov D, Rosenberg M, Simantov R, Teru H, Kurata K, Anderson KC, Hendel A, Pato A, Geffen Y. Nicotinamide-Expanded Allogeneic Natural Killer Cells with CD38 Deletion, Expressing an Enhanced CD38 Chimeric Antigen Receptor, Target Multiple Myeloma Cells. Int J Mol Sci 2023; 24:17231. [PMID: 38139060 PMCID: PMC10743602 DOI: 10.3390/ijms242417231] [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: 09/04/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Natural killer (NK) cells are a vital component of cancer immune surveillance. They provide a rapid and potent immune response, including direct cytotoxicity and mobilization of the immune system, without the need for antigen processing and presentation. NK cells may also be better tolerated than T cell therapy approaches and are susceptible to various gene manipulations. Therefore, NK cells have become the focus of extensive translational research. Gamida Cell's nicotinamide (NAM) platform for cultured NK cells provides an opportunity to enhance the therapeutic potential of NK cells. CD38 is an ectoenzyme ubiquitously expressed on the surface of various hematologic cells, including multiple myeloma (MM). It has been selected as a lead target for numerous monoclonal therapeutic antibodies against MM. Monoclonal antibodies target CD38, resulting in the lysis of MM plasma cells through various antibody-mediated mechanisms such as antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity, and antibody-dependent cellular phagocytosis, significantly improving the outcomes of patients with relapsed or refractory MM. However, this therapeutic strategy has inherent limitations, such as the anti-CD38-induced depletion of CD38-expressing NK cells, thus hindering ADCC. We have developed genetically engineered NK cells tailored to treat MM, in which CD38 was knocked-out using CRISPR-Cas9 technology and an enhanced chimeric antigen receptor (CAR) targeting CD38 was introduced using mRNA electroporation. This combined genetic approach allows for an improved cytotoxic activity directed against CD38-expressing MM cells without self-inflicted NK-cell-mediated fratricide. Preliminary results show near-complete abolition of fratricide with a 24-fold reduction in self-lysis from 19% in mock-transfected and untreated NK cells to 0.8% of self-lysis in CD38 knock-out CAR NK cells. Furthermore, we have observed significant enhancements in CD38-mediated activity in vitro, resulting in increased lysis of MM target cell lines. CD38 knock-out CAR NK cells also demonstrated significantly higher levels of NK activation markers in co-cultures with both untreated and αCD38-treated MM cell lines. These NAM-cultured NK cells with the combined genetic approach of CD38 knockout and addition of CD38 CAR represent a promising immunotherapeutic tool to target MM.
Collapse
Affiliation(s)
- Avishay Edri
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Nimrod Ben-Haim
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (N.B.-H.); (M.R.)
| | - Astar Hailu
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Nurit Brycman
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Orit Berhani-Zipori
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Julia Rifman
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Sherri Cohen
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Dima Yackoubov
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Michael Rosenberg
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (N.B.-H.); (M.R.)
| | | | - Hideshima Teru
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; (H.T.); (K.K.); (K.C.A.)
| | - Keiji Kurata
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; (H.T.); (K.K.); (K.C.A.)
| | - Kenneth Carl Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA; (H.T.); (K.K.); (K.C.A.)
| | - Ayal Hendel
- Institute of Nanotechnology and Advanced Materials, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel; (N.B.-H.); (M.R.)
| | - Aviad Pato
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| | - Yona Geffen
- Gamida-Cell, Jerusalem 34670, Israel; (A.E.); (A.H.); (N.B.); (O.B.-Z.); (J.R.); (S.C.); (D.Y.); (A.P.)
| |
Collapse
|
22
|
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
|
23
|
Karsten H, Matrisch L, Cichutek S, Fiedler W, Alsdorf W, Block A. Broadening the horizon: potential applications of CAR-T cells beyond current indications. Front Immunol 2023; 14:1285406. [PMID: 38090582 PMCID: PMC10711079 DOI: 10.3389/fimmu.2023.1285406] [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: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Engineering immune cells to treat hematological malignancies has been a major focus of research since the first resounding successes of CAR-T-cell therapies in B-ALL. Several diseases can now be treated in highly therapy-refractory or relapsed conditions. Currently, a number of CD19- or BCMA-specific CAR-T-cell therapies are approved for acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), multiple myeloma (MM), and follicular lymphoma (FL). The implementation of these therapies has significantly improved patient outcome and survival even in cases with previously very poor prognosis. In this comprehensive review, we present the current state of research, recent innovations, and the applications of CAR-T-cell therapy in a selected group of hematologic malignancies. We focus on B- and T-cell malignancies, including the entities of cutaneous and peripheral T-cell lymphoma (T-ALL, PTCL, CTCL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia (CLL), classical Hodgkin-Lymphoma (HL), Burkitt-Lymphoma (BL), hairy cell leukemia (HCL), and Waldenström's macroglobulinemia (WM). While these diseases are highly heterogenous, we highlight several similarly used approaches (combination with established therapeutics, target depletion on healthy cells), targets used in multiple diseases (CD30, CD38, TRBC1/2), and unique features that require individualized approaches. Furthermore, we focus on current limitations of CAR-T-cell therapy in individual diseases and entities such as immunocompromising tumor microenvironment (TME), risk of on-target-off-tumor effects, and differences in the occurrence of adverse events. Finally, we present an outlook into novel innovations in CAR-T-cell engineering like the use of artificial intelligence and the future role of CAR-T cells in therapy regimens in everyday clinical practice.
Collapse
Affiliation(s)
- Hendrik Karsten
- Faculty of Medicine, University of Hamburg, Hamburg, Germany
| | - Ludwig Matrisch
- Department of Rheumatology and Clinical Immunology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Faculty of Medicine, University of Lübeck, Lübeck, Germany
| | - Sophia Cichutek
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Andreas Block
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
24
|
Konishi T, Ochi T, Maruta M, Tanimoto K, Miyazaki Y, Iwamoto C, Saitou T, Imamura T, Yasukawa M, Takenaka K. Reinforced antimyeloma therapy via dual-lymphoid activation mediated by a panel of antibodies armed with bridging-BiTE. Blood 2023; 142:1789-1805. [PMID: 37738633 DOI: 10.1182/blood.2022019082] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 08/31/2023] [Accepted: 09/01/2023] [Indexed: 09/24/2023] Open
Abstract
Immunotherapy using bispecific antibodies including bispecific T-cell engager (BiTE) has the potential to enhance the efficacy of treatment for relapsed/refractory multiple myeloma. However, myeloma may still recur after treatment because of downregulation of a target antigen and/or myeloma cell heterogeneity. To strengthen immunotherapy for myeloma while overcoming its characteristics, we have newly developed a BiTE-based modality, referred to as bridging-BiTE (B-BiTE). B-BiTE was able to bind to both a human immunoglobulin G-Fc domain and the CD3 molecule. Clinically available monoclonal antibodies (mAbs) were bound with B-BiTE before administration, and the mAb/B-BiTE complex induced antitumor T-cell responses successfully while preserving and supporting natural killer cell reactivity, resulting in enhanced antimyeloma effects via dual-lymphoid activation. In contrast, any unwanted off-target immune-cell reactivity mediated by mAb/B-BiTE complexes or B-BiTE itself appeared not to be observed in vitro and in vivo. Importantly, sequential immunotherapy using 2 different mAb/B-BiTE complexes appeared to circumvent myeloma cell antigen escape, and further augmented immune responses to myeloma relative to those induced by mAb/B-BiTE monotherapy or sequential therapy with 2 mAbs in the absence of B-BiTE. Therefore, this modality facilitates easy and prompt generation of a broad panel of bispecific antibodies that can induce deep and durable antitumor responses in the presence of clinically available mAbs, supporting further advancement of reinforced immunotherapy for multiple myeloma and other refractory hematologic malignancies.
Collapse
Affiliation(s)
- Tatsuya Konishi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Toshiki Ochi
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
- Division of Immune Regulation, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Masaki Maruta
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Kazushi Tanimoto
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Yukihiro Miyazaki
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Chika Iwamoto
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Takashi Saitou
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Takeshi Imamura
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Masaki Yasukawa
- Division of Immune Regulation, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- Ehime Prefectural University of Health Sciences, Tobe, Ehime, Japan
| | - Katsuto Takenaka
- Department of Hematology, Clinical Immunology and Infectious Diseases, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
25
|
Garg S, Ni W, Griffin JD, Sattler M. Chimeric Antigen Receptor T Cell Therapy in Acute Myeloid Leukemia: Trials and Tribulations. Hematol Rep 2023; 15:608-626. [PMID: 37987319 PMCID: PMC10660693 DOI: 10.3390/hematolrep15040063] [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: 07/18/2023] [Revised: 08/01/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematological malignancy that is often associated with relapse and drug resistance after standard chemotherapy or targeted therapy, particularly in older patients. Hematopoietic stem cell transplants are looked upon as the ultimate salvage option with curative intent. Adoptive cell therapy using chimeric antigen receptors (CAR) has shown promise in B cell malignancies and is now being investigated in AML. Initial clinical trials have been disappointing in AML, and we review current strategies to improve efficacy for CAR approaches. The extensive number of clinical trials targeting different antigens likely reflects the genetic heterogeneity of AML. The limited number of patients reported in multiple early clinical studies makes it difficult to draw conclusions about CAR safety, but it does suggest that the efficacy of this approach in AML lags behind the success observed in B cell malignancies. There is a clear need not only to improve CAR design but also to identify targets in AML that show limited expression in normal myeloid lineage cells.
Collapse
Affiliation(s)
- Swati Garg
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Ni
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - James D. Griffin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Sattler
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; (W.N.); (J.D.G.); (M.S.)
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
26
|
Dutta S, Box AC, Li Y, Sardiu ME. Identifying dynamical persistent biomarker structures for rare events using modern integrative machine learning approach. Proteomics 2023; 23:e2200290. [PMID: 36852539 PMCID: PMC11503472 DOI: 10.1002/pmic.202200290] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 01/30/2023] [Accepted: 02/17/2023] [Indexed: 03/01/2023]
Abstract
The evolution of omics and computational competency has accelerated discoveries of the underlying biological processes in an unprecedented way. High throughput methodologies, such as flow cytometry, can reveal deeper insights into cell processes, thereby allowing opportunities for scientific discoveries related to health and diseases. However, working with cytometry data often imposes complex computational challenges due to high-dimensionality, large size, and nonlinearity of the data structure. In addition, cytometry data frequently exhibit diverse patterns across biomarkers and suffer from substantial class imbalances which can further complicate the problem. The existing methods of cytometry data analysis either predict cell population or perform feature selection. Through this study, we propose a "wisdom of the crowd" approach to simultaneously predict rare cell populations and perform feature selection by integrating a pool of modern machine learning (ML) algorithms. Given that our approach integrates superior performing ML models across different normalization techniques based on entropy and rank, our method can detect diverse patterns existing across the model features. Furthermore, the method identifies a dynamic biomarker structure that divides the features into persistently selected, unselected, and fluctuating assemblies indicating the role of each biomarker in rare cell prediction, which can subsequently aid in studies of disease progression.
Collapse
Affiliation(s)
- Sreejata Dutta
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Andrew C. Box
- Stowers Institute for Medical Research, Kansas City, Missouri, USA
| | - Yanming Li
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Cancer Center, Kansas City, Kansas, USA
| | - Mihaela E. Sardiu
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Cancer Center, Kansas City, Kansas, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
27
|
Patel SA, Bello E, Wilks A, Gerber JM, Sadagopan N, Cerny J. Harnessing autologous immune effector mechanisms in acute myeloid leukemia: 2023 update of trials and tribulations. Leuk Res 2023; 134:107388. [PMID: 37729719 PMCID: PMC10947503 DOI: 10.1016/j.leukres.2023.107388] [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: 07/03/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/22/2023]
Abstract
Numerous recent advances have been made in therapeutic approaches toward acute myeloid leukemia (AML). Since 2017, we have seen eleven novel Food & Drug Administration (FDA)-approved medications for AML, all of which extend beyond the classical cytarabine-based cytostatic chemotherapy. In the recent two decades, the role of immune surveillance in AML has been intensively investigated. The power of one's own innate and adaptive immunity has been harnessed pharmacologically toward the goal of clearance of AML cells. Specifically, pre-clinical studies have shown great promise for antibodies that disinhibit T cells and macrophages by blocking checkpoint receptors within the immunologic synapse, thereby resulting in the elimination of AML cells. Anti-CD33 CAR-T therapies and anti-CD3/CD123 bispecific antibodies have also exhibited encouraging results in pre-clinical and early clinical studies. However, despite these translational efforts, we currently have no immune-based therapies for AML on the market, with the exception of gemtuzumab ozogamicin. In this focused review, we discuss molecular target validation and the most relevant clinical updates for immune-based experimental therapeutics including anti-CD47 monoclonal antibodies, CAR-T therapies, and bispecific T cell engagers. We highlight barriers to the clinical translation of these therapies in AML, and we propose solutions to optimize the manufacturing and delivery of the most novel immune-based therapies in the pipeline.
Collapse
Affiliation(s)
- Shyam A Patel
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA; Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA
| | - Elisa Bello
- UMass Chan Medical School, Worcester, MA, USA
| | - Andrew Wilks
- Dept. of Medicine - Division of Hematology and Medical Oncology, Boston Medical Center, Boston University School of Medicine, Boston, MA, USA
| | - Jonathan M Gerber
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA; Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA
| | - Narayanan Sadagopan
- MedStar Health - Georgetown/Washington Hospital Center Hematology and Medical Oncology, Washington, DC, USA
| | - Jan Cerny
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA; Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
28
|
Dutta S, Mudaranthakam DP, Li Y, Sardiu ME. PerSEveML: A Web-Based Tool to Identify Persistent Biomarker Structure for Rare Events Using Integrative Machine Learning Approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.25.564000. [PMID: 38196661 PMCID: PMC10775315 DOI: 10.1101/2023.10.25.564000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Omics datasets often pose a computational challenge due to their high dimensionality, large size, and non-linear structures. Analyzing these datasets becomes especially daunting in the presence of rare events. Machine learning (ML) methods have gained traction for analyzing rare events, yet there remains a limited exploration of bioinformatics tools that integrate ML techniques to comprehend the underlying biology. Expanding upon our previously developed computational framework of an integrative machine learning approach1, we introduce PerSEveML, an interactive web-based that uses crowd-sourced intelligence to predict rare events and determine feature selection structures. PerSEveML provides a comprehensive overview of the integrative approach through evaluation metrics that help users understand the contribution of individual ML methods to the prediction process. Additionally, PerSEveML calculates entropy and rank scores, which visually organize input features into a persistent structure of selected, unselected, and fluctuating categories that help researchers uncover meaningful hypotheses regarding the underlying biology. We have evaluated PerSEveML on three diverse biologically complex data sets with extremely rare events from small to large scale and have demonstrated its ability to generate valid hypotheses. PerSEveML is available at https://biostats-shinyr.kumc.edu/PerSEveML/ and https://github.com/sreejatadutta/PerSEveML.
Collapse
Affiliation(s)
- Sreejata Dutta
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Dinesh Pal Mudaranthakam
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Cancer Center, Kansas City, USA
| | - Yanming Li
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Cancer Center, Kansas City, USA
| | - Mihaela E Sardiu
- Department of Biostatistics & Data Science, University of Kansas Medical Center, Kansas City, Kansas, USA
- University of Kansas Cancer Center, Kansas City, USA
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
29
|
Mansour AG, Teng KY, Li Z, Zhu Z, Chen H, Tian L, Ali A, Zhang J, Lu T, Ma S, Lin CM, Caligiuri MA, Yu J. Off-the-shelf CAR-engineered natural killer cells targeting FLT3 enhance killing of acute myeloid leukemia. Blood Adv 2023; 7:6225-6239. [PMID: 37379267 PMCID: PMC10582841 DOI: 10.1182/bloodadvances.2022007405] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 06/14/2023] [Accepted: 06/16/2023] [Indexed: 06/30/2023] Open
Abstract
The majority of patients with acute myeloid leukemia (AML) succumb to the disease or its complications, especially among older patients. Natural killer (NK) cells have been shown to have antileukemic activity in patients with AML; however, to our knowledge, primary NK cells armed with a chimeric antigen receptor (CAR) targeting antigens associated with AML as an "off-the-shelf" product for disease control have not been explored. We developed frozen, off-the-shelf allogeneic human NK cells engineered with a CAR recognizing FLT3 and secreting soluble interleukin-15 (IL-15) (FLT3 CAR_sIL-15 NK) to improve in vivo NK cell persistence and T-cell activation. FLT3 CAR_sIL-15 NK cells had higher cytotoxicity and interferon gamma secretion against FLT3+ AML cell lines when compared with activated NK cells lacking an FLT3 CAR or soluble IL-15. Frozen and thawed allogeneic FLT3 CAR_sIL-15 NK cells prolonged survival of both the MOLM-13 AML model as well as an orthotopic patient-derived xenograft AML model when compared with control NK cells. FLT3 CAR_sIL-15 NK cells showed no cytotoxicity against healthy blood mononuclear cells or hematopoietic stem cells. Collectively, our data suggest that FLT3 is an AML-associated antigen that can be targeted by frozen, allogeneic, off-the-shelf FLT3 CAR_sIL-15 NK cells that may provide a novel approach for the treatment of AML.
Collapse
Affiliation(s)
- Anthony G. Mansour
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Kun-Yu Teng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Zhiyao Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Zheng Zhu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Hanyu Chen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Lei Tian
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Aliya Ali
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, CA
| | - Ting Lu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
| | - Chih-Min Lin
- Department of Cellular Immunotherapy GMP Manufacturing, City of Hope National Medical Center, Los Angeles, CA
| | - Michael A. Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA
- President, City of Hope National Medical Center, Los Angeles, CA
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA
- Department of Hematology & Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Center, Los Angeles, CA
| |
Collapse
|
30
|
Liberatore C, Di Ianni M. Novel Approaches to Treatment of Acute Myeloid Leukemia Relapse Post Allogeneic Stem Cell Transplantation. Int J Mol Sci 2023; 24:15019. [PMID: 37834466 PMCID: PMC10573608 DOI: 10.3390/ijms241915019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023] Open
Abstract
The management of patients with acute myeloid leukemia (AML) relapsed post allogeneic hematopoietic stem cell transplantation (HSCT) remains a clinical challenge. Intensive treatment approaches are limited by severe toxicities in the early post-transplantation period. Therefore, hypomethylating agents (HMAs) have become the standard therapeutic approach due to favorable tolerability. Moreover, HMAs serve as a backbone for additional anti-leukemic agents. Despite discordant results, the addition of donor lymphocytes infusions (DLI) generally granted improved outcomes with manageable GvHD incidence. The recent introduction of novel targeted drugs in AML gives the opportunity to add a third element to salvage regimens. Those patients harboring targetable mutations might benefit from IDH1/2 inhibitors Ivosidenib and Enasidenib as well as FLT3 inhibitors Sorafenib and Gilteritinib in combination with HMA and DLI. Conversely, patients lacking targetable mutations actually benefit from the addition of Venetoclax. A second HSCT remains a valid option, especially for fit patients and for those who achieve a complete disease response with salvage regimens. Overall, across studies, higher response rates and longer survival were observed in cases of pre-emptive intervention for molecular relapse. Future perspectives currently rely on the development of adoptive immunotherapeutic strategies mainly represented by CAR-T cells.
Collapse
Affiliation(s)
- Carmine Liberatore
- Hematology Unit, Department of Oncology and Hematology, Santo Spirito Hospital, 65124 Pescara, Italy;
| | - Mauro Di Ianni
- Hematology Unit, Department of Oncology and Hematology, Santo Spirito Hospital, 65124 Pescara, Italy;
- Department of Medicine and Sciences of Aging, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
31
|
Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023; 8:306. [PMID: 37591844 PMCID: PMC10435569 DOI: 10.1038/s41392-023-01521-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/19/2023] Open
Abstract
The immune-cell origin of hematologic malignancies provides a unique avenue for the understanding of both the mechanisms of immune responsiveness and immune escape, which has accelerated the progress of immunotherapy. Several categories of immunotherapies have been developed and are being further evaluated in clinical trials for the treatment of blood cancers, including stem cell transplantation, immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. These immunotherapies have shown the potential to induce long-term remission in refractory or relapsed patients and have led to a paradigm shift in cancer treatment with great clinical success. Different immunotherapeutic approaches have their advantages but also shortcomings that need to be addressed. To provide clinicians with timely information on these revolutionary therapeutic approaches, the comprehensive review provides historical perspectives on the applications and clinical considerations of the immunotherapy. Here, we first outline the recent advances that have been made in the understanding of the various categories of immunotherapies in the treatment of hematologic malignancies. We further discuss the specific mechanisms of action, summarize the clinical trials and outcomes of immunotherapies in hematologic malignancies, as well as the adverse effects and toxicity management and then provide novel insights into challenges and future directions.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
32
|
Yue S, An J, Zhang Y, Li J, Zhao C, Liu J, Liang L, Sun H, Xu Y, Zhong Z. Exogenous Antigen Upregulation Empowers Antibody Targeted Nanochemotherapy of Leukemia. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209984. [PMID: 37321606 DOI: 10.1002/adma.202209984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 05/12/2023] [Indexed: 06/17/2023]
Abstract
Acute myeloid leukemia (AML) is afflicted by a high-mortality rate and few treatment options. The lack of specific surface antigens severely hampers the development of targeted therapeutics and cell therapy. Here, it is shown that exogenous all-trans retinoic acid (ATRA) mediates selective and transient CD38 upregulation on leukemia cells by up to 20-fold, which enables high-efficiency targeted nanochemotherapy of leukemia with daratumumab antibody-directed polymersomal vincristine sulfate (DPV). Strikingly, treatment of two CD38-low expressing AML orthotopic models with ATRA and DPV portfolio strategies effectively eliminates circulating leukemia cells and leukemia invasion into bone marrow and organs, leading to exceptional survival benefits with 20-40% of mice becoming leukemia-free. The combination of exogenous CD38 upregulation and antibody-directed nanotherapeutics provides a unique and powerful targeted therapy for leukemia.
Collapse
Affiliation(s)
- Shujing Yue
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Yifan Zhang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Jiaying Li
- Department of Orthopaedic Surgery, Orthopaedic Institute, The First Affiliated Hospital, Soochow University, Suzhou, 215007, P. R. China
| | - Cenzhu Zhao
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Jingyi Liu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Lanlan Liang
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Huanli Sun
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, and State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
33
|
Gao D, Hong F, He A. The role of bone marrow microenvironment on CAR-T efficacy in haematologic malignancies. Scand J Immunol 2023; 98:e13273. [PMID: 39007933 DOI: 10.1111/sji.13273] [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: 11/18/2022] [Revised: 03/30/2023] [Accepted: 04/19/2023] [Indexed: 07/16/2024]
Abstract
In recent years, chimeric antigen receptor-T (CAR-T) cell therapy has emerged as a novel immunotherapy method. It has shown significant therapeutic efficacy in the treatment of haematological B cell malignancies. In particular, the CAR-T therapy targeting CD19 has yielded unprecedented efficacy for acute B-lymphocytic leukaemia (B-ALL) and non-Hodgkin's lymphoma (NHL). In haematologic malignancies, tumour stem cells are more prone to stay in the regulatory bone marrow (BM) microenvironment (called niches), which provides a protective environment against immune attack. However, how the BM microenvironment affects the anti-tumour efficacy of CAR-T cells and its underlying mechanism is worthy of attention. In this review, we discuss the role of the BM microenvironment on the efficacy of CAR-T in haematological malignancies and propose corresponding strategies to enhance the anti-tumour activity of CAR-T therapy.
Collapse
Affiliation(s)
- Dandan Gao
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Fei Hong
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Aili He
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
- National-Local Joint Engineering Research Center of Biodiagnostics & Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
34
|
Dagar G, Gupta A, Masoodi T, Nisar S, Merhi M, Hashem S, Chauhan R, Dagar M, Mirza S, Bagga P, Kumar R, Akil ASAS, Macha MA, Haris M, Uddin S, Singh M, Bhat AA. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med 2023; 21:449. [PMID: 37420216 PMCID: PMC10327392 DOI: 10.1186/s12967-023-04292-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Traditional cancer treatments use nonspecific drugs and monoclonal antibodies to target tumor cells. Chimeric antigen receptor (CAR)-T cell therapy, however, leverages the immune system's T-cells to recognize and attack tumor cells. T-cells are isolated from patients and modified to target tumor-associated antigens. CAR-T therapy has achieved FDA approval for treating blood cancers like B-cell acute lymphoblastic leukemia, large B-cell lymphoma, and multiple myeloma by targeting CD-19 and B-cell maturation antigens. Bi-specific chimeric antigen receptors may contribute to mitigating tumor antigen escape, but their efficacy could be limited in cases where certain tumor cells do not express the targeted antigens. Despite success in blood cancers, CAR-T technology faces challenges in solid tumors, including lack of reliable tumor-associated antigens, hypoxic cores, immunosuppressive tumor environments, enhanced reactive oxygen species, and decreased T-cell infiltration. To overcome these challenges, current research aims to identify reliable tumor-associated antigens and develop cost-effective, tumor microenvironment-specific CAR-T cells. This review covers the evolution of CAR-T therapy against various tumors, including hematological and solid tumors, highlights challenges faced by CAR-T cell therapy, and suggests strategies to overcome these obstacles, such as utilizing single-cell RNA sequencing and artificial intelligence to optimize clinical-grade CAR-T cells.
Collapse
Affiliation(s)
- Gunjan Dagar
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, 3050, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ravi Chauhan
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Manisha Dagar
- Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Sameer Mirza
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, 182320, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Mohammad Haris
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
| |
Collapse
|
35
|
Shao R, Li Z, Xin H, Jiang S, Zhu Y, Liu J, Huang R, Xu K, Shi X. Biomarkers as targets for CAR-T/NK cell therapy in AML. Biomark Res 2023; 11:65. [PMID: 37330575 PMCID: PMC10276424 DOI: 10.1186/s40364-023-00501-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/11/2023] [Indexed: 06/19/2023] Open
Abstract
The most common kind of acute leukemia in adults is acute myeloid leukemia (AML), which is often treated with induction chemotherapy regimens followed by consolidation or allogeneic hematopoietic stem cell transplantation (HSCT). However, some patients continue to develop relapsed or refractory AML (R/R-AML). Small molecular targeted drugs require long-time administration. Not all the patients hold molecular targets. Novel medicines are therefore needed to enhance treatment outcomes. T cells and natural killer (NK) cells engineered with chimeric antigen receptors (CARs) that target antigens associated with AML have recently been produced and are currently being tested in both pre-clinical and clinical settings. This review provides an overview of CAR-T/NK treatments for AML.
Collapse
Affiliation(s)
- Ruonan Shao
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Zijian Li
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Honglei Xin
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Suyu Jiang
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Yilin Zhu
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Jingan Liu
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Rong Huang
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China
| | - Kailin Xu
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Xiaofeng Shi
- Second Affiliated Hospital of Nanjing Medical University, No.262, North Zhongshan Road, Nanjing, 210003, Jiangsu, China.
| |
Collapse
|
36
|
Premnath N, Madanat YF. Novel Investigational Agents and Pathways That May Influence the Future Management of Acute Myeloid Leukemia. Cancers (Basel) 2023; 15:2958. [PMID: 37296920 PMCID: PMC10252053 DOI: 10.3390/cancers15112958] [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: 04/21/2023] [Revised: 05/24/2023] [Accepted: 05/27/2023] [Indexed: 06/12/2023] Open
Abstract
Acute Myeloid leukemia (AML) is a clinically heterogeneous disease with a 5-year overall survival of 32% between 2012 to 2018. The above number severely dwindles with age and adverse risk of disease, presenting opportunities for new drug development and is an area of dire unmet need. Basic science and clinical investigators across the world have been working on many new and old molecule formulations and combination strategies to improve outcomes in this disease. In this review, we discuss select promising novel agents in various stages of clinical development for patients with AML.
Collapse
Affiliation(s)
- Naveen Premnath
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA;
| | - Yazan F. Madanat
- Division of Hematology and Medical Oncology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75235, USA;
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
37
|
Atilla E, Benabdellah K. The Black Hole: CAR T Cell Therapy in AML. Cancers (Basel) 2023; 15:2713. [PMID: 37345050 DOI: 10.3390/cancers15102713] [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: 04/15/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Despite exhaustive studies, researchers have made little progress in the field of adoptive cellular therapies for relapsed/refractory acute myeloid leukemia (AML), unlike the notable uptake for B cell malignancies. Various single antigen-targeting chimeric antigen receptor (CAR) T cell Phase I trials have been established worldwide and have recruited approximately 100 patients. The high heterogeneity at the genetic and molecular levels within and between AML patients resembles a black hole: a great gravitational field that sucks in everything. One must consider the fact that only around 30% of patients show a response; there are, however, consequential off-tumor effects. It is obvious that a new point of view is needed to achieve more promising results. This review first introduces the unique therapeutic challenges of not only CAR T cells but also other adoptive cellular therapies in AML. Next, recent single-cell sequencing data for AML to assess somatically acquired alterations at the DNA, epigenetic, RNA, and protein levels are discussed to give a perspective on cellular heterogeneity, intercellular hierarchies, and the cellular ecosystem. Finally, promising novel strategies are summarized, including more sophisticated next-generation CAR T, TCR-T, and CAR NK therapies; the approaches with which to tailor the microenvironment and target neoantigens; and allogeneic approaches.
Collapse
Affiliation(s)
- Erden Atilla
- Fred Hutchinson Cancer Research Center, Clinical Research Division, 1100 Fairview Ave N, Seattle, WA 98109, USA
- GENYO Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer/University of Granada/Andalusian Regional Government, Health Sciences Technology Park, 18016 Granada, Spain
| | - Karim Benabdellah
- GENYO Centre for Genomics and Oncological Research, Genomic Medicine Department, Pfizer/University of Granada/Andalusian Regional Government, Health Sciences Technology Park, 18016 Granada, Spain
| |
Collapse
|
38
|
Yang W, Sun X, Liu S, Xu Y, Li Y, Huang X, Liu K, Mao L, Min S, Liu L, Li S, Zhu Y, Zhang Y, Xie X, Xu K, Sun C, Yan J, Li Z. TLR8 agonist Motolimod-induced inflammatory death for treatment of acute myeloid leukemia. Biomed Pharmacother 2023; 163:114759. [PMID: 37105077 DOI: 10.1016/j.biopha.2023.114759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 04/29/2023] Open
Abstract
The clinical treatment of AML is dominated by "7 + 3" therapy, but it often shows great toxicity and limited therapeutic efficacy in application. Therefore, it is urgent to develop novel therapeutic strategies to achieve safe and efficient treatment of AML. Small-molecule inhibitors have the characteristics of high specificity, low off-target toxicity and remarkable therapeutic effect, and are receiving more and more attention in tumor therapy. In this study, we screened a library of 1972 FDA-approved small molecular compounds for those that induced the inflammatory death of AML cells, among which the TLR8 agonist Motolimod (MTL) showed stronger anti-AML activity in the animal model but slight affection on normal lymphocytes in control mice. In terms of mechanism, cellular experiments in AML cell lines proved that TLR8 and LKB1/AMPK are the key distinct mechanisms for MTL triggered caspase-3-dependent cell death and the expression of a large number of inflammatory factors. In conclusion, our findings identified the immunoactivator MTL as a single agent exerting significant anti-AML activity in vitro and in vivo, with strong potential for clinical translation.
Collapse
Affiliation(s)
- Wei Yang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China.
| | - Xiongfei Sun
- Department of hematopathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, PR China; Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, PR China
| | - Shuai Liu
- Department of Laboratory, Shenzhen Samii International Medical Center (Shenzhen Fourth People's Hospital), Shenzhen 518118, PR China
| | - Ying Xu
- Department of hematopathology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, PR China
| | - Yunlei Li
- Department of Clinical Laboratory, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, PR China
| | - Xiaoru Huang
- Institute of Biomedical Engineering, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, PR China
| | - Kaiqing Liu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Longyi Mao
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Shasha Min
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Linjiang Liu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Shi Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Yuqi Zhu
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Yu Zhang
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Xina Xie
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China
| | - Kui Xu
- Institute of Biomedical Engineering, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, PR China
| | - Changqing Sun
- Department of Clinical Laboratory, Shenzhen Baoan Pure Traditional Chinese Medicine Hospital, Shenzhen 518126, PR China
| | - Jie Yan
- The Second Affiliated Hospital, The State Key Laboratory of Respiratory Disease, Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, PR China
| | - Zesong Li
- Guangdong Provincial Key Laboratory of Systems Biology and Synthetic Biology for Urogenital Tumors, Shenzhen Key Laboratory of Genitourinary Tumor, Department of Urology, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital (Shenzhen Institute of Translational Medicine). Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging. School of Biomedical Engineering, School of Medicine, Shenzhen University, Shenzhen 518060, China.
| |
Collapse
|
39
|
Yagyu S, Nakazawa Y. piggyBac-transposon-mediated CAR-T cells for the treatment of hematological and solid malignancies. Int J Clin Oncol 2023; 28:736-747. [PMID: 36859566 DOI: 10.1007/s10147-023-02319-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Accepted: 02/17/2023] [Indexed: 03/03/2023]
Abstract
Since the introduction of the use of chimeric antigen receptor T-cell therapy (CAR-T therapy) dramatically changed the therapeutic strategy for B cell tumors, various CAR-T cell products have been developed and applied to myeloid and solid tumors. Although viral vectors have been widely used to produce genetically engineered T cells, advances in genetic engineering have led to the development of methods for producing non-viral, gene-modified CAR-T cells. Recent progress has revealed that non-viral CAR-T cells have a significant impact not only on the simplicity of the production process and the accessibility of non-viral vectors but also on the function of the cells themselves. In this review, we focus on piggyBac-transposon-based CAR-T cells among non-viral, gene-modified CAR-T cells and discuss their characteristics, preclinical development, and recent clinical applications.
Collapse
Affiliation(s)
- Shigeki Yagyu
- Innovative Research and Liaison Organization, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan. .,Center for Advanced Research of Gene and Cell Therapy, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan. .,Department of Pediatrics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kawaramachihirokoji, Kamigyo-ku, Kyoto, Japan.
| | - Yozo Nakazawa
- Center for Advanced Research of Gene and Cell Therapy, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan.,Department of Pediatrics, Shinshu University School of Medicine, 3-1-1, Asahi, Matsumoto, Nagano, Japan.,Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1, Asahi, Matsumoto, Nagano, Japan
| |
Collapse
|
40
|
Labanieh L, Mackall CL. CAR immune cells: design principles, resistance and the next generation. Nature 2023; 614:635-648. [PMID: 36813894 DOI: 10.1038/s41586-023-05707-3] [Citation(s) in RCA: 175] [Impact Index Per Article: 175.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 01/04/2023] [Indexed: 02/24/2023]
Abstract
The remarkable clinical activity of chimeric antigen receptor (CAR) therapies in B cell and plasma cell malignancies has validated the use of this therapeutic class for liquid cancers, but resistance and limited access remain as barriers to broader application. Here we review the immunobiology and design principles of current prototype CARs and present emerging platforms that are anticipated to drive future clinical advances. The field is witnessing a rapid expansion of next-generation CAR immune cell technologies designed to enhance efficacy, safety and access. Substantial progress has been made in augmenting immune cell fitness, activating endogenous immunity, arming cells to resist suppression via the tumour microenvironment and developing approaches to modulate antigen density thresholds. Increasingly sophisticated multispecific, logic-gated and regulatable CARs display the potential to overcome resistance and increase safety. Early signs of progress with stealth, virus-free and in vivo gene delivery platforms provide potential paths for reduced costs and increased access of cell therapies in the future. The continuing clinical success of CAR T cells in liquid cancers is driving the development of increasingly sophisticated immune cell therapies that are poised to translate to treatments for solid cancers and non-malignant diseases in the coming years.
Collapse
Affiliation(s)
- Louai Labanieh
- Department of Bioengineering, Stanford University, Stanford, CA, USA.,Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA.,Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University, Stanford, CA, USA. .,Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA. .,Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, USA. .,Division of Blood and Marrow Transplantation and Cell Therapy, Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
41
|
Yan T, Zhu L, Chen J. Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment. Exp Hematol Oncol 2023; 12:14. [PMID: 36707873 PMCID: PMC9883880 DOI: 10.1186/s40164-023-00373-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
The past decade has witnessed ongoing progress in immune therapy to ameliorate human health. As an emerging technique, chimeric antigen receptor (CAR) T-cell therapy has the advantages of specific killing of cancer cells, a high remission rate of cancer-induced symptoms, rapid tumor eradication, and long-lasting tumor immunity, opening a new window for tumor treatment. However, challenges remain in CAR T-cell therapy for solid tumors due to target diversity, tumor heterogeneity, and the complex microenvironment. In this review, we have outlined the development of the CAR T-cell technique, summarized the current advances in tumor-associated antigens (TAAs), and highlighted the importance of tumor-specific antigens (TSAs) or neoantigens for solid tumors. We also addressed the challenge of the TAA binding domain in CARs to overcome off-tumor toxicity. Moreover, we illustrated the dominant tumor microenvironment (TME)-induced challenges and new strategies based on TME-associated antigens (TMAs) for solid tumor CAR T-cell therapy.
Collapse
Affiliation(s)
- Ting Yan
- grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| | - Lingfeng Zhu
- grid.443397.e0000 0004 0368 7493Department of Urology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| | - Jin Chen
- grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China ,grid.443397.e0000 0004 0368 7493Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| |
Collapse
|
42
|
Xi Y, Chenglong L, Rong Z, Wen W, Yu W, Jiao C, Juan H, Feifei C, Rong X, Tao J, Hui L, Xiaobing H. Chidamide-based 3-drug combination regimen reverses molecular relapse post transplantation in AML1-ETO-positive acute myeloid leukemia. Front Pharmacol 2023; 13:1059930. [PMID: 36712661 PMCID: PMC9880285 DOI: 10.3389/fphar.2022.1059930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/22/2022] [Indexed: 01/15/2023] Open
Abstract
Objective: We aimed to explore a new method to reverse early relapse in patients with AML1-ETO-positive acute myeloid cell transplantation. Methods: A chidamide-based 3-drug combination regimen was used in our center to treat patients with AML1-ETO-positive AML post transplantation but negative flow cytometry results. A retrospective analysis was performed of the survival rate and possible influencing factors of patients with relapse treated with this regimen in our center from January 2018 to January 2022. Results: The overall response rate was 95.8% (23/24), and the median number of treatment courses was 4 (range, 3-12 courses). The total molecular complete response (MCR) was 79.1% (19/24) after all treatments, and the molecular complete response was 37.5% (9/24) after one cycle of treatment but reached 58.3% (14/24) after four cycles; overall, the proportion of MCR increased gradually with the increase in treatment cycles. The projected 5-year overall survival rate was 73.9%. The projected 5-year leukemia-free survival rate was 64.8%, and the projected 1-year cumulative relapse rate was 35.5%. The incidence of grade II-IV graft-versus-host diseases (GVHD) was 29.2% (7/24), and that of grade III-IV GVHD was 20.8% (5/24), which could be effectively controlled by glucocorticoid therapy combined with calcineurin inhibitors The total incidence of chronic GVHD was 29.2% (7/24), and all cases were localized chronic GVHD. The total infection rate was 33.3% (8/24), mainly involving bacterial and fungal infections, and the incidence of life-threatening infections was 4.17% (1/24). The treatment-related mortality rate was 0%; and the total mortality rate was 20.8% (5/24). Nausea and vomiting, thrombocytopenia, and neutropenia were common adverse reactions, all of which were Common Terminology Criteria for Adverse Events grade 2-3 events and reversible after drug withdrawal. In terms of immunity, Th1 cell counts gradually increased, Th17 cell counts gradually decreased, and the Th1/Th17 ratio gradually increased after treatment. The CD8+ T lymphocyte count increased gradually, while the CD4+ T lymphocyte count did not change significantly. Conclusion: Our chidamide-based 3-drug combination regimen led to a high remission rate and tolerable adverse reactions in patients with AML1-ETO-positive post-transplant relapse, and most patients can achieve long-term survival with this regimen.
Collapse
Affiliation(s)
- Yang Xi
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China,*Correspondence: Huang Xiaobing,
| | - Li Chenglong
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China,*Correspondence: Huang Xiaobing,
| | - Zhang Rong
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wang Wen
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Wang Yu
- Sichuan Provincial People’s Hospital (Medical Group), Dongli Hospital, Chengdu, China
| | - Chen Jiao
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Huang Juan
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Che Feifei
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Xiao Rong
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Jiang Tao
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Li Hui
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Huang Xiaobing
- Sichuan Provincial People’s Hospital, Affliated Hospital of University of Electronic Science and Technology of China, Chengdu, China,*Correspondence: Huang Xiaobing,
| |
Collapse
|
43
|
Gołos A, Góra-Tybor J, Robak T. Experimental drugs in clinical trials for acute myeloid leukemia: innovations, trends, and opportunities. Expert Opin Investig Drugs 2023; 32:53-67. [PMID: 36669827 DOI: 10.1080/13543784.2023.2171860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is a heterogeneous disease characterized by many cytogenetic and molecular alterations. Due to better knowledge of the molecular basis of AML, many targeted therapies have been introduced and registered, e.g. FMS-like tyrosine kinase 3 inhibitors, isocitrate dehydrogenase 1/2 mutation inhibitors, and Bcl-2 inhibitor. Despite that, the cure for AML remains an unmet clinical need in most patients. AREAS COVERED The review aims to present new, not yet registered drugs for AML. We searched the English literature for articles concerning AML, targeted drugs, menin inhibitors, DOT1L, BET, IDH inhibitors, FLT3, hedgehog inhibitors, Polo-like kinase inhibitors, RNA splicing, and immune therapies via PubMed. Publications from January 2000 to August 2022 were scrutinized. Additional relevant publications were obtained by reviewing the references from the chosen articles and Google search. Conference proceedings from the previous 5 years of The American Society of Hematology, the European Hematology Association, and the American Society of Clinical Oncology were searched manually. Additional relevant publications were obtained by reviewing the references. EXPERT OPINION For several years, the therapeutic approach in AML has become more individualized. Novel groups of drugs give hope for greater curability. High response rates have agents that restore the activity of the p53 protein. In addition, agents that work independently of a particular mutation seem promising for AML without any known mutation.
Collapse
Affiliation(s)
- Aleksandra Gołos
- Department of Hematooncology, Copernicus Memorial Hospital, Lodz, Poland
| | - Joanna Góra-Tybor
- Department of Hematooncology, Copernicus Memorial Hospital, Lodz, Poland.,Department of Hematology, Medical University of Lodz, Lodz, Poland
| | - Tadeusz Robak
- Department of Hematology, Medical University of Lodz, Lodz, Poland.,Department of General Hematology, Copernicus Memorial Hospital, Lodz, Poland
| |
Collapse
|
44
|
Shahzad M, Nguyen A, Hussain A, Ammad-Ud-Din M, Faisal MS, Tariq E, Ali F, Butt A, Anwar I, Chaudhary SG, Lutfi F, Ahmed N, Singh AK, Hematti P, McGuirk JP, Mushtaq MU. Outcomes with chimeric antigen receptor t-cell therapy in relapsed or refractory acute myeloid leukemia: a systematic review and meta-analysis. Front Immunol 2023; 14:1152457. [PMID: 37168849 PMCID: PMC10164930 DOI: 10.3389/fimmu.2023.1152457] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/11/2023] [Indexed: 05/13/2023] Open
Abstract
Background We conducted a systematic review and meta-analysis to evaluate outcomes following chimeric antigen receptor T cell (CAR-T) therapy in relapsed/refractory acute myeloid leukemia (RR-AML). Methods We performed a literature search on PubMed, Cochrane Library, and Clinicaltrials.gov. After screening 677 manuscripts, 13 studies were included. Data was extracted following PRISMA guidelines. Pooled analysis was done using the meta-package by Schwarzer et al. Proportions with 95% confidence intervals (CI) were computed. Results We analyzed 57 patients from 10 clinical trials and 3 case reports. The pooled complete and overall response rates were 49.5% (95% CI 0.18-0.81, I2 =65%) and 65.2% (95% CI 0.36-0.91, I2 =57%). The pooled incidence of cytokine release syndrome, immune-effector cell associated neurotoxicity syndrome, and graft-versus-host disease was estimated as 54.4% (95% CI 0.17-0.90, I2 =77%), 3.9% (95% CI 0.00-0.19, I2 =22%), and 1.6% (95%CI 0.00-0.21, I2 =33%), respectively. Conclusion CAR-T therapy has demonstrated modest efficacy in RR-AML. Major challenges include heterogeneous disease biology, lack of a unique targetable antigen, and immune exhaustion.
Collapse
Affiliation(s)
- Moazzam Shahzad
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
- Moffitt Cancer Center, University of South Florida, Tampa, FL, United States
| | - Andrea Nguyen
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Ali Hussain
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | | | - Muhammad Salman Faisal
- Division of Hematology/Oncology, Roswell Park Cancer Institute Buffalo, NY, United States
| | - Ezza Tariq
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Fatima Ali
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Atif Butt
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Iqra Anwar
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Sibgha Gull Chaudhary
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Anurag K. Singh
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Peiman Hematti
- Division of Hematology/Oncology, University of Wisconsin School of Medicine & Public Health, Madison, WL, United States
| | - Joseph P. McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Muhammad Umair Mushtaq
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
- *Correspondence: Muhammad Umair Mushtaq,
| |
Collapse
|
45
|
Present and Future Role of Immune Targets in Acute Myeloid Leukemia. Cancers (Basel) 2022; 15:cancers15010253. [PMID: 36612249 PMCID: PMC9818182 DOI: 10.3390/cancers15010253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
It is now well known that the bone marrow (BM) cell niche contributes to leukemogenesis, but emerging data support the role of the complex crosstalk between AML cells and the BM microenvironment to induce a permissive immune setting that protects leukemic stem cells (LSCs) from therapy-induced death, thus favoring disease persistence and eventual relapse. The identification of potential immune targets on AML cells and the modulation of the BM environment could lead to enhanced anti-leukemic effects of drugs, immune system reactivation, and the restoration of AML surveillance. Potential targets and effectors of this immune-based therapy could be monoclonal antibodies directed against LSC antigens such as CD33, CD123, and CLL-1 (either as direct targets or via several bispecific T-cell engagers), immune checkpoint inhibitors acting on different co-inhibitory axes (alone or in combination with conventional AML drugs), and novel cellular therapies such as chimeric antigen receptor (CAR) T-cells designed against AML-specific antigens. Though dozens of clinical trials, mostly in phases I and II, are ongoing worldwide, results have still been negatively affected by difficulties in the identification of the optimal targets on LSCs.
Collapse
|
46
|
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
|
47
|
Chang YJ, Pei XY, Huang XJ. Haematopoietic stem-cell transplantation in China in the era of targeted therapies: current advances, challenges, and future directions. Lancet Haematol 2022; 9:e919-e929. [DOI: 10.1016/s2352-3026(22)00293-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 08/25/2022] [Accepted: 08/30/2022] [Indexed: 11/30/2022]
|
48
|
Asmamaw Dejenie T, Tiruneh G/Medhin M, Dessie Terefe G, Tadele Admasu F, Wale Tesega W, Chekol Abebe E. Current updates on generations, approvals, and clinical trials of CAR T-cell therapy. Hum Vaccin Immunother 2022; 18:2114254. [PMID: 36094837 DOI: 10.1080/21645515.2022.2114254] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a novel, customized immunotherapy that is considered a 'living' and self-replicating drug to treat cancer, sometimes resulting in a complete cure. CAR T-cells are manufactured through genetic engineering of T-cells by equipping them with CARs to detect and target antigen-expressing cancer cells. CAR is designed to have an ectodomain extracellularly, a transmembrane domain spanning the cell membrane, and an endodomain intracellularly. Since its first discovery, the CAR structure has evolved greatly, from the first generation to the fifth generation, to offer new therapeutic alternatives for cancer patients. This treatment has achieved long-term and curative therapeutic efficacy in multiple blood malignancies that nowadays profoundly change the treatment landscape of lymphoma, leukemia, and multiple myeloma. But CART-cell therapy is associated with several hurdles, such as limited therapeutic efficacy, little effect on solid tumors, adverse effects, expensive cost, and feasibility issues, hindering its broader implications.
Collapse
Affiliation(s)
- Tadesse Asmamaw Dejenie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Markeshaw Tiruneh G/Medhin
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gashaw Dessie Terefe
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Fitalew Tadele Admasu
- Department of Biochemistry, College of Medicine and Health Science Arbaminch University, Arbaminch, Ethiopia
| | - Wondwossen Wale Tesega
- Department of Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Endeshaw Chekol Abebe
- Department of Biochemistry, College of Medicine and Health Science Arbaminch University, Arbaminch, Ethiopia
| |
Collapse
|
49
|
Cui Q, Liang P, Dai H, Cui W, Cai M, Ding Z, Ma Q, Yin J, Li Z, Liu S, Kang L, Yao L, Cen J, Shen H, Zhu M, Yu L, Wu D, Tang X. Case report: CD38-directed CAR-T cell therapy: A novel immunotherapy targeting CD38- positive blasts overcomes TKI and chemotherapy resistance of myeloid chronic myeloid leukemia in blastic phase. Front Immunol 2022; 13:1012981. [PMID: 36524116 PMCID: PMC9744919 DOI: 10.3389/fimmu.2022.1012981] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/08/2022] [Indexed: 11/30/2022] Open
Abstract
Resistance to tyrosine kinase inhibitor (TKI) is a tough problem in the treatment of chronic myeloid leukemia in blastic phase (CML-BP), which was often associated with acquired mutations in the kinase domain and not eliminating the leukemic stem cells. The efficacy of TKI or combination with chemotherapy in CML-BP remains unsatisfactory. Chimeric antigen receptor T (CAR-T) cell immunotherapy may overcome TKI and chemotherapy resistance. However, lack of ideal targetable antigens is a major obstacle for treating patients with myeloid malignancies. CD38 is known to be expressed on most (acute myeloid leukemia) AML cells, and its lack of expression on hematopoietic stem cells renders it as a potential therapeutic target for myeloid CML-BP. We develop a CD38-directed CAR-T cell therapy for AML, and two patients with myeloid CML-BP were enrolled (NCT04351022). Two patients, harboring E255K and T315I mutation in the ABL kinase domain, respectively, were resistant to multiple TKIs (imatinib, dasatinib, nilotinib, and ponatinib) and intensive chemotherapy. The blasts in the bone marrow of two patients exhibited high expression of CD38. After tumor reduction chemotherapy and lymphodepletion chemotherapy, 1 × 107 CAR-T-38 cells per kilogram of body weight were administered. They achieved minimal residual disease-negative and BCR::ABL1-negative complete remission and experienced grade II cytokine release syndrome manifesting as fever. Our data highlighted that CAR-T-38 cell therapy may overcome TKI and chemotherapy resistance in patients with myeloid CML-BP.
Collapse
Affiliation(s)
- Qingya Cui
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Peiqi Liang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Haiping Dai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Wei Cui
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Mengjie Cai
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Zixuan Ding
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Qinfen Ma
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Jia Yin
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Zheng Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Sining Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Liqing Kang
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China,Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd. Shanghai, China
| | - Li Yao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Jiannong Cen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Hongjie Shen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Mingqing Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Lei Yu
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, China,Shanghai Unicar-Therapy Bio-Medicine Technology Co., Ltd. Shanghai, China,*Correspondence: Lei Yu, ; Depei Wu, ; Xiaowen Tang,
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China,*Correspondence: Lei Yu, ; Depei Wu, ; Xiaowen Tang,
| | - Xiaowen Tang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China,Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China,*Correspondence: Lei Yu, ; Depei Wu, ; Xiaowen Tang,
| |
Collapse
|
50
|
Qu C, Zhang H, Cao H, Tang L, Mo H, Liu F, Zhang L, Yi Z, Long L, Yan L, Wang Z, Zhang N, Luo P, Zhang J, Liu Z, Ye W, Liu Z, Cheng Q. Tumor buster - where will the CAR-T cell therapy 'missile' go? Mol Cancer 2022; 21:201. [PMID: 36261831 PMCID: PMC9580202 DOI: 10.1186/s12943-022-01669-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 11/10/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell (CAR-T cell) therapy based on gene editing technology represents a significant breakthrough in personalized immunotherapy for human cancer. This strategy uses genetic modification to enable T cells to target tumor-specific antigens, attack specific cancer cells, and bypass tumor cell apoptosis avoidance mechanisms to some extent. This method has been extensively used to treat hematologic diseases, but the therapeutic effect in solid tumors is not ideal. Tumor antigen escape, treatment-related toxicity, and the immunosuppressive tumor microenvironment (TME) limit their use of it. Target selection is the most critical aspect in determining the prognosis of patients receiving this treatment. This review provides a comprehensive summary of all therapeutic targets used in the clinic or shown promising potential. We summarize CAR-T cell therapies’ clinical trials, applications, research frontiers, and limitations in treating different cancers. We also explore coping strategies when encountering sub-optimal tumor-associated antigens (TAA) or TAA loss. Moreover, the importance of CAR-T cell therapy in cancer immunotherapy is emphasized.
Collapse
Affiliation(s)
- Chunrun Qu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Hui Cao
- Department of Psychiatry, The Second People's Hospital of Hunan Province, The Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China.,The School of Clinical Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haoyang Mo
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Fangkun Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Yi
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Lifu Long
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,XiangYa School of Medicine, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Luzhe Yan
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Nan Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.,One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou, Zhengzhou, Henan, China
| | - Weijie Ye
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|