1
|
Liu L, Zhu W, Liu N, Gong S, Ma Q, Zhou H, Zhong N, Dai W, Zhao L, Sun R, Wang J, Shi Y, Guo Z. The first case of multiple myeloma treated with ASCT followed by Anti-BCMA CAR-T cells using retrovirus vector: A case report. Heliyon 2024; 10:e36955. [PMID: 39281539 PMCID: PMC11399616 DOI: 10.1016/j.heliyon.2024.e36955] [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: 04/19/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/18/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T)-cell therapy targeting B-cell maturation antigen (BCMA) is currently one of the promising treatment methods for relapsed/refractory multiple myeloma (MM). Herein, this study is a case report on a 41-year-old male patient with MM. Unfortunately, he still developed multidrug-resistant, refractory, and bone marrow suppression after receiving multiline high-intensity chemotherapy. After a detailed evaluation, the physician recommended autologous hematopoietic stem cell transplantation (ASCT) support, followed by sequential immunotherapy with autologous anti- BCMA CAR-T cells. The CAR-T product is a novel anti-BCMA CAR-T based on Retrovirus vectors (RV). It was worth noting that the patient achieved VGPR (very good partial remission) one month after infusion of anti-BCMA CAR-T cells. Recent tests have found that the M protein was no longer detectable and the patient has achieved CR (complete response). Although grade 3 cytokine release syndrome (CRS) appeared, the symptom was well controlled and immune effector cell-associated neurotoxicity syndrome (ICANS) did not occur. This was the first case report of RV prepared anti-BCMA CAR-T cells combined with ASCT for the treatment of MM patient in clinical practice, indicating that the RV-based anti-BCMA-CAR-T cells with ASCT have excellent therapeutic efficacy and high safety in triple-refractory MM patients.
Collapse
Affiliation(s)
- Liqiong Liu
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Wenxiang Zhu
- Shenzhen Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Shenzhen, 518033, China
| | - Ning Liu
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Shiting Gong
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, 518118, China
| | - Qihong Ma
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, 518118, China
| | - Huanhuan Zhou
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Nan Zhong
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Wei Dai
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| | - Lijun Zhao
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, 518118, China
| | - Rui Sun
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, 518118, China
| | - Jianxun Wang
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, 518118, China
| | - Yuanyuan Shi
- Shenzhen Cell Valley Biomedical Co., LTD, Shenzhen, 518118, China
| | - Zhi Guo
- Department of Hematology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, 518052, China
| |
Collapse
|
2
|
Zhang Q, Zhu X, Xiao Y. The critical role of endothelial cell in the toxicity associated with chimeric antigen receptor T cell therapy and intervention strategies. Ann Hematol 2024; 103:2197-2206. [PMID: 38329486 PMCID: PMC11224091 DOI: 10.1007/s00277-024-05640-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 01/21/2024] [Indexed: 02/09/2024]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy has shown promising results in patients with hematological malignancies. However, many patients still have poor prognoses or even fatal outcomes due to the life-threatening toxicities associated with the therapy. Moreover, even after improving the known influencing factors (such as number or type of CAR-T infusion) related to CAR-T cell infusion, the results remain unsatisfactory. In recent years, it has been found that endothelial cells (ECs), which are key components of the organization, play a crucial role in various aspects of immune system activation and inflammatory response. The levels of typical markers of endothelial activation positively correlated with the severity of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxic syndrome (ICANS), suggesting that ECs are important targets for intervention and toxicity prevention. This review focuses on the critical role of ECs in CRS and ICANS and the intervention strategies adopted.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiaojian Zhu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
3
|
Kim H, Oh J, Kim MK, Lee KH, Jeong D. Selenoprotein W engages in overactive osteoclast differentiation in multiple myeloma. Mol Biol Rep 2024; 51:587. [PMID: 38683225 PMCID: PMC11058866 DOI: 10.1007/s11033-024-09517-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Patients with multiple myeloma exhibit malignant osteolytic bone disease due to excessive osteoclast formation and function. We recently identified that osteoclastogenic stimulator selenoprotein W (SELENOW) is upregulated via ERK signaling and downregulated via p38 signaling during receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation. In the intrinsic physiological process, RANKL-induced downregulation of SELENOW maintains proper osteoclast differentiation; in contrast, forced overexpression of SELENOW leads to overactive osteoclast formation and function. METHODS AND RESULTS We observed that SELENOW is highly expressed in multiple myeloma-derived peripheral blood mononuclear cells (PBMCs) and mature osteoclasts when compared to healthy controls. Also, the level of tumor necrosis factor alpha (TNFα), a pathological osteoclastogenic factor, is increased in the PBMCs and serum of patients with multiple myeloma. ERK activation by TNFα was more marked and sustained than that by RANKL, allowing SELENOW upregulation. Excessive expression of SELENOW in osteoclast progenitors and mature osteoclasts derived from multiple myeloma facilitated efficient nuclear translocation of osteoclastogenic transcription factors NF-κB and NFATc1, which are favorable for osteoclast formation. CONCLUSION Our findings suggest a possibility that feedforward signaling of osteoclastogenic SELENOW by TNFα derived from multiple myeloma induces overactive osteoclast differentiation, leading to bone loss during multiple myeloma.
Collapse
Affiliation(s)
- Hyunsoo Kim
- Laboratory of Bone Metabolism and Control, Department of Microbiology, Yeungnam University College of Medicine, Daegu, 42415, Korea
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA19104, USA
| | - Jiin Oh
- Laboratory of Bone Metabolism and Control, Department of Microbiology, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Min Kyoung Kim
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Kyung Hee Lee
- Department of Hematology-Oncology, Yeungnam University College of Medicine, Daegu, 42415, Korea
| | - Daewon Jeong
- Laboratory of Bone Metabolism and Control, Department of Microbiology, Yeungnam University College of Medicine, Daegu, 42415, Korea.
- Company of The Bone Science, Yeungnam University College of Medicine, Daegu, 42415, Korea.
| |
Collapse
|
4
|
Zhou J, Shi F, Luo X, Lei B, Shi Z, Huang C, Zhang Y, Li X, Wang H, Li XY, He X. The persistence and antitumor efficacy of CAR-T cells are modulated by tonic signaling within the CDR. Int Immunopharmacol 2024; 126:111239. [PMID: 37979453 DOI: 10.1016/j.intimp.2023.111239] [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: 08/16/2023] [Revised: 11/07/2023] [Accepted: 11/13/2023] [Indexed: 11/20/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has demonstrated remarkable clinical efficacy, but challenges related to relapse and CAR-T cell exhaustion persist. One contributing factor to this exhaustion is CAR tonic signaling, where CAR-T cells self-activate without antigen stimulation, leading to reduced persistence and impaired antitumor activity. To address this issue, we conducted a preclinical study evaluating tonic signaling using nanobody-derived CAR-T cells. Our investigation revealed that specific characteristics of the complementary determining regions (CDRs), including low solubility, polarity, positive charge, energy, and area of ionic and positive CDR patches of amino acids, were associated with low antigen-independent tonic signaling. Significantly, we observed that stronger tonic signaling directly impacted CAR-T cell proliferation in vitro, consequently leading to CAR-T cell exhaustion and diminished persistence and effectiveness in vivo. Our findings provide compelling preclinical evidence and lay the foundation for the clinical assessment of CAR-T cells with distinct tonic signaling patterns. Understanding the role of CDRs in modulating tonic signaling holds promise for advancing the development of more efficient and durable CAR-T cell therapies, thereby enhancing the treatment of cancer and addressing the challenges of relapse in CAR-T cell therapy.
Collapse
Affiliation(s)
- Jincai Zhou
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China.
| | - Feifei Shi
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Xinran Luo
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Bixia Lei
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Zhongjun Shi
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Chenyu Huang
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Yuting Zhang
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Xiaopei Li
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Huajing Wang
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China
| | - Xian-Yang Li
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China.
| | - Xiaowen He
- R&D Department, OriCell Therapeutics Co. Ltd., 1227 Zhangheng Road, Shanghai, 201203, China.
| |
Collapse
|
5
|
Gao X, Zeng H, Zhao X, Wu H, Yan M, Li Y, Zhang G, Sun F. Efficacy and safety of venetoclax in patients with relapsed/refractory multiple myeloma: a meta-analysis. BMC Cancer 2023; 23:1058. [PMID: 37924016 PMCID: PMC10623759 DOI: 10.1186/s12885-023-11553-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 10/20/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND Venetoclax is clinically active in treating relapsed/refractory multiple myeloma (RRMM). This study evaluated the efficacy and safety of venetoclax or venetoclax with other agents in treating RRMM. METHODS PubMed, Web of Science, Embase, and Cochrane Library were comprehensively searched. We included studies investigating the efficacy and safety of venetoclax or venetoclax with other agents in treating RRMM. Overall response rates (ORR), stringent complete response rates (sCR), complete response rates (CR), very good partial response rates (VGPR), partial response rates (PR), stable disease (SD), progressive disease (PD) and adverse events were synthesized using either a random-effects model or a fixed-effects model. RESULTS A total of 7 clinical trials with 482 patients with RRMM were included. Concerning venetoclax with other agents, the pooled ORR, sCR, CR, VGPR, PR, SD, and PD were 0.76 (95% CIs: 0.62, 0.87), 0.11 (95% CIs: 0.04, 0.21), 0.18 (95% CIs: 0.11, 0.26), 0.16 (95% CIs: 0.12, 0.25), 0.29 (95% CIs: 0.25, 0.34), 0.07 (95% CIs: 0.05, 0.10), and 0.11 (95% CIs: 0.04, 0.23). The overall rate of adverse events ≥ Grade 3 was 0.84 (95% CIs: 0.77, 0.91). The most common non-hematologic adverse events were nausea, diarrhea, fatigue, back pain, and vomiting; hematologic adverse events included thrombocytopenia, neutropenia, anemia, leukopenia, and lymphopenia. CONCLUSIONS This study indicates that venetoclax alone or in combination with other agents reveals favorable treatment responses and acceptable adverse events in treating RRMM.
Collapse
Affiliation(s)
- Xiaohui Gao
- Departments of Pediatrics, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Hui Zeng
- Departments of Hematology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Xiaoyan Zhao
- Departments of Hematology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Haibing Wu
- Departments of Hematology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Minchao Yan
- Departments of Hematology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Yuan Li
- Departments of Hematology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China
| | - Gang Zhang
- Departments of Hematology, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China.
| | - Fei Sun
- Departments of Pediatrics, The Affiliated Hospital of Jiaxing University, Jiaxing, 314000, Zhejiang, China.
| |
Collapse
|
6
|
Gambles MT, Yang J, Kopeček J. Multi-targeted immunotherapeutics to treat B cell malignancies. J Control Release 2023; 358:232-258. [PMID: 37121515 PMCID: PMC10330463 DOI: 10.1016/j.jconrel.2023.04.048] [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: 01/05/2023] [Revised: 04/20/2023] [Accepted: 04/27/2023] [Indexed: 05/02/2023]
Abstract
The concept of multi-targeted immunotherapeutic systems has propelled the field of cancer immunotherapy into an exciting new era. Multi-effector molecules can be designed to engage with, and alter, the patient's immune system in a plethora of ways. The outcomes can vary from effector cell recruitment and activation upon recognition of a cancer cell, to a multipronged immune checkpoint blockade strategy disallowing evasion of the cancer cells by immune cells, or to direct cancer cell death upon engaging multiple cell surface receptors simultaneously. Here, we review the field of multi-specific immunotherapeutics implemented to treat B cell malignancies. The mechanistically diverse strategies are outlined and discussed; common B cell receptor antigen targeting strategies are outlined and summarized; and the challenges of the field are presented along with optimistic insights for the future.
Collapse
Affiliation(s)
- M Tommy Gambles
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA
| | - Jiyuan Yang
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA.
| | - Jindřich Kopeček
- Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA; Department of Molecular Pharmaceutics, University of Utah, Salt Lake City, UT 84112, USA; Department of Biomedical Engineering, University of Utah, Salt Lake City, UT 84112, USA.
| |
Collapse
|
7
|
Li W, Zhang B, Cao W, Zhang W, Li T, Liu L, Xu L, Gao F, Wang Y, Wang F, Xing H, Jiang Z, Shi J, Bian Z, Song Y. Identification of potential resistance mechanisms and therapeutic targets for the relapse of BCMA CAR-T therapy in relapsed/refractory multiple myeloma through single-cell sequencing. Exp Hematol Oncol 2023; 12:44. [PMID: 37158921 PMCID: PMC10165782 DOI: 10.1186/s40164-023-00402-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 04/13/2023] [Indexed: 05/10/2023] Open
Abstract
BACKGROUND BCMA CAR-T is highly effective for relapsed/refractory multiple myeloma(R/R-MM) and significantly improves the survival of patients. However, the short remission time and high relapse rate of MM patients treated with BCMA CAR-T remain bottlenecks that limit long-term survival. The immune microenvironment of the bone marrow (BM) in R/R-MM may be responsible for this. The present study aims to present an in-depth analysis of resistant mechanisms and to explore potential novel therapeutic targets for relapse of BCMA CAR-T treatment via single-cell RNA sequencing (scRNA-seq) of BM plasma cells and immune cells. METHODS This study used 10X Genomic scRNA-seq to identify cell populations in R/R-MM CD45+ BM cells before BCMA CAR-T treatment and relapse after BCMA CAR-T treatment. Cell Ranger pipeline and CellChat were used to perform detailed analysis. RESULTS We compared the heterogeneity of CD45+ BM cells before BCMA CAR-T treatment and relapse after BCMA CAR-T treatment. We found that the proportion of monocytes/macrophages increased, while the percentage of T cells decreased at relapse after BCMA CAR-T treatment. We then reclustered and analyzed the alterations in plasma cells, T cells, NK cells, DCs, neutrophils, and monocytes/macrophages in the BM microenvironment before BCMA CAR-T treatment and relapse after BCMA CAR-T treatment. We show here that the percentage of BCMA positive plasma cells increased at relapse after BCMA CAR-T cell therapy. Other targets such as CD38, CD24, SLAMF7, CD138, and GPRC5D were also found to be expressed in plasma cells of the R/R-MM patient at relapse after BCMA CAR-T cell therapy. Furthermore, exhausted T cells, TIGIT+NK cells, interferon-responsive DCs, and interferon-responsive neutrophils, increased in the R/R-MM patient at relapse after BCMA CAR-T cell treatment. Significantly, the proportion of IL1βhi Mφ, S100A9hi Mφ, interferon-responsive Mφ, CD16hi Mφ, MARCO hi Mφ, and S100A11hi Mφ significantly increased in the R/R-MM patient at relapse after BCMA CAR-T cell therapy. Cell-cell communication analysis indicated that monocytes/macrophages, especially the MIF and APRIL signaling pathway are key players in R/R-MM patient at relapse after BCMA CAR-T cell therapy. CONCLUSION Taken together, our data extend the understanding of intrinsic and extrinsic relapse of BCMA CAR-T treatment in R/R-MM patient and the potential mechanisms involved in the alterations of antigens and the induced immunosuppressive microenvironment, which may provide a basis for the optimization of BCMA CAR-T strategies. Further studies should be performed to confirm these findings.
Collapse
Affiliation(s)
- Wei Li
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Binglei Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Weijie Cao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Wenli Zhang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - Tiandong Li
- College of Public Health, Zhengzhou University, Zhengzhou, 450000, Henan, China
| | - Lina Liu
- Department of Hematology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, 450008, Henan, China
| | - LinPing Xu
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, 450008, China
| | - Fengcai Gao
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Yanmei Wang
- Department of Hematology, Zhengzhou People's Hospital, Zhengzhou, 450003, Henan, China
| | - Fang Wang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Haizhou Xing
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China
| | - Jianxiang Shi
- BGI College & Henan Institute of Medical and Pharmaceutical Sciences in Academy of Medical Science, Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Zhilei Bian
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China.
| | - Yongping Song
- Department of Hematology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Department of Hematology, Henan Provincial Hematology Hospital, Zhengzhou, 450000, Henan, China.
| |
Collapse
|
8
|
Luteolin inhibits the TGF-β signaling pathway to overcome bortezomib resistance in multiple myeloma. Cancer Lett 2023; 554:216019. [PMID: 36442773 DOI: 10.1016/j.canlet.2022.216019] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
Multiple myeloma (MM) is an incurable condition and the second most common hematological malignancy. Over the past few years, there has been progress in the treatment of MM, but most patients still relapse. Multiple myeloma stem-like cells (MMSCs) are believed to be the main reason for drug resistance and eventual relapse. Currently, there are not enough therapeutic agents that have been identified for eradication of MMSCs, and thus, identification of the same may alleviate the issue of relapse in patients. In the present study, we showed that luteolin (LUT), a natural compound obtained from different plants, such as vegetables, medicinal herbs, and fruits, effectively inhibits the proliferation of MM cells and overcomes bortezomib (BTZ) resistance in them in vitro and in vivo, mainly by decreasing the proportion of ALDH1+ cells. Furthermore, RNA sequencing after LUT treatment of MM cell lines and an MM xenograft mouse model revealed that the effects of the compound are mediated through inhibition of transforming growth factor-β signaling. Similarly, we found that LUT also significantly reduced the proportion of ALDH1+ cells in primary CD138+ plasma cells. In addition, LUT could overcome the BTZ treatment-induced increase in the proportion of ALDH1+ cells, and the combination of LUT and BTZ had a synergistic effect against myeloma cells. Collectively, our findings suggested that LUT is a promising agent that manifests MMSCs to overcome BTZ resistance, alone or in combination with BTZ, and thus, is a potential therapeutic drug for the treatment of MM.
Collapse
|
9
|
Chen W, Liu A, Li L. The MODIFY Study Protocol: An Open-Label, Single-Arm, Multicenter, Prospective Pragmatic Study of Ixazomib-Based Triple-Drug Therapy in Chinese Patients with Multiple Myeloma. Adv Ther 2023; 40:705-717. [PMID: 36463561 PMCID: PMC9898406 DOI: 10.1007/s12325-022-02355-3] [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] [Accepted: 10/11/2022] [Indexed: 12/05/2022]
Abstract
BACKGROUND Although the bortezomib-based triple-drug therapy is considered as a front-line therapy for multiple myeloma (MM) in Chinese patients, increased level of toxicity leads to treatment dissatisfaction. Treatment with ixazomib, an oral proteasome inhibitor, has demonstrated better efficacy and safety profile without increasing the toxicity. In this study, we investigate the safety and clinical outcomes of Chinese patients with newly diagnosed MM (NDMM) who transitioned from a bortezomib-based triple-drug therapy to an ixazomib-based triple-drug therapy in a real-world clinical setting. METHODS This will be an open-label, single-arm, multicenter, prospective, observational study will recruit Chinese patients (aged ≥ 18 years) diagnosed with NDMM using International Myeloma Working Group (IMWG) criteria and who have received a bortezomib-based triple-drug therapy for more than two cycles as initial therapy. The previous bortezomib-based triple-drug therapy may include bortezomib, cyclophosphamide, and dexamethasone or lenalidomide, bortezomib, and dexamethasone or bortezomib, doxorubicin, and dexamethasone or bortezomib, thalidomide, and dexamethasone. At the time of enrollment, patients must have achieved at least partial response as defined by IMWG criteria. Approximately 320 eligible patients at 15 top MM hospitals in China will be treated with ixazomib triple-drug therapy and followed up once every 3 months for 24 months, unless specified. The primary endpoint is to assess progression-free survival at 2 years for Chinese patients with NDMM who have transitioned from a bortezomib-based triple-drug therapy to ixazomib-based triple-drug therapy. The clinical effectiveness, safety and tolerability, patient-reported outcomes, and health economic/resource utilization will be evaluated as secondary endpoints. PLANNED OUTCOMES The results from this study may provide evidence to verify the benefits of transitioning from bortezomib-based triple-drug therapy to ixazomib-based triple-drug therapy in Chinese patients with NDMM in a real-world clinical setting. TRIAL REGISTRATION The study has been registered at clinicalTrials.gov (NCT05013190).
Collapse
Affiliation(s)
- Wenming Chen
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - Aijun Liu
- Department of Hematology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Lin Li
- Medical, Takeda (China) International Trading Co., Ltd, Shanghai, China
| |
Collapse
|
10
|
Wang Z, Chen C, Wang L, Jia Y, Qin Y. Chimeric antigen receptor T-cell therapy for multiple myeloma. Front Immunol 2022; 13:1050522. [PMID: 36618390 PMCID: PMC9814974 DOI: 10.3389/fimmu.2022.1050522] [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: 09/21/2022] [Accepted: 12/08/2022] [Indexed: 12/24/2022] Open
Abstract
Multiple myeloma (MM) is a malignant plasma cell disorder that remains incurable for most patients, as persistent clonal evolution drives new mutations which confer MM high-risk signatures and resistance to standard care. The past two decades have significantly refashioned the therapeutic options for MM, especially adoptive T cell therapy contributing to impressive response rate and clinical efficacy. Despite great promises achieved from chimeric antigen receptor T-cell (CAR-T) therapy, the poor durability and severe toxicity (cytokine release syndrome and neurotoxicity) are still huge challenges. Therefore, relapsed/refractory multiple myeloma (RRMM), characterized by the nature of clinicopathologic and molecular heterogeneity, is frequently associated with poor prognosis. B Cell Maturation Antigen (BCMA) is the most successful target for CAR-T therapy, and other potential targets either for single-target or dual-target CAR-T are actively being studied in numerous clinical trials. Moreover, mechanisms driving resistance or relapse after CAR-T therapy remain uncharacterized, which might refer to T-cell clearance, antigen escape, and immunosuppressive tumor microenvironment. Engineering CAR T-cell to improve both efficacy and safety continues to be a promising area for investigation. In this review, we aim to describe novel tumor-associated neoantigens for MM, summarize the data from current MM CAR-T clinical trials, introduce the mechanism of disease resistance/relapse after CAR-T infusion, highlight innovations capable of enhanced efficacy and reduced toxicity, and provide potential directions to optimize manufacturing processes.
Collapse
Affiliation(s)
| | | | | | - Yongxu Jia
- *Correspondence: Yongxu Jia, ; Yanru Qin,
| | - Yanru Qin
- *Correspondence: Yongxu Jia, ; Yanru Qin,
| |
Collapse
|
11
|
Sriram H, Kunjachan F, Khanka T, Gawai S, Ghogale S, Deshpande N, Girase K, Patil J, Chatterjee G, Rajpal S, Patkar NV, Bagal B, Jain H, Sengar M, Hasan SK, Khattry N, Subramanian PG, Gujral S, Tembhare PR. Expression levels and patterns of B-cell maturation antigen in newly diagnosed and relapsed multiple myeloma patients from Indian subcontinent. CYTOMETRY. PART B, CLINICAL CYTOMETRY 2022; 102:462-470. [PMID: 36346307 DOI: 10.1002/cyto.b.22099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/16/2022] [Accepted: 10/18/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND Many novel therapies are being evaluated for the treatment of Multiple myeloma (MM). The cell-surface protein B-cell maturation antigen (BCMA, CD269) has recently emerged as a promising target for CAR-T cell and monoclonal-antibody therapies in MM. However, the knowledge of the BCMA expression-pattern in myeloma patients from the Indian subcontinent is still not available. We present an in-depth study of BCMA expression-pattern on abnormal plasma cells (aPC) in Indian MM patients. METHODS We studied BM samples from 217 MM patients (211-new and 6-relapsed) with a median age of 56 years (range, 30-78 years & M:F-2.29) and 20 control samples. Expression levels/patterns of CD269 (clone-19f2) were evaluated in aPCs from MM patients and in normal PCs (nPC) from uninvolved staging bone marrow samples (controls) using multicolor flow cytometry (MFC). Expression-level of CD269 was determined as a ratio of mean fluorescent intensity (MFI-R) of CD269 in PCs to that of non-B-lymphocytes and expression-pattern (homogenous/heterogeneous) as coefficient-of-variation of immunofluorescence (CVIF). RESULTS Median (range) percentage of CD269-positive abnormal-PCs in total PCs was 71.6% (0.49-99.29%). The MFI-R (median, range) of CD269 was significantly higher in aPCs (4.13, 1.12-26.88) than nPCs (3.33, 1.23-12.87), p < .0001. Median (range) MFI of CD269 at diagnosis and relapse were 2.39 (0.77-9.57) and 2.66 (2.15-3.23) respectively. CD269 levels were similar at diagnosis and relapse, p = .5529. CONCLUSIONS We demonstrated that BCMA/CD269 is highly expressed in aPCs from a majority of MM patients, both at diagnosis and relapse. Thus, BCMA is a valuable target for therapy for Indian MM patients.
Collapse
Affiliation(s)
- Harshini Sriram
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Florence Kunjachan
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Twinkle Khanka
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sangamitra Gawai
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sitaram Ghogale
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Nilesh Deshpande
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Karishma Girase
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Jagruti Patil
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Gaurav Chatterjee
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sweta Rajpal
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Nikhil V Patkar
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Bhausaheb Bagal
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Hasmukh Jain
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Manju Sengar
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Syed Khizer Hasan
- Cell and Tumor Biology Group, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Navin Khattry
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Papagudi G Subramanian
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Sumeet Gujral
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Prashant R Tembhare
- Hematopathology Laboratory, ACTREC, Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
12
|
Soekojo CY, Chng WJ. Treatment Horizon in Multiple Myeloma. Eur J Haematol Suppl 2022; 109:425-440. [PMID: 35880395 DOI: 10.1111/ejh.13840] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 07/23/2022] [Indexed: 11/29/2022]
Abstract
OBJECTIVES This paper reviews current and emerging therapies for MM. METHODS Narrative review RESULTS: Multiple myeloma (MM) is a complex, heterogenous condition, and in recent years there has been an expansion in the number and range of treatments. Several new treatment approaches, including enhanced monoclonal antibodies, antibody-drug conjugates (ADC), bispecific T-cell engagers (BiTE) and chimeric antigen-T-cell therapy (CAR-T) are under development. CONCLUSIONS The emergence of new treatments that aim to tackle MM-associated immune dysfunction has led to improvements in overall survival.
Collapse
Affiliation(s)
- Cinnie Yentia Soekojo
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System
| | - Wee Joo Chng
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System
| |
Collapse
|
13
|
Quazi S. An Overview of CAR T Cell Mediated B Cell Maturation Antigen Therapy. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:e392-e404. [PMID: 34992008 DOI: 10.1016/j.clml.2021.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
Abstract
Multiple Myeloma (MM) is one of the incurable types of cancer in plasma cells. While immense progress has been made in the treatment of this malignancy, a large percentage of patients were unable to adapt to such therapy. Additionally, these therapies might be associated with significant diseases and are not always tolerated well in all patients. Since cancer in plasma cells has no cure, patients develop resistance to treatments, resulting in R/R MM (Refractory/Relapsed Multiple Myeloma). BCMA (B cell maturation antigen) is primarily produced on mature B cells. It's up-regulation and activation are associated with multiple myeloma in both murine and human models, indicating that this might be an effective therapeutic target for this type of malignancy. Additionally, BCMA's predictive value, association with effective clinical trials, and capacity to be utilized in previously difficult to observe patient populations, imply that it might be used as a biomarker for multiple myeloma. Numerous kinds of BCMA-targeting medicines have demonstrated antimyeloma efficacy in individuals with refractory/relapsed MM, including CAR T-cell (Chimeric antigen receptor T cell) treatments, ADCs (Antibody-drug conjugate s), bispecific antibody constructs. Among these medications, CART cell-mediated BCMA therapy has shown significant outcomes in multiple myeloma clinical trials. This review article outlines CAR T cell mediated BCMA medicines have the efficiency to change the therapeutic pattern for multiple myeloma significantly.
Collapse
Affiliation(s)
- Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore, Karnataka, India.
| |
Collapse
|
14
|
Zhang L, Shen X, Yu W, Li J, Zhang J, Zhang R, Li J, Chen L. Comprehensive meta-analysis of anti-BCMA chimeric antigen receptor T-cell therapy in relapsed or refractory multiple myeloma. Ann Med 2021; 53:1547-1559. [PMID: 34459681 PMCID: PMC8409966 DOI: 10.1080/07853890.2021.1970218] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/16/2021] [Accepted: 08/13/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR) T-cell therapy shows impressive results in clinical trials. We conducted a meta-analysis based on the most recent data to systematically describe the efficacy and safety of anti-BCMA CAR T therapy for patients with relapsed or refractory multiple myeloma (R/R MM). METHODS PubMed, Embase, Web of Science, Cochrane library, ClinicalTrials.gov, China Biology Medicine disc (CBM disc) and Wanfang Data were searched on 8 November 2020. Registration number of PROSPERO was CRD42020219127. RESULTS From 763 articles, we identified 22 appropriate studies with 681 patients. The pooled overall response rate (ORR) was 85.2% (95%CI 0.797-0.910), complete response rate (CRR) was 47.0% (95%CI 0.378-0.583), and minimal residual disease (MRD) negativity rate was 97.8% (95%CI 0.935-1.022). The pooled incidence of grade 3-4 cytokine release syndrome was 6.6% (95%CI 0.036-0.096) and neurotoxicity was 2.2% (95%CI 0.006-0.038). The median progression-free survival (PFS) was 14.0 months and median overall survival (OS) was 24.0 months. Subgroup analysis showed dual epitope-binding CAR T cells achieved the best therapy outcomes and humanized CAR T cells had the best safety profile. Patients who were older, heavily pre-treated or received lower dose of CAR T cells had worse ORR. There was no significant difference in ORR, CRR and PFS between patients with and without high-risk cytogenetic features. The PFS and CRR of non-extramedullary disease (EMD) group was superior to those of EMD group. CONCLUSION Anti-BCMA CAR T therapy is effective and safe for patients with R/R MM. It can improve the prognosis of patients with high-risk cytogenetic features while the prognosis of patients with EMD remains poor. Moreover, patients are likely to benefit from an earlier use of CAR T therapy and human-derived CAR T cells have obvious advantages based on the existing data.
Collapse
Affiliation(s)
- Lina Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Xuxing Shen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Wenjun Yu
- Department of Geriatric Medicine, Geriatric Hospital of Nanjing Medical University, Jiangsu Province Geriatric Institute, Nanjing, China
| | - Jing Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Jue Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Run Zhang
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Jianyong Li
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| | - Lijuan Chen
- Department of Hematology, The First Affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, China
| |
Collapse
|
15
|
Chimeric Antigen Receptor T-Cell Therapeutics for Multiple Myeloma: Moving Into the Spotlight. ACTA ACUST UNITED AC 2021; 27:205-212. [PMID: 34549909 DOI: 10.1097/ppo.0000000000000525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
ABSTRACT Chimeric antigen receptor (CAR) T-cell therapy has quickly emerged as a highly promising treatment for patients with relapsed and refractory multiple myeloma. There are numerous candidates under development, each with their unique characteristics and points of differentiation. The most recent US Food and Drug Administration approval of the first B-cell maturation antigen-targeted CAR-T cell therapy on March 26, 2021, has paved a path forward for the eventual evaluation of more of these investigational agents undergoing clinical trials. Herein, we highlight, from a clinical development perspective, the CAR-T cell therapies farthest along in development with updated data from the American Society of Hematology 2020 annual meeting. We also discuss potential paths of overcoming resistance to these therapies and the future direction for CAR-T cell therapeutics in multiple myeloma.
Collapse
|
16
|
Chimeric antigen receptor- and natural killer cell receptor-engineered innate killer cells in cancer immunotherapy. Cell Mol Immunol 2021; 18:2083-2100. [PMID: 34267335 PMCID: PMC8429625 DOI: 10.1038/s41423-021-00732-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 06/18/2021] [Indexed: 02/06/2023] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T-cell (CAR-T) therapy has demonstrated impressive therapeutic efficacy against hematological malignancies, but multiple challenges have hindered its application, particularly for the eradication of solid tumors. Innate killer cells (IKCs), particularly NK cells, NKT cells, and γδ T cells, employ specific antigen-independent innate tumor recognition and cytotoxic mechanisms that simultaneously display high antitumor efficacy and prevent tumor escape caused by antigen loss or modulation. IKCs are associated with a low risk of developing GVHD, thus offering new opportunities for allogeneic "off-the-shelf" cellular therapeutic products. The unique innate features, wide tumor recognition range, and potent antitumor functions of IKCs make them potentially excellent candidates for cancer immunotherapy, particularly serving as platforms for CAR development. In this review, we first provide a brief summary of the challenges hampering CAR-T-cell therapy applications and then discuss the latest CAR-NK-cell research, covering the advantages, applications, and clinical translation of CAR- and NK-cell receptor (NKR)-engineered IKCs. Advances in synthetic biology and the development of novel genetic engineering techniques, such as gene-editing and cellular reprogramming, will enable the further optimization of IKC-based anticancer therapies.
Collapse
|
17
|
Liu H, Pan C, Song W, Liu D, Li Z, Zheng L. Novel strategies for immuno-oncology breakthroughs with cell therapy. Biomark Res 2021; 9:62. [PMID: 34332618 PMCID: PMC8325826 DOI: 10.1186/s40364-021-00316-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/16/2021] [Indexed: 12/19/2022] Open
Abstract
Cell therapy has evolved rapidly in the past several years with more than 250 clinical trials ongoing around the world. While more indications of cellular therapy with chimeric antigen receptor - engineered T cells (CAR-T) are approved for hematologic malignancies, new concepts and strategies of cellular therapy for solid tumors are emerging and are discussed. These developments include better selections of targets by shifting from tumor-associated antigens to personalized tumor-specific neoantigens, an enhancement of T cell trafficking by breaking the stromal barriers, and a rejuvenation of exhausted T cells by targeting immunosuppressive mechanisms in the tumor microenvironment (TME). Despite significant remaining challenges, we believe that cell therapy will once again lead and revolutionize cancer immunotherapy before long because of the maturation of technologies in T cell engineering, target selection and T cell delivery. This review highlighted the recent progresses reported at the 2020 China Immuno-Oncology Workshop co-organized by the Chinese American Hematologist and Oncologist Network (CAHON), the China National Medical Product Administration (NMPA), and Tsinghua University.
Collapse
Affiliation(s)
- Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.
- University of Chicago, Chicago, IL, USA.
| | - Chongxian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Harvard University, Boston, MA, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.
- Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
18
|
Roex G, Timmers M, Wouters K, Campillo-Davo D, Flumens D, Schroyens W, Chu Y, Berneman ZN, Lion E, Luo F, Anguille S. Safety and clinical efficacy of BCMA CAR-T-cell therapy in multiple myeloma. J Hematol Oncol 2020; 13:164. [PMID: 33272302 PMCID: PMC7713173 DOI: 10.1186/s13045-020-01001-1] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022] Open
Abstract
Background B-cell maturation antigen (BCMA)-targeted chimeric antigen receptor (CAR)-T-cell therapy is an emerging treatment option for multiple myeloma. The aim of this systematic review and meta-analysis was to determine its safety and clinical activity and to identify factors influencing these outcomes. Methods We performed a database search using the terms “BCMA,” “CAR,” and “multiple myeloma” for clinical studies published between 01/01/2015 and 01/01/2020. The methodology is further detailed in PROSPERO (CRD42020125332). Results Twenty-three different CAR-T-cell products have been used so far in 640 patients. Cytokine release syndrome was observed in 80.3% (69.0–88.2); 10.5% (6.8–16.0) had neurotoxicity. A higher neurotoxicity rate was reported in studies that included more heavily pretreated patients: 19.1% (13.3–26.7; I2 = 45%) versus 2.8% (1.3–6.1; I2 = 0%) (p < 0.0001). The pooled overall response rate was 80.5% (73.5–85.9); complete responses (CR) were observed in 44.8% (35.3–54.6). A pooled CR rate of 71.9% (62.8–79.6; I2 = 0%) was noted in studies using alpaca/llama-based constructs, whereas it was only 18.0% (6.5–41.1; I2 = 67%) in studies that used retroviral vectors for CAR transduction. Median progression-free survival (PFS) was 12.2 (11.4–17.4) months, which compared favorably to the expected PFS of 1.9 (1.5–3.7) months (HR 0.14; p < 0.0001). Conclusions Although considerable toxicity was observed, BCMA-targeted CAR-T-cell therapy is highly efficacious even in advanced multiple myeloma. Subgroup analysis confirmed the anticipated inter-study heterogeneity and identified potential factors contributing to safety and efficacy. The results of this meta-analysis may assist the future design of CAR-T-cell studies and lead to optimized BCMA CAR-T-cell products.
Collapse
Affiliation(s)
- Gils Roex
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Marijke Timmers
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Kristien Wouters
- Clinical Trial Center, Antwerp University Hospital, Edegem, Belgium
| | - Diana Campillo-Davo
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Donovan Flumens
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Wilfried Schroyens
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Yiwei Chu
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Zwi N Berneman
- Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium
| | - Eva Lion
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China.,Department of Digestive Diseases, Huashan Hospital of Fudan University, Shanghai, China
| | - Sébastien Anguille
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute, Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium. .,Division of Hematology and Center for Cell Therapy & Regenerative Medicine, Antwerp University Hospital, Edegem, Belgium.
| |
Collapse
|
19
|
Yan Y, Zeng S, Gong Z, Xu Z. Clinical implication of cellular vaccine in glioma: current advances and future prospects. J Exp Clin Cancer Res 2020; 39:257. [PMID: 33228738 PMCID: PMC7685666 DOI: 10.1186/s13046-020-01778-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023] Open
Abstract
Gliomas, especially glioblastomas, represent one of the most aggressive and difficult-to-treat human brain tumors. In the last few decades, clinical immunotherapy has been developed and has provided exceptional achievements in checkpoint inhibitors and vaccines for cancer treatment. Immunization with cellular vaccines has the advantage of containing specific antigens and acceptable safety to potentially improve cancer therapy. Based on T cells, dendritic cells (DC), tumor cells and natural killer cells, the safety and feasibility of cellular vaccines have been validated in clinical trials for glioma treatment. For TAA engineered T cells, therapy mainly uses chimeric antigen receptors (IL13Rα2, EGFRvIII and HER2) and DNA methylation-induced technology (CT antigen) to activate the immune response. Autologous dendritic cells/tumor antigen vaccine (ADCTA) pulsed with tumor lysate and peptides elicit antigen-specific and cytotoxic T cell responses in patients with malignant gliomas, while its pro-survival effect is biased. Vaccinations using autologous tumor cells modified with TAAs or fusion with fibroblast cells are characterized by both effective humoral and cell-mediated immunity. Even though few therapeutic effects have been observed, most of this therapy showed safety and feasibility, asking for larger cohort studies and better guidelines to optimize cellular vaccine efficiency in anti-glioma therapy.
Collapse
Affiliation(s)
- Yuanliang Yan
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Shuangshuang Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China
| | - Zhijie Xu
- Department of Pathology, Xiangya Hospital, Central South University, 87 Xiangya Road, Hunan, 410008, Changsha, China.
| |
Collapse
|
20
|
Targeted-Alpha-Therapy Combining Astatine-211 and anti-CD138 Antibody in A Preclinical Syngeneic Mouse Model of Multiple Myeloma Minimal Residual Disease. Cancers (Basel) 2020; 12:cancers12092721. [PMID: 32971984 PMCID: PMC7564412 DOI: 10.3390/cancers12092721] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Multiple myeloma is a cancer that remains incurable. Among the many therapies under evaluation, antibodies can be used as vehicles to target and deliver toxic radiation to the tumour cells. Our objective was therefore to investigate the potential of targeted alpha therapy, combining an anti-CD138 mAb with astatine-211, to destroy the residual cells responsible for relapse. We have shown in a mouse model that mimics human disease, that destroying multiple myeloma cells is feasible with low toxicity by injecting an anti-CD138 mAb coupled with astatine-211. This approach could eradicate residual cells after initial treatment and thus prevent recurrence. Abstract Despite therapeutic progress in recent years with the introduction of targeted therapies (daratumumab, elotuzumab), multiple myeloma remains an incurable cancer. The question is therefore to investigate the potential of targeted alpha therapy, combining an anti-CD138 antibody with astatine-211, to destroy the residual cells that cause relapses. A preclinical syngeneic mouse model, consisting of IV injection of 1 million of 5T33 cells in a KaLwRij C57/BL6 mouse, was treated 10 days later with an anti-mCD138 antibody, called 9E7.4, radiolabeled with astatine-211. Four activities of the 211At-9E7.4 radioimmunoconjugate were tested in two independent experiments: 370 kBq (n = 16), 555 kBq (n = 10), 740 kBq (n = 17) and 1100 kBq (n = 6). An isotype control was also tested at 555 kBq (n = 10). Biodistribution, survival rate, hematological parameters, enzymatic hepatic toxicity, histological examination and organ dosimetry were considered. The survival median of untreated mice was 45 days after engraftment. While the activity of 1100 kBq was highly toxic, the activity of 740 kBq offered the best efficacy with 65% of overall survival 150 days after the treatment with no evident sign of toxicity. This work demonstrates the pertinence of treating minimal residual disease of multiple myeloma with an anti-CD138 antibody coupled to astatine-211.
Collapse
|
21
|
Yu B, Jiang T, Liu D. BCMA-targeted immunotherapy for multiple myeloma. J Hematol Oncol 2020; 13:125. [PMID: 32943087 PMCID: PMC7499842 DOI: 10.1186/s13045-020-00962-7] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 09/07/2020] [Indexed: 12/30/2022] Open
Abstract
B cell maturation antigen (BCMA) is a novel treatment target for multiple myeloma (MM) due to its highly selective expression in malignant plasma cells (PCs). Multiple BCMA-targeted therapeutics, including antibody-drug conjugates (ADC), chimeric antigen receptor (CAR)-T cells, and bispecific T cell engagers (BiTE), have achieved remarkable clinical response in patients with relapsed and refractory MM. Belantamab mafodotin-blmf (GSK2857916), a BCMA-targeted ADC, has just been approved for highly refractory MM. In this article, we summarized the molecular and physiological properties of BCMA as well as BCMA-targeted immunotherapeutic agents in different stages of clinical development.
Collapse
Affiliation(s)
- Bo Yu
- Department of Medicine, Lincoln Medical Center, Bronx, NY USA
| | - Tianbo Jiang
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY USA
| | - Delong Liu
- Department of Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY USA
| |
Collapse
|
22
|
Jiao Y, Yi M, Xu L, Chu Q, Yan Y, Luo S, Wu K. CD38: targeted therapy in multiple myeloma and therapeutic potential for solid cancers. Expert Opin Investig Drugs 2020; 29:1295-1308. [PMID: 32822558 DOI: 10.1080/13543784.2020.1814253] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION CD38 is expressed by some cells of hematological malignancies and tumor-related immunosuppressive cells, including regulatory T cells, regulatory B cells, and myeloid-derived suppressor cells. CD38 is an effective target in some hematological malignancies such as multiple myeloma (MM). Daratumumab (Dara), a CD38-targeting antibody, can eliminate CD38high immune suppressor cells and is regarded as a standard therapy for MM because of its outstanding clinical efficacy. Other CD38 monospecific antibodies, such as isatuximab, MOR202, and TAK079, showed promising effects in clinical trials. AREA COVERED This review examines the expression, function, and targeting of CD38 in MM and its potential to deplete immunosuppressive cells in solid cancers. We summarize the distribution and biological function of CD38 and discuss the application of anti-CD38 drugs in hematological malignancies. We also analyz the role of CD38+ immune cells in the tumor microenvironment to encourage additional investigations that target CD38 in solid cancers. PubMed and ClinicalTrials were searched to identify relevant literature from the database inception to 30 April 2020. EXPERT OPINION There is convincing evidence that CD38-targeted immunotherapeutics reduce CD38+ immune suppressor cells. This result suggests that CD38 can be exploited to treat solid tumors by regulating the immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Ying Jiao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| | - Ming Yi
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| | - Linping Xu
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital , Zhengzhou, China
| | - Qian Chu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China
| | - Yongxiang Yan
- R & D Department, Wuhan YZY Biopharma Co., Ltd , Wuhan, China
| | - Suxia Luo
- Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital , Zhengzhou, China
| | - Kongming Wu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology , Wuhan, China.,Department of Medical Oncology, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital , Zhengzhou, China
| |
Collapse
|
23
|
García-Guerrero E, Sierro-Martínez B, Pérez-Simón JA. Overcoming Chimeric Antigen Receptor (CAR) Modified T-Cell Therapy Limitations in Multiple Myeloma. Front Immunol 2020; 11:1128. [PMID: 32582204 PMCID: PMC7290012 DOI: 10.3389/fimmu.2020.01128] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 05/07/2020] [Indexed: 12/11/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable disease regardless of recent advances in the field. Therefore, a substantial unmet need exists to treat patients with relapsed/refractory myeloma. The use of novel agents such as daratumumab, elotuzumab, carfilzomib, or pomalidomide, among others, usually cannot completely eradicate myeloma cells. Although these new drugs have had a significant impact on the prognosis of MM patients, the vast majority ultimately become refractory or can no longer be treated due to toxicity of prior treatment, and thus succumb to the disease. Cellular therapies represent a novel approach with a unique mechanism of action against myeloma with the potential to defeat drug resistance and achieve long-term remissions. Genetic modification of cells to express a novel receptor with tumor antigen specificity is currently being explored in myeloma. Chimeric antigen receptor gene-modified T-cells (CAR T-cells) have shown to be the most promising approach so far. CAR T-cells have shown to induce durable complete remissions in other advanced hematologic malignancies like acute lymphocytic leukemia (ALL) and diffuse large B-cell lymphoma (DLBCL). With this background, significant efforts are underway to develop CAR-based therapies for MM. Currently, several antigen targets, including CD138, CD19, immunoglobulin kappa (Ig-Kappa) and B-cell maturation antigen (BCMA), are being used in clinical trials to treat myeloma patients. Some of these trials have shown promising results, especially in terms of response rates. However, the absence of a plateau is observed in most studies which correlates with the absence of durable remissions. Therefore, several potential limitations such as lack of effectiveness, off-tumor toxicities, and antigen loss or interference with soluble proteins could hamper the efficacy of CAR T-cells in myeloma. In this review, we will focus on clinical outcomes reported with CAR T-cells in myeloma, as well as on CAR T-cell limitations and how to overcome them with next generation of CAR T-cells.
Collapse
Affiliation(s)
- Estefanía García-Guerrero
- Instituto de Biomedicina de Sevilla, UGC de Hematología, Hospital Universitario Virgen del Rocío and Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Universidad de Sevilla, Seville, Spain
| | - Belén Sierro-Martínez
- Instituto de Biomedicina de Sevilla, UGC de Hematología, Hospital Universitario Virgen del Rocío and Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Universidad de Sevilla, Seville, Spain
| | - Jose Antonio Pérez-Simón
- Instituto de Biomedicina de Sevilla, UGC de Hematología, Hospital Universitario Virgen del Rocío and Consejo Superior de Investigaciones Científicas (CSIC) and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Universidad de Sevilla, Seville, Spain
| |
Collapse
|
24
|
Kang L, Zhang J, Li M, Xu N, Qi W, Tan J, Lou X, Yu Z, Sun J, Wang Z, Fu C, Tang X, Dai H, Chen J, Wu D, Yu L. Characterization of novel dual tandem CD19/BCMA chimeric antigen receptor T cells to potentially treat multiple myeloma. Biomark Res 2020; 8:14. [PMID: 32435496 PMCID: PMC7222432 DOI: 10.1186/s40364-020-00192-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 04/27/2020] [Indexed: 02/07/2023] Open
Abstract
Background Treatment with chimeric antigen receptor (CAR)-engineered T cells directed against the B-cell maturation antigen (BCMA) promoted transient recovery from multiple myeloma (MM). However, the absence of this antigen on immature plasma cells may limit the efficacy of this modality and facilitate relapse. The purpose of this study is to characterize a novel CAR that includes both a single-chain variable fragment (scFv)-BCMA and an scFv-CD19 in tandem orientation (tan-CAR) in an attempt to target both BCMA and CD19 expression on MM cells. Method The scFv sequences from the anti-CD19 antibody FMC63 and the anti-BCMA antibody C11D5.3 were ligated in tandem with transmembrane and T-cell signaling domains to generate the tan-CAR construct. Specificity and efficacy of activated tan-CAR T cells were analyzed using in vitro proliferation, cytokine release, and cytolysis assays. We also evaluated the in vivo efficacy with a xenograft mouse model that included target tumor cells that expressed CD19 or BCMA and compared the results to those obtained with conventional CAR T cells. Results The in vitro studies revealed specific activation of tan-CAR T cells by K562 cells that overexpressed CD19 and/or BCMA. Cell proliferation, cytokine release, and cytolytic activity were all comparable to the responses of single scFv CAR T cells. Importantly, in vivo studies of tan-CAR T cells revealed specific inhibition of tumor growth in the mouse xenograft model that included cells expressing both CD19 and BCMA. Systemic administration of tan-CAR T cells resulted in complete tumor remission, in contrast to the reduced efficacies of BCMA-CAR T and CD19-CAR T alone in this setting. Conclusion We report the successful design and execution of novel tan-CAR T cells that promote significant anti-tumor efficacy against both CD19 and BCMA antigen-positive tumor cells in vitro and in vivo. The data from this study reveal a novel strategy that may help to reduce the rate of relapse in the treatment with single scFv-CAR T cells.
Collapse
Affiliation(s)
- Liqing Kang
- 1Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Jian Zhang
- 2National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,3Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Minghao Li
- 1Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Nan Xu
- 1Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Wei Qi
- 1Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Jingwen Tan
- 1Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO, 3663 North Zhongshan Road, Shanghai, 200065 China
| | - Xiaoyan Lou
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| | - Zhou Yu
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| | - Juanjuan Sun
- Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| | - Zhenkun Wang
- 5Central Laboratory of Hematology and Oncology, First Affiliated Hospital, Harbin Medical University, Harbin, 150001 Heilongjiang Province China
| | - Chengcheng Fu
- 2National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,3Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiaowen Tang
- 2National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,3Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Haiping Dai
- 2National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,3Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jia Chen
- 2National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,3Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Depei Wu
- 2National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China.,3Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Lei Yu
- 1Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, NO, 3663 North Zhongshan Road, Shanghai, 200065 China.,Shanghai Unicar-Therapy Bio-medicine Technology Co., Ltd, No 1525 Minqiang Road, Shanghai, 201612 China
| |
Collapse
|
25
|
Leblay N, Maity R, Hasan F, Neri P. Deregulation of Adaptive T Cell Immunity in Multiple Myeloma: Insights Into Mechanisms and Therapeutic Opportunities. Front Oncol 2020; 10:636. [PMID: 32432039 PMCID: PMC7214816 DOI: 10.3389/fonc.2020.00636] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022] Open
Abstract
Immunotherapy has recently emerged as a promising treatment option for multiple myeloma (MM) patients. Profound immune dysfunction and evasion of immune surveillance are known to characterize MM evolution and disease progression. Along with genomic changes observed in malignant plasma cells, the bone marrow (BM) milieu creates a protective environment sustained by the complex interaction of BM stromal cells (BMSCs) and malignant cells that using bidirectional connections and cytokines released stimulate disease progression, drug resistance and enable immune escape. Local immune suppression and T-cell exhaustion are important mediating factors of clinical outcomes and responses to immune-based approaches. Thus, further characterization of the defects present in the immune system of MM patients is essential to develop novel therapies and to repurpose the existing ones. This review seeks to provide insights into the mechanisms that promote tumor escape, cause inadequate T-cell stimulation and impaired cytotoxicity in MM. Furthermore, it highlights current immunotherapies being used to restore adaptive T-cell immune responses in MM and describes strategies created to escape these multiple immune evasion mechanisms.
Collapse
Affiliation(s)
- Noémie Leblay
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Ranjan Maity
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Fajer Hasan
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| | - Paola Neri
- Arnie Charbonneau Cancer Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
26
|
Pan C, Liu H, Robins E, Song W, Liu D, Li Z, Zheng L. Next-generation immuno-oncology agents: current momentum shifts in cancer immunotherapy. J Hematol Oncol 2020; 13:29. [PMID: 32245497 PMCID: PMC7119170 DOI: 10.1186/s13045-020-00862-w] [Citation(s) in RCA: 144] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022] Open
Abstract
Cancer immunotherapy has reached a critical point, now that immune checkpoint inhibitors and two CAR-T products have received market approval in treating 16 types of cancers and 1 tissue-agnostic cancer indication. Accompanying these advances, the 2018 Nobel Prize was awarded for the discovery of immune checkpoint pathways, which has led to the revolution of anti-cancer treatments. However, expanding the indications of immuno-oncology agents and overcoming treatment resistance face mounting challenges. Although combination immunotherapy is an obvious strategy to pursue, the fact that there have been more failures than successes in this effort has served as a wake-up call, placing emphasis on the importance of building a solid scientific foundation for the development of next-generation immuno-oncology (IO) agents. The 2019 China Cancer Immunotherapy Workshop was held to discuss the current challenges and opportunities in IO. At this conference, emerging concepts and strategies for IO development were proposed, focusing squarely on correcting the immunological defects in the tumor microenvironment. New targets such as Siglec-15 and new directions including neoantigens, cancer vaccines, oncolytic viruses, and cytokines were reviewed. Emerging immunotherapies were discussed in the areas of overcoming primary and secondary resistance to existing immune checkpoint inhibitors, activating effector cells, and targeting immunosuppressive mechanisms in the tumor microenvironment. In this article, we highlight old and new waves of IO therapy development, and provide perspectives on the latest momentum shifts in cancer immunotherapy.
Collapse
Affiliation(s)
- Chongxian Pan
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- University of California, Davis, CA, USA
| | - Hongtao Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- University of Chicago, Chicago, IL, USA
| | - Elizabeth Robins
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Wenru Song
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Kira Pharmaceuticals, Cambridge, MA, USA
| | - Delong Liu
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- New York Medical College, Valhalla, NY, USA
| | - Zihai Li
- Chinese American Hematologist and Oncologist Network, New York, NY, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH, USA
| | - Lei Zheng
- Chinese American Hematologist and Oncologist Network, New York, NY, USA.
- Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|