1
|
Mancuso K, Barbato S, Talarico M, Tacchetti P, Zamagni E, Cavo M. Idecabtagene vicleucel (ide-cel) for the treatment of triple-class exposed relapsed and refractory multiple myeloma. Expert Opin Biol Ther 2025; 25:27-46. [PMID: 39651553 DOI: 10.1080/14712598.2024.2433518] [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: 08/07/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024]
Abstract
INTRODUCTION Modern anti-myeloma therapies have broken new ground in the treatment of the disease, and the incorporation of ide-cel in the treatment landscape represents one of the major scientific and clinical advances. AREAS COVERED Ide-cel was the first cell-based gene therapy approved for the treatment of triple-class exposed relapsed/refractory myeloma patients, showing impressive results, and demonstrating superiority over standard regimens in terms of efficacy, potential treatment-free intervals, and improved quality of life in heavily pretreated patients and in high-risk disease. This review summarizes the state-of-the-art of the most recent updates deriving from the use of ide-cel within ongoing, or upcoming, clinical trials, and from real-life experiences. EXPERT OPINION As the use of chimeric antigen receptor (CAR)-T therapy is likely to progressively increase over time and current indications expand to earlier treatment lines, efforts should be directed toward ameliorating overall management to facilitate proactive planning for treatment sequencing and provide adequate time for logistical planning. Importantly, the potential limited availability of CAR-T therapy highlights the importance of careful patient selection and coordination among centers. Meanwhile, attempts are underway to improve tolerance and reduce toxicity while enhancing anti-myeloma activity.
Collapse
Affiliation(s)
- Katia Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Simona Barbato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Marco Talarico
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Michele Cavo
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| |
Collapse
|
2
|
Sima H, Shao W. Advancements in the design and function of bispecific CAR-T cells targeting B Cell-Associated tumor antigens. Int Immunopharmacol 2024; 142:113166. [PMID: 39298818 DOI: 10.1016/j.intimp.2024.113166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Single-targeted CAR-T has exhibited notable success in treating B-cell tumors, effectively improving patient outcomes. However, the recurrence rate among patients remains above fifty percent, primarily attributed to antigen escape and the diminished immune persistence of CAR-T cells. Over recent years, there has been a surge of interest in bispecific CAR-T cell therapies, marked by an increasing number of research articles and clinical applications annually. This paper undertakes a comprehensive review of influential studies on the design of bispecific CAR-T in recent years, examining their impact on bispecific CAR-T efficacy concerning disease classification, targeted antigens, and CAR design. Notable distinctions in antigen targeting within B-ALL, NHL, and MM are explored, along with an analysis of how CAR scFv, transmembrane region, hinge region, and co-stimulatory region design influence Bi-CAR-T efficacy across different tumors. The summary provided aims to serve as a reference for designing novel and improved CAR-Ts, facilitating more efficient treatment for B-cell malignant tumors.
Collapse
Affiliation(s)
- Helin Sima
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
3
|
Sainatham C, Yadav D, Dilli Babu A, Tallapalli JR, Kanagala SG, Filippov E, Murillo Chavez F, Ahmed N, Lutfi F. The current socioeconomic and regulatory landscape of immune effector cell therapies. Front Med (Lausanne) 2024; 11:1462307. [PMID: 39697210 PMCID: PMC11652178 DOI: 10.3389/fmed.2024.1462307] [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: 07/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Immune cell effector therapies, including chimeric antigen receptor (CAR)-T cells, T-cell receptor (TCR) T cells, natural killer (NK) cells, and macrophage-based therapies, represent a transformative approach to cancer treatment, harnessing the immune system to target and eradicate malignant cells. CAR-T cell therapy, the most established among these, involves engineering T cells to express CARs specific to cancer cell antigens, showing remarkable efficacy in hematologic malignancies like leukemias, B-cell lymphomas, and multiple myeloma. Similarly, TCR-modified therapies, which reprogram T cells to recognize intracellular tumor antigens presented by major histocompatibility complex (MHC) molecules, offer promise for a range of solid tumors. NK-cell therapies leverage NK cells' innate cytotoxicity, providing an allogeneic approach that avoids some of the immune-related complications associated with T-cell-based therapies. Macrophage-based therapies, still in early stages of the development, focus on reprogramming macrophages to stimulate an immune response against cancer cells in the tumor microenvironment. Despite their promise, socioeconomic and regulatory challenges hinder the accessibility and scalability of immune cell effector therapies. These treatments are costly, with CAR-T therapies currently exceeding $400,000 per patient, creating significant disparities in access based on socioeconomic status and geographic location. The high manufacturing costs stem from the personalized, labor-intensive processes of harvesting, modifying, and expanding patients' cells. Moreover, complex logistics for manufacturing and delivering these therapies limit their reach, particularly in low-resource settings. Regulatory pathways further complicate the landscape. In the United States., the Food and Drug Administrations' (FDA) accelerated approval processes for cell-based therapies facilitate innovation but do not address cost-related barriers. In Europe, the European Medicines Agency (EMA) offers adaptive pathways, yet decentralized reimbursement systems create uneven access across member states. Additionally, differing regulatory standards for manufacturing and quality control worldwide pose hurdles for global harmonization and access. To expand the reach of immune effector cell therapies, a multipronged approach is needed-streamlined regulatory frameworks, policies to reduce treatment costs, and international collaborations to standardize manufacturing. Addressing these socioeconomic and regulatory obstacles is essential to make these life-saving therapies accessible to a broader patient population worldwide. We present a literature review on the current landscape of immune effector cell therapies and barriers of access to currently approved standard of care therapy at various levels.
Collapse
Affiliation(s)
- Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Aravind Dilli Babu
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Jayanth Reddy Tallapalli
- Division of Infectious Diseases, Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sai Gautham Kanagala
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital Center, New York, NY, United States
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Franco Murillo Chavez
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Nausheen Ahmed
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
4
|
Li S, Shi L, Zhao L, Guo Q, Li J, Liu ZL, Guo Z, Cao YJ. Split-design approach enhances the therapeutic efficacy of ligand-based CAR-T cells against multiple B-cell malignancies. Nat Commun 2024; 15:9751. [PMID: 39528513 PMCID: PMC11555413 DOI: 10.1038/s41467-024-54150-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
To address immune escape, multi-specific CAR-T-cell strategies use natural ligands that specifically bind multiple receptors on malignant cells. In this context, we propose a split CAR design comprising a universal receptor expressed on T cells and ligand-based switch molecules, which preserves the natural trimeric structure of ligands like APRIL and BAFF. Following optimization of the hinges and switch labeling sites, the split-design CAR-T cells ensure the native conformation of ligands, facilitating the optimal formation of immune synapses between target cancer cells and CAR-T cells. Our CAR-T-cell strategy demonstrates antitumor activities against various B-cell malignancy models in female mice, potentially preventing immune escape following conventional CAR-T-cell therapies in the case of antigen loss or switching. This ligand-based split CAR design introduces an idea for optimizing CAR recognition, enhancing efficacy and potentially improving safety in clinical translation, and may be broadly applicable to cellular therapies based on natural receptors or ligands.
Collapse
Affiliation(s)
- Shuhong Li
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Licai Shi
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Lijun Zhao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Qiaoru Guo
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China
| | - Jun Li
- Fundamenta Therapeutics Co., Ltd, Suzhou, Jiangsu, China
| | - Ze-Lin Liu
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Yu J Cao
- State Key Laboratory of Chemical Oncogenomics, Shenzhen Key Laboratory of Chemical Genomics, Peking University Shenzhen Graduate School, Shenzhen, Guangdong, China.
- Institute of Chemical Biology, Shenzhen Bay Laboratory, Shenzhen, China.
| |
Collapse
|
5
|
Zhou D, Sun Q, Xia J, Gu W, Qian J, Zhuang W, Yan Z, Cheng H, Chen W, Zhu F, Qi K, Li D, Sang W, Zhu L, Ma S, Li H, Zhang H, Qiu T, Yan D, Zhang Y, Peng S, Chang AH, Xu K, Li Z. Anti-BCMA/GPRC5D bispecific CAR T cells in patients with relapsed or refractory multiple myeloma: a single-arm, single-centre, phase 1 trial. Lancet Haematol 2024; 11:e751-e760. [PMID: 39059405 DOI: 10.1016/s2352-3026(24)00176-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND Some challenges still exist with single-target B-cell maturation antigen (BCMA) chimeric antigen receptor (CAR) T-cell therapies due to variable or negative BCMA expression, although they have yielded remarkable efficacy in relapsed or refractory multiple myeloma. We developed anti-BCMA/GPRC5D bispecific CARs to mitigate the limitations and potentiate the functions of CAR T cells. METHODS This single-arm, phase 1 trial was conducted at the Affiliated Hospital of Xuzhou Medical University (Xuzhou, China). The trial enrolled patients aged 18-75 years with relapsed or refractory multiple myeloma and an Eastern Cooperative Oncology Group performance status of 0-3. Anti-BCMA/GPRC5D bispecific CAR T cells were administered at 0·5 × 106, 1·0 × 106, 2·0 × 106, and 4·0 × 106 CAR T cells per kg in the dose-escalation phase, with additional patients included at the dose selected for the dose-expansion phase. The primary endpoint was safety, which included dose-limiting toxicity and maximum tolerated dose. Activity was also evaluated as a secondary endpoint. The maximum tolerated dose was chosen for the dose-expansion phase. Safety and activity analyses were done in all patients who received anti-BCMA/GPRC5D bispecific CAR T cells as defined in the protocol. This trial is registered with ClinicalTrials.gov (NCT05509530) and is complete. FINDINGS Between Sept 1, 2022, and Nov 3, 2023, 24 patients were enrolled and underwent apheresis. Three patients were excluded after apheresis (two patients discontinued due to rapid disease progression and one patient was withdrawn because of failed manufacture of CAR T cells), so 21 patients were infused with anti-BCMA/GPRC5D bispecific CAR T cells. Median follow-up was 5·8 months (IQR 5·2-6·7). Median age was 62 years (IQR 56-67). Eight (38%) patients were male, and 13 (62%) female. All patients were Chinese. At the 4·0 × 106 CAR T cells per kg dose, two patients had dose-limiting toxicities, of whom one died of subarachnoid haemorrhage (which was not considered to be related to the study treatment). The maximum tolerated dose was identified as 2·0 × 106 CAR T cells per kg. The most common grade 3 or worse adverse events were haematological toxicities in 19 (90%) patients (except lymphopenia). 15 (71%) patients had cytokine release syndrome, of which all cases were grade 1 or 2. One case of grade 1 immune effector cell-associated neurotoxicity syndrome (ICANS) was observed in a patient who received 4·0 × 106 CAR T cells per kg. No ICANS or grade 3 or worse organ toxicities were observed in patients who received 0·5-2·0 × 106 CAR T cells per kg. The overall response rate was 86% (18 of 21 patients), with 13 (62%) patients having a complete response or better, and 17 (81%) patients having measurable residual disease negativity. Of the 12 patients who received 2·0 × 106 CAR T cells per kg (three in the dose-escalation phase and an addition nine in the dose-expansion phase), the overall response rate was 92% (11 of 12 patients) with nine (75%) patients having a complete response or better. INTERPRETATION Anti-BCMA/GPRC5D bispecific CAR T cells show a good safety profile and encouraging activity in patients with relapsed or refractory multiple myeloma. FUNDING National Natural Science Foundation of China. TRANSLATION For the Chinese translation of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Dian Zhou
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Qian Sun
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Jieyun Xia
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Weiying Gu
- Department of Hematology, The First People's Hospital of Changzhou, Third Affiliated to Suzhou University, Changzhou, China
| | - Jun Qian
- Department of Hematology, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Wanchuan Zhuang
- Department of Hematology, Lianyungang Second People's Hospital affiliated to Bengbu Medical College, Lianyungang, China
| | - Zhiling Yan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Hai Cheng
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Wei Chen
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Feng Zhu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Kunming Qi
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Depeng Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Wei Sang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Lili Zhu
- Department of Pediatrics, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Sha Ma
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Hujun Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Huanxin Zhang
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Tingting Qiu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Dongmei Yan
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | | | | | - Alex H Chang
- Shanghai YaKe Biotechnology, Shanghai, China; Engineering Research Center of Gene Technology, Ministry of Education, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Kailin Xu
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China
| | - Zhenyu Li
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China; Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China; Jiangsu Key Laboratory of Bone Marrow Stem Cells, Xuzhou, China.
| |
Collapse
|
6
|
Schindler K, Ruppel KE, Müller C, Koehl U, Fricke S, Schmiedel D. Linker-specific monoclonal antibodies present a simple and reliable detection method for scFv-based CAR NK cells. Mol Ther Methods Clin Dev 2024; 32:101328. [PMID: 39286335 PMCID: PMC11403257 DOI: 10.1016/j.omtm.2024.101328] [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: 04/05/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024]
Abstract
Chimeric antigen receptor (CAR) T cell therapies have demonstrated significant successes in treating cancer. Currently, there are six approved CAR T cell products available on the market that target different malignancies of the B cell lineage. However, to overcome the limitations of CAR T cell therapies, other immune cells are being investigated for CAR-based cell therapies. CAR natural killer (NK) cells can be applied as allogeneic cell therapy, providing an economical, safe, and efficient alternative to autologous CAR T cells. To improve CAR research and future in-patient monitoring of cell therapeutics, a simple, reliable, and versatile CAR detection reagent is crucial. As most existing CARs contain a single-chain variable fragment (scFv) with either a Whitlow or a G4S linker site, linker-specific monoclonal antibodies (mAbs) can detect a broad range of CARs. This study demonstrates that these linker-specific mAbs can detect different CAR NK cells in vitro, spiked in whole blood, and within patient-derived tumor spheroids with high specificity and sensitivity, providing an effective and almost universal alternative for scFv-based CAR detection. Additionally, we confirm that linker-specific antibodies can be used for functional testing and enrichment of CAR NK cells, thereby providing a useful research tool to fast-track the development of novel CAR-based therapies.
Collapse
Affiliation(s)
- Katharina Schindler
- Department for Cell and Gene Therapy Development, Fraunhofer Institute
for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
| | - Katharina Eva Ruppel
- Department for Cell and Gene Therapy Development, Fraunhofer Institute
for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
| | - Claudia Müller
- Department of Preclinical Development and Validation, Fraunhofer
Institute for Cell Therapy and Immunology (IZI), 04103 Leipzig,
Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases, 60596
Frankfurt, Germany
| | - Ulrike Koehl
- Department for Cell and Gene Therapy Development, Fraunhofer Institute
for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
- Institute for Clinical Immunology, University of Leipzig, 04103 Leipzig,
Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases, 60596
Frankfurt, Germany
| | - Stephan Fricke
- Department for Cell and Gene Therapy Development, Fraunhofer Institute
for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
- Fraunhofer Cluster of Excellence Immune-Mediated Diseases, 60596
Frankfurt, Germany
- Medicine Campus MEDiC of the Technical University of Dresden at Klinikum
Chemnitz gGmbH, 09116 Chemnitz, Germany
| | - Dominik Schmiedel
- Department for Cell and Gene Therapy Development, Fraunhofer Institute
for Cell Therapy and Immunology (IZI), 04103 Leipzig, Germany
- Institute for Clinical Immunology, University of Leipzig, 04103 Leipzig,
Germany
| |
Collapse
|
7
|
Nasiri F, Asaadi Y, Mirzadeh F, Abdolahi S, Molaei S, Gavgani SP, Rahbarizadeh F. Updates on CAR T cell therapy in multiple myeloma. Biomark Res 2024; 12:102. [PMID: 39261906 PMCID: PMC11391811 DOI: 10.1186/s40364-024-00634-5] [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: 04/04/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024] Open
Abstract
Multiple myeloma (MM) is a hematological cancer characterized by the abnormal proliferation of plasma cells. Initial treatments often include immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and monoclonal antibodies (mAbs). Despite salient progress in diagnosis and treatment, most MM patients typically have a median life expectancy of only four to five years after starting treatment. In recent developments, the success of chimeric antigen receptor (CAR) T-cells in treating B-cell malignancies exemplifies a new paradigm shift in advanced immunotherapy techniques with promising therapeutic outcomes. Ide-cel and cilta-cel stand as the only two FDA-approved BCMA-targeted CAR T-cells for MM patients, a recognition achieved despite extensive preclinical and clinical research efforts in this domain. Challenges remain regarding certain aspects of CAR T-cell manufacturing and administration processes, including the lack of accessibility and durability due to T-cell characteristics, along with expensive and time-consuming processes limiting health plan coverage. Moreover, MM features, such as tumor antigen heterogeneity, antigen presentation alterations, complex tumor microenvironments, and challenges in CAR-T trafficking, contribute to CAR T-cell exhaustion and subsequent therapy relapse or refractory status. Additionally, the occurrence of adverse events such as cytokine release syndrome, neurotoxicity, and on-target, off-tumor toxicities present obstacles to CAR T-cell therapies. Consequently, ongoing CAR T-cell trials are diligently addressing these challenges and barriers. In this review, we provide an overview of the effectiveness of currently available CAR T-cell treatments for MM, explore the primary resistance mechanisms to these treatments, suggest strategies for improving long-lasting remissions, and investigate the potential for combination therapies involving CAR T-cells.
Collapse
Affiliation(s)
- Fatemeh Nasiri
- Department of Internal Medicine, College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Yasaman Asaadi
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Farzaneh Mirzadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Shahrokh Abdolahi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Sedigheh Molaei
- School of Medicine, Qom University of Medical Sciences, Qom, Iran
| | - Somayeh Piri Gavgani
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
8
|
Xiong Y, Libby KA, Su X. The physical landscape of CAR-T synapse. Biophys J 2024; 123:2199-2210. [PMID: 37715447 PMCID: PMC11331049 DOI: 10.1016/j.bpj.2023.09.004] [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: 07/05/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells form dynamic immunological synapses with their cancer cell targets. After a CAR-antigen engagement, the CAR-T synapse forms, matures, and finally disassembles, accompanied by substantial remodeling of cell surface proteins, lipids, and glycans. In this review, we provide perspectives for understanding protein distribution, membrane topology, and force transmission across the CAR-T synapse. We highlight the features of CAR-T synapses that differ from T cell receptor synapses, including the disorganized protein pattern, adjustable synapse width, diverse mechano-responding properties, and resulting signaling consequences. Through a range of examples, we illustrate how revealing the biophysical nature of the CAR-T synapse could guide the design of CAR-Ts with improved anti-tumor function.
Collapse
Affiliation(s)
- Yiwei Xiong
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut
| | - Kendra A Libby
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut; Yale Cancer Center, Yale University, New Haven, Connecticut; Yale Stem Cell Center, Yale University, New Haven, Connecticut.
| |
Collapse
|
9
|
Ng BD, Rajagopalan A, Kousa AI, Fischman JS, Chen S, Massa A, Elias HK, Manuele D, Galiano M, Lemarquis AL, Boardman AP, DeWolf S, Pierce J, Bogen B, James SE, van den Brink MRM. IL-18-secreting multiantigen targeting CAR T cells eliminate antigen-low myeloma in an immunocompetent mouse model. Blood 2024; 144:171-186. [PMID: 38579288 PMCID: PMC11302468 DOI: 10.1182/blood.2023022293] [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: 09/15/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/07/2024] Open
Abstract
ABSTRACT Multiple myeloma is a plasma cell malignancy that is currently incurable with conventional therapies. Following the success of CD19-targeted chimeric antigen receptor (CAR) T cells in leukemia and lymphoma, CAR T cells targeting B-cell maturation antigen (BCMA) more recently demonstrated impressive activity in relapsed and refractory myeloma patients. However, BCMA-directed therapy can fail due to weak expression of BCMA on myeloma cells, suggesting that novel approaches to better address this antigen-low disease may improve patient outcomes. We hypothesized that engineered secretion of the proinflammatory cytokine interleukin-18 (IL-18) and multiantigen targeting could improve CAR T-cell activity against BCMA-low myeloma. In a syngeneic murine model of myeloma, CAR T cells targeting the myeloma-associated antigens BCMA and B-cell activating factor receptor (BAFF-R) failed to eliminate myeloma when these antigens were weakly expressed, whereas IL-18-secreting CAR T cells targeting these antigens promoted myeloma clearance. IL-18-secreting CAR T cells developed an effector-like T-cell phenotype, promoted interferon-gamma production, reprogrammed the myeloma bone marrow microenvironment through type-I/II interferon signaling, and activated macrophages to mediate antimyeloma activity. Simultaneous targeting of weakly-expressed BCMA and BAFF-R with dual-CAR T cells enhanced T-cell:target-cell avidity, increased overall CAR signal strength, and stimulated antimyeloma activity. Dual-antigen targeting augmented CAR T-cell secretion of engineered IL-18 and facilitated elimination of larger myeloma burdens in vivo. Our results demonstrate that combination of engineered IL-18 secretion and multiantigen targeting can eliminate myeloma with weak antigen expression through distinct mechanisms.
Collapse
Affiliation(s)
- Brandon D. Ng
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pharmacology, Weill Cornell Medicine, New York, NY
| | - Adhithi Rajagopalan
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Anastasia I. Kousa
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Jacob S. Fischman
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Immunology Graduate Group, University of Pennsylvania, Philadelphia, PA
| | - Sophia Chen
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alyssa Massa
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
| | - Harold K. Elias
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
| | - Dylan Manuele
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Michael Galiano
- Molecular Cytology Core, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Andri L. Lemarquis
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander P. Boardman
- Lymphoma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Susan DeWolf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jonah Pierce
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY
| | | | - Scott E. James
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marcel R. M. van den Brink
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
- City of Hope Comprehensive Cancer Center, Duarte, CA
- Department of Immunology and Microbial Pathogenesis, Weill Cornell Medicine, New York, NY
- Department of Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY
- Bone Marrow Transplantation Service, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
10
|
Schlegel LS, Werbrouck C, Boettcher M, Schlegel P. Universal CAR 2.0 to overcome current limitations in CAR therapy. Front Immunol 2024; 15:1383894. [PMID: 38962014 PMCID: PMC11219820 DOI: 10.3389/fimmu.2024.1383894] [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: 02/08/2024] [Accepted: 06/03/2024] [Indexed: 07/05/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has effectively complemented the treatment of advanced relapsed and refractory hematological cancers. The remarkable achievements of CD19- and BCMA-CAR T therapies have raised high expectations within the fields of hematology and oncology. These groundbreaking successes are propelling a collective aspiration to extend the reach of CAR therapies beyond B-lineage malignancies. Advanced CAR technologies have created a momentum to surmount the limitations of conventional CAR concepts. Most importantly, innovations that enable combinatorial targeting to address target antigen heterogeneity, using versatile adapter CAR concepts in conjunction with recent transformative next-generation CAR design, offer the promise to overcome both the bottleneck associated with CAR manufacturing and patient-individualized treatment regimens. In this comprehensive review, we delineate the fundamental prerequisites, navigate through pivotal challenges, and elucidate strategic approaches, all aimed at paving the way for the future establishment of multitargeted immunotherapies using universal CAR technologies.
Collapse
Affiliation(s)
- Lara Sophie Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Coralie Werbrouck
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Michael Boettcher
- Department of Pediatric Surgery, University Medical Centre Mannheim, University of Heidelberg, Heidelberg, Germany
| | - Patrick Schlegel
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Pediatric Hematology and Oncology, Westmead Children’s Hospital, Sydney, NSW, Australia
| |
Collapse
|
11
|
Miller K, Hashmi H, Rajeeve S. Beyond BCMA: the next wave of CAR T cell therapy in multiple myeloma. Front Oncol 2024; 14:1398902. [PMID: 38800372 PMCID: PMC11116580 DOI: 10.3389/fonc.2024.1398902] [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: 03/11/2024] [Accepted: 04/24/2024] [Indexed: 05/29/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment landscape of relapsed/refractory multiple myeloma. The current Food and Drug Administration approved CAR T cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel both target B cell maturation antigen (BCMA), which is expressed on the surface of malignant plasma cells. Despite deep initial responses in most patients, relapse after anti-BCMA CAR T cell therapy is common. Investigations of acquired resistance to anti-BCMA CAR T cell therapy are underway. Meanwhile, other viable antigenic targets are being pursued, including G protein-coupled receptor class C group 5 member D (GPRC5D), signaling lymphocytic activation molecule family member 7 (SLAMF7), and CD38, among others. CAR T cells targeting these antigens, alone or in combination with anti-BCMA approaches, appear to be highly promising as they move from preclinical studies to early phase clinical trials. This review summarizes the current data with novel CAR T cell targets beyond BCMA that have the potential to enter the treatment landscape in the near future.
Collapse
Affiliation(s)
| | | | - Sridevi Rajeeve
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
12
|
Sheykhhasan M, Ahmadieh-Yazdi A, Vicidomini R, Poondla N, Tanzadehpanah H, Dirbaziyan A, Mahaki H, Manoochehri H, Kalhor N, Dama P. CAR T therapies in multiple myeloma: unleashing the future. Cancer Gene Ther 2024; 31:667-686. [PMID: 38438559 PMCID: PMC11101341 DOI: 10.1038/s41417-024-00750-2] [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: 08/12/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/06/2024]
Abstract
In recent years, the field of cancer treatment has witnessed remarkable breakthroughs that have revolutionized the landscape of care for cancer patients. While traditional pillars such as surgery, chemotherapy, and radiation therapy have long been available, a cutting-edge therapeutic approach called CAR T-cell therapy has emerged as a game-changer in treating multiple myeloma (MM). This novel treatment method complements options like autologous stem cell transplants and immunomodulatory medications, such as proteasome inhibitors, by utilizing protein complexes or anti-CD38 antibodies with potent complement-dependent cytotoxic effects. Despite the challenges and obstacles associated with these treatments, the recent approval of the second FDA multiple myeloma CAR T-cell therapy has sparked immense promise in the field. Thus far, the results indicate its potential as a highly effective therapeutic solution. Moreover, ongoing preclinical and clinical trials are exploring the capabilities of CAR T-cells in targeting specific antigens on myeloma cells, offering hope for patients with relapsed/refractory MM (RRMM). These advancements have shown the potential for CAR T cell-based medicines or combination therapies to elicit greater treatment responses and minimize side effects. In this context, it is crucial to delve into the history and functions of CAR T-cells while acknowledging their limitations. We can strategize and develop innovative approaches to overcome these barriers by understanding their challenges. This article aims to provide insights into the application of CAR T-cells in treating MM, shedding light on their potential, limitations, and strategies employed to enhance their efficacy.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Amirhossein Ahmadieh-Yazdi
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi, University of Medical Sciences, Yazd, Iran
| | - Rosario Vicidomini
- Section on Cellular Communication, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ashkan Dirbaziyan
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - Paola Dama
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|
13
|
Ünlü S, Sánchez Navarro BG, Cakan E, Berchtold D, Meleka Hanna R, Vural S, Vural A, Meisel A, Fichtner ML. Exploring the depths of IgG4: insights into autoimmunity and novel treatments. Front Immunol 2024; 15:1346671. [PMID: 38698867 PMCID: PMC11063302 DOI: 10.3389/fimmu.2024.1346671] [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: 11/29/2023] [Accepted: 02/29/2024] [Indexed: 05/05/2024] Open
Abstract
IgG4 subclass antibodies represent the rarest subclass of IgG antibodies, comprising only 3-5% of antibodies circulating in the bloodstream. These antibodies possess unique structural features, notably their ability to undergo a process known as fragment-antigen binding (Fab)-arm exchange, wherein they exchange half-molecules with other IgG4 antibodies. Functionally, IgG4 antibodies primarily block and exert immunomodulatory effects, particularly in the context of IgE isotype-mediated hypersensitivity reactions. In the context of disease, IgG4 antibodies are prominently observed in various autoimmune diseases combined under the term IgG4 autoimmune diseases (IgG4-AID). These diseases include myasthenia gravis (MG) with autoantibodies against muscle-specific tyrosine kinase (MuSK), nodo-paranodopathies with autoantibodies against paranodal and nodal proteins, pemphigus vulgaris and foliaceus with antibodies against desmoglein and encephalitis with antibodies against LGI1/CASPR2. Additionally, IgG4 antibodies are a prominent feature in the rare entity of IgG4 related disease (IgG4-RD). Intriguingly, both IgG4-AID and IgG4-RD demonstrate a remarkable responsiveness to anti-CD20-mediated B cell depletion therapy (BCDT), suggesting shared underlying immunopathologies. This review aims to provide a comprehensive exploration of B cells, antibody subclasses, and their general properties before examining the distinctive characteristics of IgG4 subclass antibodies in the context of health, IgG4-AID and IgG4-RD. Furthermore, we will examine potential therapeutic strategies for these conditions, with a special focus on leveraging insights gained from anti-CD20-mediated BCDT. Through this analysis, we aim to enhance our understanding of the pathogenesis of IgG4-mediated diseases and identify promising possibilities for targeted therapeutic intervention.
Collapse
Affiliation(s)
- Selen Ünlü
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Koç University School of Medicine, Istanbul, Türkiye
| | - Blanca G. Sánchez Navarro
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Elif Cakan
- Ragon Institute of Massachusetts General Hospital, Massachusetts Institute of Technology and Harvard University, Cambridge, MA, United States
| | - Daniel Berchtold
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Rafael Meleka Hanna
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Secil Vural
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Dermatology and Venereology, Koç University School of Medicine, İstanbul, Türkiye
| | - Atay Vural
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Neurology, Koç University School of Medicine, İstanbul, Türkiye
| | - Andreas Meisel
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Miriam L. Fichtner
- Koç University Research Center for Translational Medicine (KUTTAM), İstanbul, Türkiye
- Department of Neurology with Experimental Neurology, Integrated Myasthenia Gravis Center, Neuroscience Clinical Research Center, Charité Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
14
|
Mishra A, Maiti R, Mohan P, Gupta P. Antigen loss following CAR-T cell therapy: Mechanisms, implications, and potential solutions. Eur J Haematol 2024; 112:211-222. [PMID: 37705357 DOI: 10.1111/ejh.14101] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/05/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Chimeric Antigen Receptor T-cell (CAR-T cell) therapy has emerged as a groundbreaking immunotherapeutic approach for treating various hematological malignancies. CAR-T cells are engineered to express synthetic receptors that target specific antigens on cancer cells, leading to their eradication. While the therapy has shown remarkable efficacy, a significant challenge that has been observed in 30%-70% of patients showing recurrent disease is antigen loss or downregulation. We searched PubMed/MEDLINE, EMBASE, and Google scholar for articles on antigen loss/escape following Chimeric antigen receptor T-cell therapy in malignancies. Antigen loss refers to the loss or reduction in the expression of the target antigen on cancer cells, rendering CAR-T cells ineffective. This phenomenon poses a significant clinical concern, as it can lead to disease relapse and limited treatment options. This review explores the mechanisms underlying antigen loss following CAR-T cell therapy, its implications on treatment outcomes, and potential strategies to overcome the problem.
Collapse
Affiliation(s)
- Archana Mishra
- Department of Pharmacology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Rituparna Maiti
- Department of Pharmacology, All India Institute of Medical Sciences, Bhubaneswar, Odisha, India
| | - Prafull Mohan
- Clinical Pharmacologist, Armed Forces Medical Services, Guwahati, India
| | - Pooja Gupta
- Department of Pharmacology, All India Institute of Medical Sciences, Delhi, India
| |
Collapse
|
15
|
Scheller L, Tebuka E, Rambau PF, Einsele H, Hudecek M, Prommersberger SR, Danhof S. BCMA CAR-T cells in multiple myeloma-ready for take-off? Leuk Lymphoma 2024; 65:143-157. [PMID: 37997705 DOI: 10.1080/10428194.2023.2276676] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023]
Abstract
Although the approval of new drugs has improved the clinical outcome of multiple myeloma (MM), it was widely regarded as incurable over the past decades. However, recent advancements in groundbreaking immunotherapies, such as chimeric antigen receptor T cells (CAR-T), have yielded remarkable results in heavily pretreated relapse/refractory patients, instilling hope for a potential cure. CAR-T are genetically modified cells armed with a novel receptor to specifically recognize and kill tumor cells. Among the potential targets for MM, the B-cell maturation antigen (BCMA) stands out since it is highly and almost exclusively expressed on plasma cells. Here, we review the currently approved BCMA-directed CAR-T products and ongoing clinical trials in MM. Furthermore, we explore innovative approaches to enhance BCMA-directed CAR-T and overcome potential reasons for treatment failure. Additionally, we explore the side effects associated with these novel therapies and shed light on accessibility of CAR-T therapy around the world.
Collapse
Affiliation(s)
- Lukas Scheller
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
- Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Universitätsklinikum Würzburg, Würzburg, Germany
| | - Erius Tebuka
- Department of Pathology, Catholic University of Health and Allied Sciences (CUHAS), Mwanza, Tanzania
- Else-Kröner-Center Würzburg-Mwanza, Catholic University of Health and Allied Sciences (CUHAS), Mwanza, Tanzania
| | - Peter Fabian Rambau
- Department of Pathology, Catholic University of Health and Allied Sciences (CUHAS), Mwanza, Tanzania
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Sabrina Rebecca Prommersberger
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Larson RC, Kann MC, Graham C, Mount CW, Castano AP, Lee WH, Bouffard AA, Takei HN, Almazan AJ, Scarfó I, Berger TR, Schmidts A, Frigault MJ, Gallagher KME, Maus MV. Anti-TACI single and dual-targeting CAR T cells overcome BCMA antigen loss in multiple myeloma. Nat Commun 2023; 14:7509. [PMID: 37980341 PMCID: PMC10657357 DOI: 10.1038/s41467-023-43416-7] [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: 08/18/2023] [Accepted: 11/09/2023] [Indexed: 11/20/2023] Open
Abstract
Chimeric Antigen Receptor (CAR) T cells directed to B cell maturation antigen (BCMA) mediate profound responses in patients with multiple myeloma, but most patients do not achieve long-term complete remissions. In addition, recent evidence suggests that high-affinity binding to BCMA can result in on-target, off-tumor activity in the basal ganglia and can lead to fatal Parkinsonian-like disease. Here we develop CAR T cells against multiple myeloma using a binder to targeting transmembrane activator and CAML interactor (TACI) in mono and dual-specific formats with anti-BCMA. These CARs have robust, antigen-specific activity in vitro and in vivo. We also show that TACI RNA expression is limited in the basal ganglia, which may circumvent some of the toxicities recently reported with BCMA CARs. Thus, single-targeting TACI CARs may have a safer toxicity profile, whereas dual-specific BCMA-TACI CAR T cells have potential to avoid the antigen escape that can occur with single-antigen targeting.
Collapse
Affiliation(s)
- Rebecca C Larson
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Michael C Kann
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Charlotte Graham
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher W Mount
- Department of Pathology and Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Ana P Castano
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Won-Ho Lee
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Amanda A Bouffard
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Hana N Takei
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Antonio J Almazan
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Irene Scarfó
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Trisha R Berger
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea Schmidts
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew J Frigault
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Kathleen M E Gallagher
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA
| | - Marcela V Maus
- Cellular Immunotherapy Program, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Cancer Center, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
17
|
Yang Z, Wang Y. Clinical development of chimeric antigen receptor-T cell therapy for hematological malignancies. Chin Med J (Engl) 2023; 136:2285-2296. [PMID: 37358555 PMCID: PMC10538902 DOI: 10.1097/cm9.0000000000002549] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Indexed: 06/27/2023] Open
Abstract
ABSTRACT Cellular therapies have revolutionized the treatment of hematological malignancies since their conception and rapid development. Chimeric antigen receptor (CAR)-T cell therapy is the most widely applied cellular therapy. Since the Food and Drug Administration approved two CD19-CAR-T products for clinical treatment of relapsed/refractory acute lymphoblastic leukemia and diffuse large B cell lymphoma in 2017, five more CAR-T cell products were subsequently approved for treating multiple myeloma or B cell malignancies. Moreover, clinical trials of CAR-T cell therapy for treating other hematological malignancies are ongoing. Both China and the United States have contributed significantly to the development of clinical trials. However, CAR-T cell therapy has many limitations such as a high relapse rate, adverse side effects, and restricted availability. Various methods are being implemented in clinical trials to address these issues, some of which have demonstrated promising breakthroughs. This review summarizes developments in CAR-T cell trials and advances in CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhihuan Yang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, Tianjin Key Laboratory of Cell Therapy for Blood Disease, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | | |
Collapse
|
18
|
Liang T, Song Y, Gu L, Wang Y, Ma W. Insight into the Progress in CAR-T Cell Therapy and Combination with Other Therapies for Glioblastoma. Int J Gen Med 2023; 16:4121-4141. [PMID: 37720174 PMCID: PMC10503554 DOI: 10.2147/ijgm.s418837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 08/02/2023] [Indexed: 09/19/2023] Open
Abstract
Glioblastoma (GBM) is the most common malignant primary brain cancer in adults. It is always resistant to existing treatments, including surgical resection, postoperative radiotherapy, and chemotherapy, which leads to a dismal prognosis and a high relapse rate. Therefore, novel curative therapies are urgently needed for GBM. Chimeric antigen receptor T (CAR-T) cell therapy has significantly improved life expectancy for hematological malignancies patients, and thus it increases the interest in applying CAR-T cell therapy for solid tumors. In the recently published research, it is indicated that there are numerous obstacles to achieve clinical benefits for solid tumors, especially for GBM, because of GBM anatomical characteristics (the blood-brain barrier and suppressive tumor microenvironment) and the tumor heterogeneity. CAR-T cells are difficult to penetrate blood-brain barrier, and immunosuppressive tumor microenvironment (TME), which induces CAR-T cell exhaustion, impairs CAR-T cell therapy response. Moreover, under the pressure of CAR-T cell therapy, the tumor heterogeneity and tumor plasticity drive tumor evolution and therapy resistance, such as antigen escape. Nonetheless, scientists strive for strategies to overcome these hurdles, including novel CAR-T cell designs and regional delivery. For instance, the structure of multi-antigen-targeted CAR-T cells can enrich CAR-T accumulation in tumor TME and eliminate abundant tumor cells to avoid tumor antigen heterogeneity. Additionally, paired with an immune modifier and one or more stimulating domains, different generation of innovations in the structure and manufacturing of CAR-T cells have improved efficacy and persistence. While single CAR-T cell therapy receives limited clinical survival benefit. Compared with single CAR-T cell therapy, the combination therapies have supplemented the treatment paradigm. Combinatorial treatment methods consolidate the CAR-T cells efficacy by regulating the tumor microenvironment, optimizing the CAR structure, targeting the CAR-T cells to the tumor cells, reversing the tumor-immune escape mechanisms, and represent a promising avenue against GBM, based on multiple impressive research. Moreover, exciting results are also reported to be realized through combining effective therapies with CAR-T cells in preclinical and clinical trials samples, have aroused inspiration to explore the antitumor function of combination therapies. In summary, this study aims to summarize the limitation of CAR-T cell therapies and introduces novel strategies to enhance CAR-T cell function as well as prospect the potential of the therapeutic combination.
Collapse
Affiliation(s)
- Tingyu Liang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yixuan Song
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Lingui Gu
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
19
|
Fan J, Yu Y, Yan L, Yuan Y, Sun B, Yang D, Liu N, Guo J, Zhang J, Zhao X. GAS6-based CAR-T cells exhibit potent antitumor activity against pancreatic cancer. J Hematol Oncol 2023; 16:77. [PMID: 37475048 PMCID: PMC10357739 DOI: 10.1186/s13045-023-01467-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/15/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND The receptor tyrosine kinases TAM family (TYRO3, AXL, and MERTK) are highly expressed in multiple forms of cancer cells and tumor-associated macrophages and promote the development of cancers including pancreatic tumor. Targeting TAM receptors could be a promising therapeutic option. METHODS We designed a novel CAR based on the extracellular domain of growth arrest-specific protein 6 (GAS6), a natural ligand for all TAM members. The ability of CAR-T to kill pancreatic cancer cells is tested in vitro and in vivo, and the safety is evaluated in mice and nonhuman primate. RESULTS GAS6-CAR-T cells efficiently kill TAM-positive pancreatic cancer cell lines, gemcitabine-resistant cancer cells, and cancer stem-like cells in vitro. GAS6-CAR-T cells also significantly suppressed the growth of PANC1 xenografts and patient-derived xenografts in mice. Furthermore, these CAR-T cells did not induce obvious side effects in nonhuman primate or mice although the CAR was demonstrated to recognize mouse TAM. CONCLUSIONS Our findings indicate that GAS6-CAR-T-cell therapy may be effective for pancreatic cancers with low toxicity.
Collapse
Affiliation(s)
- Jiawei Fan
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Ye Yu
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Lanzhen Yan
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yuncang Yuan
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bin Sun
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Dong Yang
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Nan Liu
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jing Guo
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Jie Zhang
- Core Facilities of West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Xudong Zhao
- Division of Abdominal Tumor Multimodality Treatment and Laboratory of Animal Tumor Models, Cancer Center and State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics and Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
20
|
Baert L, Manfroi B, Quintero M, Chavarria O, Barbon PV, Clement E, Zeller A, Van Kuppevelt T, Sturm N, Moreaux J, Tveita A, Bogen B, McKee T, Huard B. 3-O sulfation of syndecan-1 mediated by the sulfotransferase HS3ST3a1 enhances myeloma aggressiveness. Matrix Biol 2023; 120:60-75. [PMID: 37201729 DOI: 10.1016/j.matbio.2023.05.005] [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: 12/05/2022] [Revised: 05/12/2023] [Accepted: 05/15/2023] [Indexed: 05/20/2023]
Abstract
Multiple myeloma is a hematological neoplasm derived from plasma cells invariably developing in the bone marrow (BM). The persisting clinical challenge in MM resides in its high ability to resist drugs as shown by the frequent relapses observed in patients regardless of the treatment applied. In a mouse model of MM, we identified a subpopulation of cells harboring increased resistance to current MM drugs. These cells bound a proliferation inducing ligand (APRIL), a key MM promoting/survival factor. APRIL binding involved the heparan sulfate (HS) chain present on syndecan-1 (SDC-1), and correlated with reactivity to the anti-HS antibody 10e4. 10e4+cells had a high proliferation activity, and were able to form colonies in 3-D cultures. 10e4+ cells were the only cells able to develop in BM after intravenous injection. They also resisted drugs in vivo, since their number increased after treatment in BM. Notably, 10e4+ cells differentiated into 10e4- cells upon in vitro and in vivo expansion. Expression of one sulfotransferase, HS3ST3a1, allowed modification of syndecan-1 to confer reactivity to 10e4 and binding to APRIL. HS3ST3a1 deletion inhibited tumorigenesis in BM. Notably, the two populations coexisted at a variable frequency in the BM of MM patients at diagnosis. In total, our results indicate that 3-O-sulfation on SDC-1 carried out by HS3ST3a1 defines aggressive MM cells, and that targeting of this enzyme could possibly be used to better control drug resistance.
Collapse
Affiliation(s)
- L Baert
- Institute for Advanced Biosciences, University Grenoble-Alpes, INSERM U1209, La Tronche, France
| | - B Manfroi
- Institute for Advanced Biosciences, University Grenoble-Alpes, INSERM U1209, La Tronche, France
| | - M Quintero
- translational innovation in medicine and complexity, University Grenoble-Alpes, CNRS UMR5525, La Tronche, France
| | - O Chavarria
- Institute for Advanced Biosciences, University Grenoble-Alpes, INSERM U1209, La Tronche, France
| | - P V Barbon
- Institute for Advanced Biosciences, University Grenoble-Alpes, INSERM U1209, La Tronche, France
| | - E Clement
- translational innovation in medicine and complexity, University Grenoble-Alpes, CNRS UMR5525, La Tronche, France
| | - A Zeller
- Department of Pathology and Immunology, university Hospitals, Geneva, Switzerland
| | - T Van Kuppevelt
- Rabdoud university medical center, Nijmegen, the Netherlands
| | - N Sturm
- translational innovation in medicine and complexity, University Grenoble-Alpes, CNRS UMR5525, La Tronche, France; Department of Pathology, university Hospital, Grenoble, France
| | - J Moreaux
- Department of Biological Hematology, University Hospital, Montpellier, France; Institute of Human Genetics, centre national de la recherche scientifique, University Montpellier, France
| | - A Tveita
- Department of Immunology and transfusion medicine, Institute for Immunology, university Hospital, Oslo, Norway
| | - B Bogen
- Department of Immunology and transfusion medicine, Institute for Immunology, university Hospital, Oslo, Norway; University of Oslo, Norway
| | - T McKee
- Department of clinical pathology, university Hospitals, Geneva, Switzerland
| | - B Huard
- translational innovation in medicine and complexity, University Grenoble-Alpes, CNRS UMR5525, La Tronche, France.
| |
Collapse
|
21
|
Lee L, Lim WC, Galas-Filipowicz D, Fung K, Taylor J, Patel D, Akbar Z, Alvarez Mediavilla E, Wawrzyniecka P, Shome D, Reijmers RM, Gregg T, Wood L, Day W, Cerec V, Ferrari M, Thomas S, Cordoba S, Onuoha S, Khokhar N, Peddareddigari V, Al-Hajj M, Cavet J, Zweegman S, Rodriguez-Justo M, Youg K, Pule M, Popat R. Limited efficacy of APRIL CAR in patients with multiple myeloma indicate challenges in the use of natural ligands for CAR T-cell therapy. J Immunother Cancer 2023; 11:e006699. [PMID: 37399355 DOI: 10.1136/jitc-2023-006699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND We used a proliferating ligand (APRIL) to construct a ligand-based third generation chimeric antigen receptor (CAR) able to target two myeloma antigens, B-cell maturation antigen (BCMA) and transmembrane activator and CAML interactor. METHODS The APRIL CAR was evaluated in a Phase 1 clinical trial (NCT03287804, AUTO2) in patients with relapsed, refractory multiple myeloma. Eleven patients received 13 doses, the first 15×106 CARs, and subsequent patients received 75,225,600 and 900×106 CARs in a 3+3 escalation design. RESULTS The APRIL CAR was well tolerated. Five (45.5%) patients developed Grade 1 cytokine release syndrome and there was no neurotoxicity. However, responses were only observed in 45.5% patients (1×very good partial response, 3×partial response, 1×minimal response). Exploring the mechanistic basis for poor responses, we then compared the APRIL CAR to two other BCMA CARs in a series of in vitro assays, observing reduced interleukin-2 secretion and lack of sustained tumor control by APRIL CAR regardless of transduction method or co-stimulatory domain. There was also impaired interferon signaling of APRIL CAR and no evidence of autoactivation. Thus focusing on APRIL itself, we confirmed similar affinity to BCMA and protein stability in comparison to BCMA CAR binders but reduced binding by cell-expressed APRIL to soluble BCMA and reduced avidity to tumor cells. This indicated either suboptimal folding or stability of membrane-bound APRIL attenuating CAR activation. CONCLUSIONS The APRIL CAR was well tolerated, but the clinical responses observed in AUTO2 were disappointing. Subsequently, when comparing the APRIL CAR to other BCMA CARs, we observed in vitro functional deficiencies due to reduced target binding by cell-expressed ligand.
Collapse
Affiliation(s)
- Lydia Lee
- Research Department of Haematology, UCL Cancer Institute, London, UK
| | | | | | - Kent Fung
- Research Department of Haematology, UCL Cancer Institute, London, UK
| | | | - Dominic Patel
- Department of Pathology, UCL Cancer Institute, London, UK
| | | | | | | | | | | | | | - Leigh Wood
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| | | | | | | | | | | | | | | | | | | | - Jim Cavet
- The Christie NHS Foundation Trust, Manchester, UK
| | | | | | - Kwee Youg
- Research Department of Haematology, UCL Cancer Institute, London, UK
| | - Martin Pule
- Research Department of Haematology, UCL Cancer Institute, London, UK
- Autolus Ltd, London, UK
| | - Rakesh Popat
- Department of Haematology, University College London Hospitals NHS Foundation Trust, London, UK
| |
Collapse
|
22
|
Yao L, Wang JT, Jayasinghe RG, O'Neal J, Tsai CF, Rettig MP, Song Y, Liu R, Zhao Y, Ibrahim OM, Fiala MA, Fortier JM, Chen S, Gehrs L, Rodrigues FM, Wendl MC, Kohnen D, Shinkle A, Cao S, Foltz SM, Zhou DC, Storrs E, Wyczalkowski MA, Mani S, Goldsmith SR, Zhu Y, Hamilton M, Liu T, Chen F, Vij R, Ding L, DiPersio JF. Single-Cell Discovery and Multiomic Characterization of Therapeutic Targets in Multiple Myeloma. Cancer Res 2023; 83:1214-1233. [PMID: 36779841 PMCID: PMC10102848 DOI: 10.1158/0008-5472.can-22-1769] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 12/10/2022] [Accepted: 02/07/2023] [Indexed: 02/14/2023]
Abstract
Multiple myeloma (MM) is a highly refractory hematologic cancer. Targeted immunotherapy has shown promise in MM but remains hindered by the challenge of identifying specific yet broadly representative tumor markers. We analyzed 53 bone marrow (BM) aspirates from 41 MM patients using an unbiased, high-throughput pipeline for therapeutic target discovery via single-cell transcriptomic profiling, yielding 38 MM marker genes encoding cell-surface proteins and 15 encoding intracellular proteins. Of these, 20 candidate genes were highlighted that are not yet under clinical study, 11 of which were previously uncharacterized as therapeutic targets. The findings were cross-validated using bulk RNA sequencing, flow cytometry, and proteomic mass spectrometry of MM cell lines and patient BM, demonstrating high overall concordance across data types. Independent discovery using bulk RNA sequencing reiterated top candidates, further affirming the ability of single-cell transcriptomics to accurately capture marker expression despite limitations in sample size or sequencing depth. Target dynamics and heterogeneity were further examined using both transcriptomic and immuno-imaging methods. In summary, this study presents a robust and broadly applicable strategy for identifying tumor markers to better inform the development of targeted cancer therapy. SIGNIFICANCE Single-cell transcriptomic profiling and multiomic cross-validation to uncover therapeutic targets identifies 38 myeloma marker genes, including 11 transcribing surface proteins with previously uncharacterized potential for targeted antitumor therapy.
Collapse
Affiliation(s)
- Lijun Yao
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Julia T. Wang
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Reyka G. Jayasinghe
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Julie O'Neal
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Chia-Feng Tsai
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Michael P. Rettig
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Yizhe Song
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Ruiyang Liu
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Yanyan Zhao
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Omar M. Ibrahim
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Mark A. Fiala
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Julie M. Fortier
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Siqi Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Leah Gehrs
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Fernanda Martins Rodrigues
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Michael C. Wendl
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Daniel Kohnen
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Andrew Shinkle
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Song Cao
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Steven M. Foltz
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Daniel Cui Zhou
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Erik Storrs
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Matthew A. Wyczalkowski
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Smrithi Mani
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Scott R. Goldsmith
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Ying Zhu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Mark Hamilton
- Multiple Myeloma Research Foundation, Norwalk, Connecticut
| | - Tao Liu
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington
| | - Feng Chen
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Ravi Vij
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| | - Li Ding
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
- McDonnell Genome Institute, Washington University in St. Louis, St. Louis, Missouri
| | - John F. DiPersio
- Department of Medicine, Washington University in St. Louis, St. Louis, Missouri
| |
Collapse
|
23
|
The BAFF-APRIL System in Cancer. Cancers (Basel) 2023; 15:cancers15061791. [PMID: 36980677 PMCID: PMC10046288 DOI: 10.3390/cancers15061791] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/13/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
B cell-activating factor (BAFF; also known as CD257, TNFSF13B, BLyS) and a proliferation-inducing ligand (APRIL; also known as CD256, TNFSF13) belong to the tumor necrosis factor (TNF) family. BAFF was initially discovered as a B-cell survival factor, whereas APRIL was first identified as a protein highly expressed in various cancers. These discoveries were followed by over two decades of extensive research effort, which identified overlapping signaling cascades between BAFF and APRIL, controlling immune homeostasis in health and driving pathogenesis in autoimmunity and cancer, the latter being the focus of this review. High levels of BAFF, APRIL, and their receptors have been detected in different cancers and found to be associated with disease severity and treatment response. Here, we have summarized the role of the BAFF-APRIL system in immune cell differentiation and immune tolerance and detailed its pathogenic functions in hematological and solid cancers. We also highlight the emerging therapeutics targeting the BAFF-APRIL system in different cancer types.
Collapse
|
24
|
Zhang X, Zhang H, Lan H, Wu J, Xiao Y. CAR-T cell therapy in multiple myeloma: Current limitations and potential strategies. Front Immunol 2023; 14:1101495. [PMID: 36891310 PMCID: PMC9986336 DOI: 10.3389/fimmu.2023.1101495] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Over the last decade, the survival outcome of patients with multiple myeloma (MM) has been substantially improved with the emergence of novel therapeutic agents, such as proteasome inhibitors, immunomodulatory drugs, anti-CD38 monoclonal antibodies, selective inhibitors of nuclear export (SINEs), and T cell redirecting bispecific antibodies. However, MM remains an incurable neoplastic plasma cell disorder, and almost all MM patients inevitably relapse due to drug resistance. Encouragingly, B cell maturation antigen (BCMA)-targeted chimeric antigen receptor T (CAR-T) cell therapy has achieved impressive success in the treatment of relapsed/refractory (R/R) MM and brought new hopes for R/R MM patients in recent years. Due to antigen escape, the poor persistence of CAR-T cells, and the complicated tumor microenvironment, a significant population of MM patients still experience relapse after anti-BCMA CAR-T cell therapy. Additionally, the high manufacturing costs and time-consuming manufacturing processes caused by the personalized manufacturing procedures also limit the broad clinical application of CAR-T cell therapy. Therefore, in this review, we discuss current limitations of CAR-T cell therapy in MM, such as the resistance to CAR-T cell therapy and the limited accessibility of CAR-T cell therapy, and summarize some optimization strategies to overcome these challenges, including optimizing CAR structure, such as utilizing dual-targeted/multi-targeted CAR-T cells and armored CAR-T cells, optimizing manufacturing processes, combing CAR-T cell therapy with existing or emerging therapeutic approaches, and performing subsequent anti-myeloma therapy after CAR-T cell therapy as salvage therapy or maintenance/consolidation therapy.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Huixuan Lan
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jinming Wu
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yang Xiao
- Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
25
|
Liu Z, Zhang J, Liu H, Shen H, Meng N, Qi X, Ding K, Song J, Fu R, Ding D, Feng G. BSA-AIE Nanoparticles with Boosted ROS Generation for Immunogenic Cell Death Immunotherapy of Multiple Myeloma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2208692. [PMID: 36529696 DOI: 10.1002/adma.202208692] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/15/2022] [Indexed: 06/17/2023]
Abstract
The main obstacle of multiple myeloma (MM) therapy is the compromised immune microenvironment, which leads to MM relapses and extramedullary disease progression. In this study, a novel strategy is reported of enhanced immunogenic cell death (ICD) immunotherapy with aggregation-induced emission (AIE) photosensitizer-loaded bovine serum albumin (BSA) nanoparticles (referred as BSA/TPA-Erdn), which can activate T cells, convert the cold tumor to hot, and reverse T cell senescence to restore the immune microenvironment for MM treatment. Loading AIE photosensitizer into the hydrophobic domain of BSA proteins significantly immobilizes the molecular geometry, which massively increases reactive oxygen species (ROS) generation and elicits a promising ICD immune response. Employing a NOD-SCID IL-2receptor gamma null mice model with MM patients' monocytes, it is shown that BSA/TPA-Erdn can simulate human dentric cell maturation, activate functional T lymphocytes, and increase additional polarization and differentiation signals to deliver a promising immunotherapy performance. Intriguingly, for the first time, it is shown that BSA/TPA-Erdn can greatly reverse T cell senescence, a main challenge in treating MM. Additionally, BSA/TPA-Erdn can effectively recruit more functional T lymphocytes into MM tumor. As a consequence, BSA/TPA-Erdn restores MM immune microenvironment and shows the best MM tumor eradication performance, which shall pave new insights for MM treatment in clinical practices.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jingtian Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive, Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Hongli Shen
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Nanhao Meng
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Xinwen Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive, Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Kai Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Jia Song
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive, Materials, Ministry of Education, and College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Guangxue Feng
- State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, AIE Institute, School of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| |
Collapse
|
26
|
Yang J, Zhou W, Li D, Niu T, Wang W. BCMA-targeting chimeric antigen receptor T-cell therapy for multiple myeloma. Cancer Lett 2023; 553:215949. [PMID: 36216149 DOI: 10.1016/j.canlet.2022.215949] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022]
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy, despite the development of numerous innovative therapies during the past two decades. Immunotherapies are changing the treatment paradigm of MM and have improved the overall response and survival of patients with relapsed/refractory (RR) MM. B cell maturation antigen (BCMA), selectively expressed in normal and malignant plasma cells, has been targeted by several immunotherapeutic modalities. Chimeric antigen receptor (CAR) T cells, the breakthrough in cancer immunotherapy, have revolutionized the treatment of B cell malignancies and remarkably improved the prognosis of RRMM. BCMA-targeting CAR T cell therapy is the most developed CAR T cell therapy for MM, and the US Food and Drug Administration has already approved idecabtagene vicleucel (Ide-cel) and ciltacabtagene autoleucel (Cilta-cel) for MM. However, the development of novel BCMA-targeting CAR T cell therapies remains in progress. This review focuses on BCMA-targeting CAR T cell therapy, covering all stages of investigational progress, including the innovative preclinical studies, the initial phase I clinical trials, and the more developed phase II clinical trials. It also discusses possible measures to improve the efficacy and safety of this therapy.
Collapse
Affiliation(s)
- Jinrong Yang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Weilin Zhou
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Dan Li
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| | - Wei Wang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
27
|
Ulbricht C, Cao Y, Niesner RA, Hauser AE. In good times and in bad: How plasma cells resolve stress for a life-long union with the bone marrow. Front Immunol 2023; 14:1112922. [PMID: 37033993 PMCID: PMC10080396 DOI: 10.3389/fimmu.2023.1112922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/14/2023] [Indexed: 04/11/2023] Open
Affiliation(s)
- Carolin Ulbricht
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Yu Cao
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
| | - Raluca A. Niesner
- Biophysical Analysis, Deutsches Rheuma-Forschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
- Dynamic and Functional in vivo Imaging, Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja E. Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Immune Dynamics, Deutsches Rheuma-Forschungszentrum (DRFZ), A Leibniz Institute, Berlin, Germany
- *Correspondence: Anja E. Hauser,
| |
Collapse
|
28
|
Banerjee R, Lee SS, Cowan AJ. Innovation in BCMA CAR-T therapy: Building beyond the Model T. Front Oncol 2022; 12:1070353. [PMID: 36505779 PMCID: PMC9729952 DOI: 10.3389/fonc.2022.1070353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
Autologous chimeric antigen receptor T-cell (CAR-T) therapies targeting B-cell maturation antigen (BCMA) have revolutionized the field of multiple myeloma in the same way that the Ford Model T revolutionized the original CAR world a century ago. However, we are only beginning to understand how to improve the efficacy and usability of these cellular therapies. In this review, we explore three automotive analogies for innovation with BCMA CAR-T therapies: stronger engines, better mileage, and hassle-free delivery. Firstly, we can build stronger engines in terms of BCMA targeting: improved antigen binding, tools to modulate antigen density, and armoring to better reach the antigen itself. Secondly, we can improve "mileage" in terms of response durability through ex vivo CAR design and in vivo immune manipulation. Thirdly, we can implement hassle-free delivery through rapid manufacturing protocols and off-the-shelf products. Just as the Model T set a benchmark for car manufacturing over 100 years ago, idecabtagene vicleucel and ciltacabtagene autoleucel have now set the starting point for BCMA CAR-T therapy with their approvals. As with any emerging technology, whether automotive or cellular, the best in innovation and optimization is yet to come.
Collapse
Affiliation(s)
- Rahul Banerjee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Sarah S. Lee
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Andrew J. Cowan
- Division of Medical Oncology, Department of Medicine, University of Washington, Seattle, WA, United States
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| |
Collapse
|
29
|
Camviel N, Wolf B, Croce G, Gfeller D, Zoete V, Arber C. Both APRIL and antibody-fragment-based CAR T cells for myeloma induce BCMA downmodulation by trogocytosis and internalization. J Immunother Cancer 2022; 10:jitc-2022-005091. [PMID: 36323436 PMCID: PMC9639149 DOI: 10.1136/jitc-2022-005091] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T cell therapy targeting B cell maturation antigen (BCMA) on multiple myeloma (MM) produces fast but not long-lasting responses. Reasons for treatment failure are poorly understood. CARs simultaneously targeting two antigens may represent an alternative. Here, we (1) designed and characterized novel A proliferation inducing ligand (APRIL) based dual-antigen targeting CARs, and (2) investigated mechanisms of resistance to CAR T cells with three different BCMA-binding moieties (APRIL, single-chain-variable-fragment, heavy-chain-only). METHODS Three new APRIL-CARs were designed and characterized. Human APRIL-CAR T cells were evaluated for their cytotoxic function in vitro and in vivo, for their polyfunctionality, immune synapse formation, memory, exhaustion phenotype and tonic signaling activity. To investigate resistance mechanisms, we analyzed BCMA levels and cellular localization and quantified CAR T cell-target cell interactions by live microscopy. Impact on pathway activation and tumor cell proliferation was assessed in vitro and in vivo. RESULTS APRIL-CAR T cells in a trimeric ligand binding conformation conferred fast but not sustained antitumor responses in vivo in mouse xenograft models. In vitro trimer-BBζ CAR T cells were more polyfunctional and formed stronger immune synapses than monomer-BBζ CAR T cells. After CAR T cell-myeloma cell contact, BCMA was rapidly downmodulated on target cells with all evaluated binding moieties. CAR T cells acquired BCMA by trogocytosis, and BCMA on MM cells was rapidly internalized. Since BCMA can be re-expressed during progression and persisting CAR T cells may not protect patients from relapse, we investigated whether non-functional CAR T cells play a role in tumor progression. While CAR T cell-MM cell interactions activated BCMA pathway, we did not find enhanced tumor growth in vitro or in vivo. CONCLUSION Antitumor responses with APRIL-CAR T cells were fast but not sustained. Rapid BCMA downmodulation occurred independently of whether an APRIL or antibody-based binding moiety was used. BCMA internalization mostly contributed to this effect, but trogocytosis by CAR T cells was also observed. Our study sheds light on the mechanisms underlying CAR T cell failure in MM when targeting BCMA and can inform the development of improved treatment strategies.
Collapse
Affiliation(s)
- Nicolas Camviel
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| | - Benita Wolf
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| | - Giancarlo Croce
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - David Gfeller
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Vincent Zoete
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland,Swiss Institute of Bioinformatics (SIB), Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL CHUV, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland,Ludwig Institute for Cancer Research Lausanne Branch, Lausanne, Switzerland
| |
Collapse
|
30
|
Fichtner ML, Hoehn KB, Ford EE, Mane-Damas M, Oh S, Waters P, Payne AS, Smith ML, Watson CT, Losen M, Martinez-Martinez P, Nowak RJ, Kleinstein SH, O'Connor KC. Reemergence of pathogenic, autoantibody-producing B cell clones in myasthenia gravis following B cell depletion therapy. Acta Neuropathol Commun 2022; 10:154. [PMID: 36307868 PMCID: PMC9617453 DOI: 10.1186/s40478-022-01454-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 09/30/2022] [Indexed: 11/12/2022] Open
Abstract
Myasthenia gravis (MG) is an autoantibody-mediated autoimmune disorder of the neuromuscular junction. A small subset of patients (<10%) with MG, have autoantibodies targeting muscle-specific tyrosine kinase (MuSK). MuSK MG patients respond well to CD20-mediated B cell depletion therapy (BCDT); most achieve complete stable remission. However, relapse often occurs. To further understand the immunomechanisms underlying relapse, we studied autoantibody-producing B cells over the course of BCDT. We developed a fluorescently labeled antigen to enrich for MuSK-specific B cells, which was validated with a novel Nalm6 cell line engineered to express a human MuSK-specific B cell receptor. B cells (≅ 2.6 million) from 12 different samples collected from nine MuSK MG patients were screened for MuSK specificity. We successfully isolated two MuSK-specific IgG4 subclass-expressing plasmablasts from two of these patients, who were experiencing a relapse after a BCDT-induced remission. Human recombinant MuSK mAbs were then generated to validate binding specificity and characterize their molecular properties. Both mAbs were strong MuSK binders, they recognized the Ig1-like domain of MuSK, and showed pathogenic capacity when tested in an acetylcholine receptor (AChR) clustering assay. The presence of persistent clonal relatives of these MuSK-specific B cell clones was investigated through B cell receptor repertoire tracing of 63,977 unique clones derived from longitudinal samples collected from these two patients. Clonal variants were detected at multiple timepoints spanning more than five years and reemerged after BCDT-mediated remission, predating disease relapse by several months. These findings demonstrate that a reservoir of rare pathogenic MuSK autoantibody-expressing B cell clones survive BCDT and reemerge into circulation prior to manifestation of clinical relapse. Overall, this study provides both a mechanistic understanding of MuSK MG relapse and a valuable candidate biomarker for relapse prediction.
Collapse
Affiliation(s)
- Miriam L Fichtner
- Department of Neurology, Yale University School of Medicine, 300 George Street - Room 353J, New Haven, CT, 06511, USA
- Department of Immunobiology, Yale University School of Medicine, 300 George Street - Room 353J, New Haven, CT, 06511, USA
| | - Kenneth B Hoehn
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Easton E Ford
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Marina Mane-Damas
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Sangwook Oh
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Waters
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Aimee S Payne
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Melissa L Smith
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Corey T Watson
- Department of Microbiology and Immunology, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, USA
| | - Mario Losen
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Pilar Martinez-Martinez
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
| | - Richard J Nowak
- Department of Neurology, Yale University School of Medicine, 300 George Street - Room 353J, New Haven, CT, 06511, USA
| | - Steven H Kleinstein
- Department of Immunobiology, Yale University School of Medicine, 300 George Street - Room 353J, New Haven, CT, 06511, USA
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Program in Computational Biology & Bioinformatics, Yale University, New Haven, CT, USA
| | - Kevin C O'Connor
- Department of Neurology, Yale University School of Medicine, 300 George Street - Room 353J, New Haven, CT, 06511, USA.
- Department of Immunobiology, Yale University School of Medicine, 300 George Street - Room 353J, New Haven, CT, 06511, USA.
| |
Collapse
|
31
|
Engineering T-cells with chimeric antigen receptors to combat hematological cancers: an update on clinical trials. Cancer Immunol Immunother 2022; 71:2301-2311. [PMID: 35199207 PMCID: PMC9463290 DOI: 10.1007/s00262-022-03163-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 01/24/2022] [Indexed: 11/25/2022]
Abstract
Chimeric antigen receptor (CAR) redirected T-cells has shown efficacy in the treatment of B-cell leukemia/lymphoma, however, high numbers of relapses occur due to loss of targeted antigen or intrinsic failure of the CAR T-cells. In this situation modifications of the basic strategy are envisaged to reduce the risk of relapse, some of them are in early clinical exploration. These include simultaneous targeting of multiple antigens or combination of CAR T-cell therapy with other treatment modalities such as checkpoint inhibitors. The review evaluates and discusses these modified advanced therapies and pre-clinical approaches with respect to their potential to control leukemia and lymphoma in the long-term.
Collapse
|
32
|
Mazinani M, Rahbarizadeh F. CAR-T cell potency: from structural elements to vector backbone components. Biomark Res 2022; 10:70. [PMID: 36123710 PMCID: PMC9487061 DOI: 10.1186/s40364-022-00417-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy, in which a patient’s own T lymphocytes are engineered to recognize and kill cancer cells, has achieved remarkable success in some hematological malignancies in preclinical and clinical trials, resulting in six FDA-approved CAR-T products currently available in the market. Once equipped with a CAR construct, T cells act as living drugs and recognize and eliminate the target tumor cells in an MHC-independent manner. In this review, we first described all structural modular of CAR in detail, focusing on more recent findings. We then pointed out behind-the-scene elements contributing to CAR expression and reviewed how CAR expression can be drastically affected by the elements embedded in the viral vector backbone.
Collapse
Affiliation(s)
- Marzieh Mazinani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran. .,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
33
|
Ramírez-Chacón A, Betriu-Méndez S, Bartoló-Ibars A, González A, Martí M, Juan M. Ligand-based CAR-T cell: Different strategies to drive T cells in future new treatments. Front Immunol 2022; 13:932559. [PMID: 36172370 PMCID: PMC9511026 DOI: 10.3389/fimmu.2022.932559] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-based therapies are presented as innovative treatments for multiple malignancies. Despite their clinical success, there is scientific evidence of the limitations of these therapies mainly due to immunogenicity issues, toxicities associated with the infusion of the product, and relapses of the tumor. As a result, novel approaches are appearing aiming to solve and/or mitigate the harmful effects of CAR-T therapies. These include strategies based on the use of ligands as binding moieties or ligand-based CAR-T cells. Several proposals are currently under development, with some undergoing clinical trials to assess their potential benefits. In addition to these, therapies such as chimeric autoantibody receptor (CAAR), B-cell receptor antigen for reverse targeting (BAR), and even chimeric human leukocyte antigen (HLA) antibody receptor (CHAR) have emerged, benefiting from the advantages of antigenic ligands as antibody-binding motifs. This review focuses on the potential role that ligands can play in current and future antitumor treatments and in other types of diseases, such as autoimmune diseases or problems associated with transplantation.
Collapse
Affiliation(s)
- Alejandro Ramírez-Chacón
- Immunology Unit, Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Laboratory of Cellular Immunology, Institute of Biotechnology and Biomedicine (IBB), Cerdanyola del Vallès, Spain
| | - Sergi Betriu-Méndez
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
| | - Ariadna Bartoló-Ibars
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
| | - Azucena González
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
- Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mercè Martí
- Immunology Unit, Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Spain
- Laboratory of Cellular Immunology, Institute of Biotechnology and Biomedicine (IBB), Cerdanyola del Vallès, Spain
| | - Manel Juan
- Immunology Department, Hospital Clínic de Barcelona, Centre de Diagnòstic Biomèdic (CDB), Barcelona, Spain
- Immunology Department, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) – Fundació Clínic per a la Recerca Biomèdica (FCRB) Universitat de Barcelona (UB), Barcelona, Spain
- Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- *Correspondence: Manel Juan,
| |
Collapse
|
34
|
Stern LA, Gholamin S, Moraga I, Yang X, Saravanakumar S, Cohen JR, Starr R, Aguilar B, Salvary V, Hibbard JC, Kalbasi A, Shepphird JK, O’Hearn J, Garcia KC, Brown CE. Engineered IL13 variants direct specificity of IL13Rα2-targeted CAR T cell therapy. Proc Natl Acad Sci U S A 2022; 119:e2112006119. [PMID: 35939683 PMCID: PMC9388138 DOI: 10.1073/pnas.2112006119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/03/2022] [Indexed: 11/28/2022] Open
Abstract
IL13Rα2 is an attractive target due to its overexpression in a variety of cancers and rare expression in healthy tissue, motivating expansion of interleukin 13 (IL13)-based chimeric antigen receptor (CAR) T cell therapy from glioblastoma into systemic malignancies. IL13Rα1, the other binding partner of IL13, is ubiquitously expressed in healthy tissue, raising concerns about the therapeutic window of systemic administration. IL13 mutants with diminished binding affinity to IL13Rα1 were previously generated by structure-guided protein engineering. In this study, two such variants, termed C4 and D7, are characterized for their ability to mediate IL13Rα2-specific response as binding domains for CAR T cells. Despite IL13Rα1 and IL13Rα2 sharing similar binding interfaces on IL13, mutations to IL13 that decrease binding affinity for IL13Rα1 did not drastically change binding affinity for IL13Rα2. Micromolar affinity to IL13Rα1 was sufficient to pacify IL13-mutein CAR T cells in the presence of IL13Rα1-overexpressing cells in vitro. Interestingly, effector activity of D7 CAR T cells, but not C4 CAR T cells, was demonstrated when cocultured with IL13Rα1/IL4Rα-coexpressing cancer cells. While low-affinity interactions with IL13Rα1 did not result in observable toxicities in mice, in vivo biodistribution studies demonstrated that C4 and D7 CAR T cells were better able to traffic away from IL13Rα1+ lung tissue than were wild-type (WT) CAR T cells. These results demonstrate the utility of structure-guided engineering of ligand-based binding domains with appropriate selectivity while validating IL13-mutein CARs with improved selectivity for application to systemic IL13Rα2-expressing malignancies.
Collapse
Affiliation(s)
- Lawrence A. Stern
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Sharareh Gholamin
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Division of Biology and Bioengineering, California Institute of Technology, Pasadena, CA 91125
| | - Ignacio Moraga
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345
| | - Xin Yang
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Supraja Saravanakumar
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Joseph R. Cohen
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Renate Starr
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Brenda Aguilar
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Vanessa Salvary
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Jonathan C. Hibbard
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - Anusha Kalbasi
- Department of Radiation Oncology, Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, CA 90024
| | - Jennifer K. Shepphird
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - James O’Hearn
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| | - K. Christopher Garcia
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305-5345
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA 94305-5345
- HHMI, Stanford University, Stanford, CA 94305-5345
- School of Medicine, Stanford University, Stanford, CA 94305-5345
| | - Christine E. Brown
- Department of Hematology & Hematopoietic Cell Transplantation, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010
| |
Collapse
|
35
|
Hervás-Salcedo R, Martín-Antonio B. A Journey through the Inter-Cellular Interactions in the Bone Marrow in Multiple Myeloma: Implications for the Next Generation of Treatments. Cancers (Basel) 2022; 14:3796. [PMID: 35954459 PMCID: PMC9367481 DOI: 10.3390/cancers14153796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/29/2022] [Accepted: 08/02/2022] [Indexed: 02/05/2023] Open
Abstract
Tumors are composed of a plethora of extracellular matrix, tumor and non-tumor cells that form a tumor microenvironment (TME) that nurtures the tumor cells and creates a favorable environment where tumor cells grow and proliferate. In multiple myeloma (MM), the TME is the bone marrow (BM). Non-tumor cells can belong either to the non-hematological compartment that secretes soluble mediators to create a favorable environment for MM cells to grow, or to the immune cell compartment that perform an anti-MM activity in healthy conditions. Indeed, marrow-infiltrating lymphocytes (MILs) are associated with a good prognosis in MM patients and have served as the basis for developing different immunotherapy strategies. However, MM cells and other cells in the BM can polarize their phenotype and activity, creating an immunosuppressive environment where immune cells do not perform their cytotoxic activity properly, promoting tumor progression. Understanding cell-cell interactions in the BM and their impact on MM proliferation and the performance of tumor surveillance will help in designing efficient anti-MM therapies. Here, we take a journey through the BM, describing the interactions of MM cells with cells of the non-hematological and hematological compartment to highlight their impact on MM progression and the development of novel MM treatments.
Collapse
Affiliation(s)
| | - Beatriz Martín-Antonio
- Department of Experimental Hematology, Instituto de Investigación Sanitaria-Fundación Jiménez Diaz (IIS-FJD), University Autonomous of Madrid (UAM), 28040 Madrid, Spain
| |
Collapse
|
36
|
Holstein SA, Asimakopoulos F, Azab AK, Bianchi G, Bhutani M, Crews LA, Cupedo T, Giles H, Gooding S, Hillengass J, John L, Kaiser S, Lee L, Maclachlan K, Pasquini MC, Pichiorri F, Shah N, Shokeen M, Shy BR, Smith EL, Verona R, Usmani SZ, McCarthy PL. Proceedings from the Blood and Marrow Transplant Clinical Trials Network Myeloma Intergroup Workshop on Immune and Cellular Therapy in Multiple Myeloma. Transplant Cell Ther 2022; 28:446-454. [PMID: 35605882 PMCID: PMC9357156 DOI: 10.1016/j.jtct.2022.05.019] [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: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 11/30/2022]
Abstract
The Blood and Marrow Transplant Clinical Trials Network (BMT CTN) Myeloma Intergroup conducted a workshop on Immune and Cellular Therapy in Multiple Myeloma on January 7, 2022. This workshop included presentations by basic, translational, and clinical researchers with expertise in plasma cell dyscrasias. Four main topics were discussed: platforms for myeloma disease evaluation, insights into pathophysiology, therapeutic target and resistance mechanisms, and cellular therapy for multiple myeloma. Here we provide a comprehensive summary of these workshop presentations.
Collapse
Affiliation(s)
| | - Fotis Asimakopoulos
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | | | - Giada Bianchi
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Leslie A Crews
- Moores Cancer Center, University of California San Diego, La Jolla, California
| | - Tom Cupedo
- ErasmusMC Cancer Institute Rotterdam, Rotterdam, The Netherlands
| | - Hannah Giles
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom
| | - Sarah Gooding
- MRC Molecular Hematology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Lukas John
- University Hospital Heidelberg, Heidelberg, Germany
| | | | - Lydia Lee
- University College London, London, United Kingdom
| | | | | | - Flavia Pichiorri
- Judy and Bernard Briskin Center for Multiple Myeloma Research, Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, California; Department of Hematologic Malignancies Translational Science, Beckman Research Institute, City of Hope, Duarte, California
| | - Nina Shah
- University of California San Francisco, San Francisco, California
| | - Monica Shokeen
- Washington University School of Medicine, St. Louis, Missouri
| | - Brian R Shy
- University of California San Francisco, San Francisco, California
| | - Eric L Smith
- Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Raluca Verona
- Janssen Research & Development, Spring House, Pennsylvania
| | - Saad Z Usmani
- Memorial Sloan Kettering Cancer Center, New York, New York
| | | |
Collapse
|
37
|
Maciocia PM, Wawrzyniecka PA, Maciocia NC, Burley A, Karpanasamy T, Devereaux S, Hoekx M, O'Connor D, Leon T, Rapoz-D'Silva T, Pocock R, Rahman S, Gritti G, Yánez DC, Ross S, Crompton T, Williams O, Lee L, Pule MA, Mansour MR. Anti-CCR9 chimeric antigen receptor T cells for T-cell acute lymphoblastic leukemia. Blood 2022; 140:25-37. [PMID: 35507686 DOI: 10.1182/blood.2021013648] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 04/19/2022] [Indexed: 11/20/2022] Open
Abstract
T cell acute lymphoblastic leukemia (T-ALL) is an aggressive malignancy of immature T lymphocytes, associated with higher rates of induction failure compared with those in B cell acute lymphoblastic leukemia. The potent immunotherapeutic approaches applied in B cell acute lymphoblastic leukemia, which have revolutionized the treatment paradigm, have proven more challenging in T-ALL, largely due to a lack of target antigens expressed on malignant but not healthy T cells. Unlike B cell depletion, T-cell aplasia is highly toxic. Here, we show that the chemokine receptor CCR9 is expressed in >70% of cases of T-ALL, including >85% of relapsed/refractory disease, and only on a small fraction (<5%) of normal T cells. Using cell line models and patient-derived xenografts, we found that chimeric antigen receptor (CAR) T-cells targeting CCR9 are resistant to fratricide and have potent antileukemic activity both in vitro and in vivo, even at low target antigen density. We propose that anti-CCR9 CAR-T cells could be a highly effective treatment strategy for T-ALL, avoiding T cell aplasia and the need for genome engineering that complicate other approaches.
Collapse
Affiliation(s)
- Paul M Maciocia
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Patrycja A Wawrzyniecka
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Nicola C Maciocia
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Amy Burley
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Thaneswari Karpanasamy
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Sam Devereaux
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Malika Hoekx
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - David O'Connor
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Theresa Leon
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Tanya Rapoz-D'Silva
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Rachael Pocock
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Sunniyat Rahman
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Giuseppe Gritti
- Department of Haematology, Ospedale Papa Giovanni XXIII, Bergamo, Italy; and
| | - Diana C Yánez
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Susan Ross
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Tessa Crompton
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Owen Williams
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Lydia Lee
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Martin A Pule
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
| | - Marc R Mansour
- Department of Haematology, Cancer Institute, University College London, London, United Kingdom
- Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| |
Collapse
|
38
|
Zhang X, Zhu L, Zhang H, Chen S, Xiao Y. CAR-T Cell Therapy in Hematological Malignancies: Current Opportunities and Challenges. Front Immunol 2022; 13:927153. [PMID: 35757715 PMCID: PMC9226391 DOI: 10.3389/fimmu.2022.927153] [Citation(s) in RCA: 107] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy represents a major breakthrough in cancer treatment, and it has achieved unprecedented success in hematological malignancies, especially in relapsed/refractory (R/R) B cell malignancies. At present, CD19 and BCMA are the most common targets in CAR-T cell therapy, and numerous novel therapeutic targets are being explored. However, the adverse events related to CAR-T cell therapy might be serious or even life-threatening, such as cytokine release syndrome (CRS), CAR-T-cell-related encephalopathy syndrome (CRES), infections, cytopenia, and CRS-related coagulopathy. In addition, due to antigen escape, the limited CAR-T cell persistence, and immunosuppressive tumor microenvironment, a considerable proportion of patients relapse after CAR-T cell therapy. Thus, in this review, we focus on the progress and challenges of CAR-T cell therapy in hematological malignancies, such as attractive therapeutic targets, CAR-T related toxicities, and resistance to CAR-T cell therapy, and provide some practical recommendations.
Collapse
Affiliation(s)
- Xiaomin Zhang
- Department of Hematology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lingling Zhu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Hui Zhang
- School of Medicine, Jishou University, Jishou, China
| | - Shanshan Chen
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yang Xiao
- Institute of Clinical Medicine College, Guangzhou University of Chinese Medicine, Guangzhou, China.,Department of Hematology, Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, Shenzhen, China
| |
Collapse
|
39
|
Kegyes D, Constantinescu C, Vrancken L, Rasche L, Gregoire C, Tigu B, Gulei D, Dima D, Tanase A, Einsele H, Ciurea S, Tomuleasa C, Caers J. Patient selection for CAR T or BiTE therapy in multiple myeloma: Which treatment for each patient? J Hematol Oncol 2022; 15:78. [PMID: 35672793 PMCID: PMC9171942 DOI: 10.1186/s13045-022-01296-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/22/2022] [Indexed: 01/09/2023] Open
Abstract
Multiple myeloma (MM) is a plasma cell malignancy that affects an increasing number of patients worldwide. Despite all the efforts to understand its pathogenesis and develop new treatment modalities, MM remains an incurable disease. Novel immunotherapies, such as CAR T cell therapy (CAR) and bispecific T cell engagers (BiTE), are intensively targeting different surface antigens, such as BMCA, SLAMF7 (CS1), GPRC5D, FCRH5 or CD38. However, stem cell transplantation is still indispensable in transplant-eligible patients. Studies suggest that the early use of immunotherapy may improve outcomes significantly. In this review, we summarize the currently available clinical literature on CAR and BiTE in MM. Furthermore, we will compare these two T cell-based immunotherapies and discuss potential therapeutic approaches to promote development of new clinical trials, using T cell-based immunotherapies, even as bridging therapies to a transplant.
Collapse
Affiliation(s)
- David Kegyes
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Catalin Constantinescu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Louise Vrancken
- Laboratory of Hematology, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Leo Rasche
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Celine Gregoire
- Laboratory of Hematology, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| | - Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Diana Gulei
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania
| | - Alina Tanase
- Department of Stem Cell Transplantation, Fundeni Clinical Institute, Bucharest, Romania
| | - Hermann Einsele
- Department of Internal Medicine II, University of Würzburg, Würzburg, Germany
| | - Stefan Ciurea
- Hematopoietic Stem Cell Transplantation and Cellular Therapy Program, Division of Hematology/Oncology, Chao Family Comprehensive Cancer Center, University of California, Irvine, USA
| | - Ciprian Tomuleasa
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania.
- Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj-Napoca, Romania.
| | - Jo Caers
- Laboratory of Hematology, University of Liège, Liège, Belgium
- Department of Hematology, CHU de Liège, Liège, Belgium
| |
Collapse
|
40
|
Khan AN, Chowdhury A, Karulkar A, Jaiswal AK, Banik A, Asija S, Purwar R. Immunogenicity of CAR-T Cell Therapeutics: Evidence, Mechanism and Mitigation. Front Immunol 2022; 13:886546. [PMID: 35677038 PMCID: PMC9169153 DOI: 10.3389/fimmu.2022.886546] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/22/2022] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy demonstrated remarkable success in long-term remission of cancers and other autoimmune diseases. Currently, six products (Kymriah, Yescarta, Tecartus, Breyanzi, Abecma, and Carvykti) are approved by the US-FDA for treatment of a few hematological malignancies. All the six products are autologous CAR-T cell therapies, where delivery of CAR, which comprises of scFv (single-chain variable fragment) derived from monoclonal antibodies for tumor target antigen recognition is through a lentiviral vector. Although available CAR-T therapies yielded impressive response rates in a large number of patients in comparison to conventional treatment strategies, there are potential challenges in the field which limit their efficacy. One of the major challenges is the induction of humoral and/or cellular immune response in patients elicited due to scFv domain of CAR construct, which is of non-human origin in majority of the commercially available products. Generation of anti-CAR antibodies may lead to the clearance of the therapeutic CAR-T cells, increasing the likelihood of tumor relapse and lower the CAR-T cells efficacy upon reinfusion. These immune responses influence CAR-T cell expansion and persistence, that might affect the overall clinical response. In this review, we will discuss the impact of immunogenicity of the CAR transgene on treatment outcomes. Finally, this review will highlight the mitigation strategies to limit the immunogenic potential of CARs and improve the therapeutic outcome.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Rahul Purwar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
41
|
Ohmine K, Uchibori R. Novel immunotherapies in multiple myeloma. Int J Hematol 2022; 115:799-810. [PMID: 35583724 DOI: 10.1007/s12185-022-03365-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 04/18/2022] [Accepted: 04/19/2022] [Indexed: 11/24/2022]
Abstract
For a substantial period, options for the treatment of multiple myeloma (MM) were limited; however, the advent of novel therapies into clinical practice in the 1990s resulted in dramatic changes in the prognosis of the disease. Subsequently, new proteasome inhibitors and immunomodulators with innovations in efficacy and toxicity were introduced; yet there remains a spectrum of patients with poor outcomes with current treatment strategies. One of the causes of disease progression in MM is the loss of the ability of the dysfunctional immune environment to control virulent cell clones. In recent years, therapies to overcome the immunosuppressive tumor microenvironment and activate the host immune system have shown promise in MM, especially in relapsed and refractory disease. Clinical use of this approach has been approved for several immunotherapies, and a number of studies are currently underway in clinical trials. This review outlines three of the newest and most promising approaches being investigated to enhance the immune system against MM: (1) overcoming immunosuppression with checkpoint inhibitors, (2) boosting immunity against tumors with vaccines, and (3) enhancing immune effectors with adoptive cell therapy. Information on the latest clinical trials in each class will be provided, and further developments will be discussed.
Collapse
Affiliation(s)
- Ken Ohmine
- Division of Hematology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
- Division of Immuno-Gene and Cell Therapy (Takara Bio), Jichi Medical University, Shimotsuke, Tochigi, Japan.
| | - Ryosuke Uchibori
- Division of Immuno-Gene and Cell Therapy (Takara Bio), Jichi Medical University, Shimotsuke, Tochigi, Japan
| |
Collapse
|
42
|
Preclinical validation and phase I trial of 4-hydroxysalicylanilide, targeting ribonucleotide reductase mediated dNTP synthesis in multiple myeloma. J Biomed Sci 2022; 29:32. [PMID: 35546402 PMCID: PMC9097096 DOI: 10.1186/s12929-022-00813-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/29/2022] [Indexed: 11/25/2022] Open
Abstract
Background Aberrant DNA repair pathways contribute to malignant transformation or disease progression and the acquisition of drug resistance in multiple myeloma (MM); therefore, these pathways could be therapeutically exploited. Ribonucleotide reductase (RNR) is the rate-limiting enzyme for the biosynthesis of deoxyribonucleotides (dNTPs), which are essential for DNA replication and DNA damage repair. In this study, we explored the efficacy of the novel RNR inhibitor, 4-hydroxysalicylanilide (HDS), in myeloma cells and xenograft model. In addition, we assessed the clinical activity and safety of HDS in patients with MM. Methods We applied bioinformatic, genetic, and pharmacological approaches to demonstrate that HDS was an RNR inhibitor that directly bound to RNR subunit M2 (RRM2). The activity of HDS alone or in synergy with standard treatments was evaluated in vitro and in vivo. We also initiated a phase I clinical trial of single-agent HDS in MM patients (ClinicalTrials.gov: NCT03670173) to assess safety and efficacy. Results HDS inhibited the activity of RNR by directly targeting RRM2. HDS decreased the RNR-mediated dNTP synthesis and concomitantly inhibited DNA damage repair, resulting in the accumulation of endogenous unrepaired DNA double-strand breaks (DSBs), thus inhibiting MM cell proliferation and inducing apoptosis. Moreover, HDS overcame the protective effects of IL-6, IGF-1 and bone marrow stromal cells (BMSCs) on MM cells. HDS prolonged survival in a MM xenograft model and induced synergistic anti-myeloma activity in combination with melphalan and bortezomib. HDS also showed a favorable safety profile and demonstrated clinical activity against MM. Conclusions Our study provides a rationale for the clinical evaluation of HDS as an anti-myeloma agent, either alone or in combination with standard treatments for MM. Trial registration: ClinicalTrials.gov, NCT03670173, Registered 12 September 2018. Supplementary information The online version contains supplementary material available at 10.1186/s12929-022-00813-2.
Collapse
|
43
|
Hu K, Huang Y, Hu Y, Huang H. Progress on CAR-T cell therapy for hematological malignancies. Zhejiang Da Xue Xue Bao Yi Xue Ban 2022; 51:192-203. [PMID: 36161291 PMCID: PMC9353627 DOI: 10.3724/zdxbyxb-2022-0055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/20/2022] [Indexed: 06/16/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is an effective treatment for hematological malignancies, which have experienced the development of CD19 CAR-T cells for B lymphoblastic leukemia and lymphoma, B cell maturation antigen (BCMA) CAR-T cells for multiple myeloid, and more recently, the development of CD7 CAR-T cells for T cell malignancies. There are more obstacles for myeloid malignancies compared to other hematological malignancies in this field, thus concerning researches are in more diverse ways. In order to obtain more effective clinical CAR-T cells with lower side effects, scientists have developed multi-target CAR-T cells, universal CAR-T cells, as well as CAR-T cells, CAR-NK cells, CAR-iMac cells derived from induced pluripotent stem cells (iPSC) by genetic engineering. Chinese scientists have made significant contribution to the invention and manufacture of origin CAR-T cells and the establishment of an intact clinical research system. This review introduces the latest progress involving CAR-T cell therapy for hematological malignancies including B lymphoblastic malignancies, T lymphoblastic malignancies and myeloid malignancies, and also discuss the future developments including multi-target, universal and iPSC-derived CAR-related cell therapy.
Collapse
Affiliation(s)
- Kejia Hu
- 1. Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- 2. Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- 3. Institute of Hematology, Zhejiang University, Hangzhou 310058, China
- 4. Zhejiang Provincial Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Yue Huang
- 1. Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- 2. Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- 3. Institute of Hematology, Zhejiang University, Hangzhou 310058, China
- 4. Zhejiang Provincial Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - Yongxian Hu
- 1. Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- 2. Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- 3. Institute of Hematology, Zhejiang University, Hangzhou 310058, China
- 4. Zhejiang Provincial Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| | - He Huang
- 1. Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
- 2. Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou 311121, China
- 3. Institute of Hematology, Zhejiang University, Hangzhou 310058, China
- 4. Zhejiang Provincial Laboratory for Stem Cell and Immunity Therapy, Hangzhou 310058, China
| |
Collapse
|
44
|
He C, Zhang M, Liu L, Han Y, Xu Z, Xiong Y, Yan F, Su D, Chen H, Zheng Y, Cheng F. Cellular membrane-based vesicles displaying a reconstructed B cell maturation antigen for multiple myeloma therapy by dual targeting APRIL and BAFF. Acta Biomater 2022; 143:406-417. [PMID: 35218967 DOI: 10.1016/j.actbio.2022.02.028] [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: 10/30/2021] [Revised: 01/30/2022] [Accepted: 02/18/2022] [Indexed: 11/19/2022]
Abstract
Excessive secretion of cytokines (such as APRIL and BAFF) in the bone marrow microenvironment (BMM) plays an essential role in the formation of relapsed or refractory multiple myeloma (MM). Blocking the binding of excessive cytokines to their receptors is becoming a promising approach for MM therapy. Here, we proposed a strategy of engineering cell membrane-based nanovesicles (NVs) to reconstruct B cell maturation antigen (BCMA), a receptor of APRIL and BAFF, to capture excess APRIL/BAFF in BMM as a bait protein. Our results showed that reconstructed BCMA expressed on the membrane of NVs (Re-BCMA-NVs) retained the ability of binding to soluble and surface-bound APRIL/BAFF in BMM. Consequently, Re-BCMA-NVs blocked the activation of the NF-κB pathway, downregulating the expression of anti-apoptosis genes and cell cycle-related genes, and hence inhibiting MM cell survival. Importantly, Re-BCMA-NVs showed a synergistic anti-MM effect when administrated together with bortezomib (BTZ) in vitro and in vivo. Our NVs targeting multiple cytokines in cancer microenvironment provides a solution to enhance sensitivity of MM cells to BTZ-based therapy. STATEMENT OF SIGNIFICANCE: Excessive APRIL and BAFF is reported to promote the survival of MM cell and facilitate the formation of resistance to bortezomib therapy. In this study, we bioengineered cell membrane derived reconstructed BCMA nanovesicles (Re-BCMA-NVs) to capture both soluble and cell-surface APRIL and BAFF. These NVs inhibited the activation of NF-κB pathway and thus inhibit the survival of MM cells in 2D, 3D and subcutaneous mouse tumor models. Importantly, Re-BCMA-NVs showed a synergistic anti-MM effect when administrated together with bortezomib in vitro and in vivo. Taken together, our NVs targeting multiple cytokines in cancer microenvironment provides a solution to enhance sensitivity of MM cells to bortezomib-based therapy.
Collapse
Affiliation(s)
- Chao He
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Manqi Zhang
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Lingling Liu
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China
| | - Yuhang Han
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Zhanxue Xu
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Yue Xiong
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Fuxia Yan
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Dandan Su
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China
| | - Hongbo Chen
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| | - Yongjiang Zheng
- Department of Hematology, Institute of Hematology, The Third Affiliated Hospital of Sun Yat-Sen University, 600 Tianhe Road, Guangzhou 510630, Guangdong, China.
| | - Fang Cheng
- School of pharmaceutical sciences (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, China.
| |
Collapse
|
45
|
Choi T, Kang Y. Chimeric antigen receptor (CAR) T-cell therapy for multiple myeloma. Pharmacol Ther 2022; 232:108007. [PMID: 34582835 PMCID: PMC8930424 DOI: 10.1016/j.pharmthera.2021.108007] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/23/2022]
Abstract
Although treatment outcomes of multiple myeloma patients have improved significantly during the last two decades, myeloma is still an incurable disease. There are newly emerging immunotherapies to treat multiple myeloma including monoclonal antibodies, antibody-drug conjugate, bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy. Impressive response rate and clinical efficacy in heavily pretreated myeloma patients led to the FDA approval of the first myeloma CAR-T therapy in March 2021. Among many different targets for myeloma CAR-T therapies, B Cell Maturation Antigen (BCMA) has been the most successful target so far, but other targets which can be used either for single-target or dual-target CAR-T's are actively being explored. Clinical efficacy and safety of current myeloma CAR-T therapies will be presented here. Potential mechanisms leading to resistance include clearance of CAR-T cells, antigenic escape, and immunosuppressive tumor microenvironment. Novel strategies to enhance myeloma CAR-T will also be described. In this article, we provide a comprehensive review of the current data and the future directions of myeloma CAR-T therapies.
Collapse
Affiliation(s)
- Taewoong Choi
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, Durham, NC, USA.
| |
Collapse
|
46
|
Omer B, Cardenas MG, Pfeiffer T, Daum R, Huynh M, Sharma S, Nouraee N, Xie C, Tat C, Perconti S, Van Pelt S, Scherer L, DeRenzo C, Shum T, Gottschalk S, Arber C, Rooney CM. A Costimulatory CAR Improves TCR-based Cancer Immunotherapy. Cancer Immunol Res 2022; 10:512-524. [PMID: 35176142 DOI: 10.1158/2326-6066.cir-21-0307] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 11/11/2021] [Accepted: 02/11/2022] [Indexed: 11/16/2022]
Abstract
T-cell receptors (TCR) recognize intracellular and extracellular cancer antigens, allowing T cells to target many tumor antigens. To sustain proliferation and persistence, T cells require not only signaling through the TCR (signal 1), but also costimulatory (signal 2) and cytokine (signal 3) signaling. Because most cancer cells lack costimulatory molecules, TCR engagement at the tumor site results in incomplete T-cell activation and transient antitumor effects. To overcome this lack of signal 2, we genetically modified tumor-specific T cells with a costimulatory chimeric antigen receptor (CoCAR). Like classical CARs, CoCARs combine the antigen-binding domain of an antibody with costimulatory endodomains to trigger T-cell proliferation, but CoCARs lack the cytotoxic CD3ζ chain to avoid toxicity to normal tissues. We first tested a CD19-targeting CoCAR in combination with an HLA-A*02:01-restricted, survivin-specific transgenic TCR (sTCR) in serial cocultures with leukemia cells coexpressing the cognate peptide-HLA complex (signal 1) and CD19 (signal 2). The CoCAR enabled sTCR+ T cells to kill tumors over a median of four additional tumor challenges. CoCAR activity depended on CD19 but was maintained in tumors with heterogeneous CD19 expression. In a murine tumor model, sTCR+CoCAR+ T cells improved tumor control and prolonged survival compared with sTCR+ T cells. We further evaluated the CoCAR in Epstein-Barr virus-specific T cells (EBVST). CoCAR-expressing EBVSTs expanded more rapidly than nontransduced EBVSTs and delayed tumor progression in an EBV+ murine lymphoma model. Overall, we demonstrated that the CoCAR can increase the activity of T cells expressing both native and transgenic TCRs and enhance antitumor responses.
Collapse
Affiliation(s)
- Bilal Omer
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Mara G Cardenas
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Thomas Pfeiffer
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Rachel Daum
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Mai Huynh
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Sandhya Sharma
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Nazila Nouraee
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Cicilyn Xie
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Candise Tat
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Silvana Perconti
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Stacey Van Pelt
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| | - Lauren Scherer
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Chris DeRenzo
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas.,Department of Bone Marrow Transplant and Cellular Therapy, St. Jude's Children's Research Hospital, Memphis, Tennessee
| | - Thomas Shum
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Stephen Gottschalk
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Texas Children's Cancer and Hematology Centers, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas.,Department of Bone Marrow Transplant and Cellular Therapy, St. Jude's Children's Research Hospital, Memphis, Tennessee
| | - Caroline Arber
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas.,Department of Oncology UNIL-CHUV, Lausanne University Hospital, University of Lausanne and Ludwig Institute for Cancer Research Lausanne, Epalinges, Switzerland
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
47
|
Roex G, Campillo-Davo D, Flumens D, Shaw PAG, Krekelbergh L, De Reu H, Berneman ZN, Lion E, Anguille S. Two for one: targeting BCMA and CD19 in B-cell malignancies with off-the-shelf dual-CAR NK-92 cells. J Transl Med 2022; 20:124. [PMID: 35287669 PMCID: PMC8919645 DOI: 10.1186/s12967-022-03326-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 03/01/2022] [Indexed: 01/04/2023] Open
Abstract
Abstract
Background
Chimeric antigen receptor (CAR) T-cell therapy has proven to be a valuable new treatment option for patients with B-cell malignancies. However, by applying selective pressure, outgrowth of antigen-negative tumor cells can occur, eventually resulting in relapse. Subsequent rescue by administration of CAR-T cells with different antigen-specificity indicates that those tumor cells are still sensitive to CAR-T treatment and points towards a multi-target strategy. Due to their natural tumor sensitivity and highly cytotoxic nature, natural killer (NK) cells are a compelling alternative to T cells, especially considering the availability of an off-the-shelf unlimited supply in the form of the clinically validated NK-92 cell line.
Methods
Given our goal to develop a flexible system whereby the CAR expression repertoire of the effector cells can be rapidly adapted to the changing antigen expression profile of the target cells, electrotransfection with CD19-/BCMA-CAR mRNA was chosen as CAR loading method in this study. We evaluated the functionality of mRNA-engineered dual-CAR NK-92 against tumor B-cell lines and primary patient samples. In order to test the clinical applicability of the proposed cell therapy product, the effect of irradiation on the proliferative rate and functionality of dual-CAR NK-92 cells was investigated.
Results
Co-electroporation of CD19 and BMCA CAR mRNA was highly efficient, resulting in 88.1% dual-CAR NK-92 cells. In terms of CD107a degranulation, and secretion of interferon (IFN)-γ and granzyme B, dual-CAR NK-92 significantly outperformed single-CAR NK-92. More importantly, the killing capacity of dual-CAR NK-92 exceeded 60% of single and dual antigen-expressing cell lines, as well as primary tumor cells, in a 4h co-culture assay at low effector to target ratios, matching that of single-CAR counterparts. Furthermore, our results confirm that dual-CAR NK-92 irradiated with 10 Gy cease to proliferate and are gradually cleared while maintaining their killing capacity.
Conclusions
Here, using the clinically validated NK-92 cell line as a therapeutic cell source, we established a readily accessible and flexible platform for the generation of highly functional dual-targeted CAR-NK cells.
Collapse
|
48
|
Nobari ST, Nojadeh JN, Talebi M. B-cell maturation antigen targeting strategies in multiple myeloma treatment, advantages and disadvantages. J Transl Med 2022; 20:82. [PMID: 35144648 PMCID: PMC8832753 DOI: 10.1186/s12967-022-03285-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 01/29/2022] [Indexed: 01/02/2023] Open
Abstract
B cell maturation antigen (BCMA), a transmembrane glycoprotein member of the tumor necrosis factor receptor superfamily 17 (TNFRSF17), highly expressed on the plasma cells of Multiple myeloma (MM) patients, as well as the normal population. BCMA is used as a biomarker for MM. Two members of the TNF superfamily proteins, including B-cell activating factor (BAFF) and A proliferation-inducing ligand (APRIL), are closely related to BCMA and play an important role in plasma cell survival and progression of MM. Despite the maximum specificity of the monoclonal antibody technologies, introducing the tumor-specific antigen(s) is not applicable for all malignancies, such as MM that there plenty of relatively specific antigens such as GPCR5D, MUC1, SLAMF7 and etc., but higher expression of BCMA on these cells in comparison with normal ones can be regarded as a relatively exclusive marker. Currently, different monoclonal antibody (mAb) technologies applied in anti-MM therapies such as daratuzumab, SAR650984, GSK2857916, and CAR-T cell therapies are some of these tools that are reviewed in the present manuscript. By the way, the structure, function, and signaling of the BCMA and related molecule(s) role in normal plasma cells and MM development, evaluated as well as the potential side effects of its targeting by different CAR-T cells generations. In conclusion, BCMA can be regarded as an ideal molecule to be targeted in immunotherapeutic methods, regarding lower potential systemic and local side effects.
Collapse
Affiliation(s)
- Shirin Teymouri Nobari
- Department of Medical Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Nouri Nojadeh
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Talebi
- Department of Applied Cells Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
49
|
Kyte JA. Strategies for Improving the Efficacy of CAR T Cells in Solid Cancers. Cancers (Basel) 2022; 14:cancers14030571. [PMID: 35158839 PMCID: PMC8833730 DOI: 10.3390/cancers14030571] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/17/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Cell therapy with genetically retargeted T cells shows strong clinical efficacy against leukaemia and lymphoma. To make this therapy efficient against solid cancers, a series of hurdles must be addressed. This includes the need to enable the T cells to survive long term in patients and to overcome immunosuppressive mechanisms in the tumour. Further, it is essential to prevent tumour cells from escaping by losing the protein that is recognised by the infused cells. The present article provides an overview of the key strategies that are currently being investigated to overcome these hurdles. A series of approaches have been described in preclinical models, but these remain untested in patients. The further progress of the field will depend on evaluating more strategies in a proper clinical setting. Abstract Therapy with T cells equipped with chimeric antigen receptors (CARs) shows strong efficacy against leukaemia and lymphoma, but not yet against solid cancers. This has been attributed to insufficient T cell persistence, tumour heterogeneity and an immunosuppressive tumour microenvironment. The present article provides an overview of key strategies that are currently investigated to overcome these hurdles. Basic aspects of CAR design are revisited, relevant for tuning the stimulatory signal to the requirements of solid tumours. Novel approaches for enhancing T cell persistence are highlighted, based on epigenetic or post-translational modifications. Further, the article describes CAR T strategies that are being developed for overcoming tumour heterogeneity and the escape of cancer stem cells, as well as for countering prevalent mechanisms of immune suppression in solid cancers. In general, personalised medicine is faced with a lack of drugs matching the patient’s profile. The advances and flexibility of modern gene engineering may allow for the filling of some of these gaps with tailored CAR T approaches addressing mechanisms identified as important in the individual patient. At this point, however, CAR T cell therapy remains unproved in solid cancers. The further progress of the field will depend on bringing novel strategies into clinical evaluation, while maintaining safety.
Collapse
Affiliation(s)
- Jon Amund Kyte
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Mail Box 4950 Nydalen, 0424 Oslo, Norway;
- Department of Clinical Cancer Research, Oslo University Hospital, Mail Box 4950 Nydalen, 0424 Oslo, Norway
| |
Collapse
|
50
|
Zeng W, Zhang P. Resistance and recurrence of malignancies after CAR-T cell therapy. Exp Cell Res 2022; 410:112971. [PMID: 34906583 DOI: 10.1016/j.yexcr.2021.112971] [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: 09/24/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 11/04/2022]
Abstract
The emergence of chimeric antigen receptor T (CAR-T) cell therapy has ushered a new era in cancer therapy, especially the treatment of hematological malignancies. However, resistance and recurrence still occur in some patients after CAR-T cell treatment. CAR-T cell inefficiency and tumor escape have emerged as the main challenges for the long-term disease control of B cell malignancies by this promising immunotherapy. In solid tumor treatment, CAR-T cells must also overcome many hurdles from the tumor or immune-suppressed tumor environment, which have become obstacles to the advancement of CAR-T therapy. Therefore, an understanding of the mechanisms underlying post-CAR treatment failure in patients is necessary. In this review, we characterize some mechanisms of resistance and recurrence after CAR-T cell therapy and correspondingly suggest reasonable treatment strategies.
Collapse
Affiliation(s)
- Wanying Zeng
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China
| | - Pumin Zhang
- National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Bio-targeting Theranostics, Guangxi Medical University, Nanning, Guangxi, 530021, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang Province, 310003, China; Institute of Translational Medicine, Zhejiang University Medical School, Hangzhou, Zhejiang Province, 310058, China.
| |
Collapse
|