1
|
Fattahi AS, Jafari M, Farahavar G, Abolmaali SS, Tamaddon AM. Expanding horizons in cancer therapy by immunoconjugates targeting tumor microenvironments. Crit Rev Oncol Hematol 2024; 201:104437. [PMID: 38977144 DOI: 10.1016/j.critrevonc.2024.104437] [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: 03/20/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/10/2024] Open
Abstract
Immunoconjugates are promising molecules combining antibodies with different agents, such as toxins, drugs, radionuclides, or cytokines that primarily aim to target tumor cells. However, tumor microenvironment (TME), which comprises a complex network of various cells and molecular cues guiding tumor growth and progression, remains a major challenge for effective cancer therapy. Our review underscores the pivotal role of TME in cancer therapy with immunoconjugates, examining the intricate interactions with TME and recent advancements in TME-targeted immunoconjugates. We explore strategies for targeting TME components, utilizing diverse antibodies such as neutralizing, immunomodulatory, immune checkpoint inhibitors, immunostimulatory, and bispecific antibodies. Additionally, we discuss different immunoconjugates, elucidating their mechanisms of action, advantages, limitations, and applications in cancer immunotherapy. Furthermore, we highlight emerging technologies enhancing the safety and efficacy of immunoconjugates, such as antibody engineering, combination therapies, and nanotechnology. Finally, we summarize current advancements, perspectives, and future developments of TME-targeted immunoconjugates.
Collapse
Affiliation(s)
- Amir Saamaan Fattahi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mahboobeh Jafari
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ghazal Farahavar
- Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery School of Pharmacy, Shiraz University of Medical Sciences, Iran.
| |
Collapse
|
2
|
Lin Z, Zhang Y, Liu X, Luo H, Li Q, Gao Q, Wang X, Wen J, Li L, Feng Y, Wang F, Huang J, Zhai X, Zhang L, Niu T, Zheng Y. Decreased RNA-binding protein heterogeneous nuclear ribonucleoprotein U improves multiple myeloma sensitivity to lenalidomide. Br J Haematol 2024; 205:594-606. [PMID: 38685577 DOI: 10.1111/bjh.19468] [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: 12/08/2023] [Revised: 03/20/2024] [Accepted: 04/03/2024] [Indexed: 05/02/2024]
Abstract
Multiple myeloma (MM) is an incurable plasma cell cancer in the bone marrow. Immunomodulatory drugs, such as lenalidomide (LEN) and pomalidomide, are backbone agents in MM treatment, and LEN resistance is commonly seen in the MM clinic. In this study, we presented that heterogeneous nuclear ribonucleoprotein U (hnRNPU) affected MM resistance to LEN via the regulation of target mRNA translation. hnRNPULow MM cells exhibited upregulated CRBN and IKZF1 proteins, stringent IKZF1/3 protein degradation upon LEN addition and increased sensitivity to LEN. RNA pulldown assays and RNA electrophoretic mobility shift assays revealed that hnRNPU bound to the 3'-untranslated region of CRBN and IKZF1 mRNA. A sucrose gradient assay suggested that hnRNPU specifically regulated CRBN and IKZF1 mRNA translation. The competition of hnRNPU binding to its target mRNAs by small RNAs with hnRNPU-binding sites restored MM sensitivity to LEN. hnRNPU function in vivo was confirmed in an immunocompetent MM mouse model constructed by the inoculation of Crbn-humanized murine 5TGM1 cells into CrbnI391V/+ mice. Overall, this study suggests a novel mechanism of LEN sensitivity in which hnRNPU represses CRBN and IKZF1 mRNA translation.
Collapse
Affiliation(s)
- Zhimei Lin
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
- Department of Hematology, The Affiliated Hospital of Chengdu University, Chengdu, China
| | - Yue Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiang Liu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Hongmei Luo
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Qianwen Gao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xin Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingjing Wen
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
- Department of Hematology, Mian-Yang Central Hospital, Mianyang, China
| | - Linfeng Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yu Feng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Fangfang Wang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Jingcao Huang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xinyu Zhai
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhang
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Yuhuan Zheng
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Lu Q, Yang D, Li H, Niu T, Tong A. Multiple myeloma: signaling pathways and targeted therapy. MOLECULAR BIOMEDICINE 2024; 5:25. [PMID: 38961036 PMCID: PMC11222366 DOI: 10.1186/s43556-024-00188-w] [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: 03/18/2024] [Accepted: 05/21/2024] [Indexed: 07/05/2024] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy of plasma cells, characterized by osteolytic bone lesions, anemia, hypercalcemia, renal failure, and the accumulation of malignant plasma cells. The pathogenesis of MM involves the interaction between MM cells and the bone marrow microenvironment through soluble cytokines and cell adhesion molecules, which activate various signaling pathways such as PI3K/AKT/mTOR, RAS/MAPK, JAK/STAT, Wnt/β-catenin, and NF-κB pathways. Aberrant activation of these pathways contributes to the proliferation, survival, migration, and drug resistance of myeloma cells, making them attractive targets for therapeutic intervention. Currently, approved drugs targeting these signaling pathways in MM are limited, with many inhibitors and inducers still in preclinical or clinical research stages. Therapeutic options for MM include non-targeted drugs like alkylating agents, corticosteroids, immunomodulatory drugs, proteasome inhibitors, and histone deacetylase inhibitors. Additionally, targeted drugs such as monoclonal antibodies, chimeric antigen receptor T cells, bispecific T-cell engagers, and bispecific antibodies are being used in MM treatment. Despite significant advancements in MM treatment, the disease remains incurable, emphasizing the need for the development of novel or combined targeted therapies based on emerging theoretical knowledge, technologies, and platforms. In this review, we highlight the key role of signaling pathways in the malignant progression and treatment of MM, exploring advances in targeted therapy and potential treatments to offer further insights for improving MM management and outcomes.
Collapse
Affiliation(s)
- Qizhong Lu
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Donghui Yang
- College of Veterinary Medicine, Shaanxi Center of Stem Cells Engineering and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hexian Li
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Aiping Tong
- State Key Laboratory of Biotherapy and Cancer Center, Research Unit of Gene and Immunotherapy, Chinese Academy of Medical Sciences, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, 610212, China.
| |
Collapse
|
4
|
Wang H, Wang L, Luan H, Xiao J, Zhao Z, Yu P, Deng M, Liu Y, Ji S, Ma J, Zhou Y, Zhang J, Meng X, Zhang J, Zhao X, Li C, Li F, Wang D, Wei S, Hui L, Nie S, Jin C, An Z, Zhang N, Wang Y, Zhang CC, Li Z. LILRB4 on multiple myeloma cells promotes bone lesion by p-SHP2/NF-κB/RELT signal pathway. J Exp Clin Cancer Res 2024; 43:183. [PMID: 38951916 PMCID: PMC11218313 DOI: 10.1186/s13046-024-03110-y] [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: 04/21/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Leukocyte Ig-like receptor B family 4 (LILRB4) as an immune checkpoint on myeloid cells is a potential target for tumor therapy. Extensive osteolytic bone lesion is the most characteristic feature of multiple myeloma. It is unclear whether ectopic LILRB4 on multiple myeloma regulates bone lesion. METHODS The conditioned medium (CM) from LILRB4-WT and -KO cells was used to analyze the effects of LILRB4 on osteoclasts and osteoblasts. Xenograft, syngeneic and patient derived xenograft models were constructed, and micro-CT, H&E staining were used to observe the bone lesion. RNA-seq, cytokine array, qPCR, the activity of luciferase, Co-IP and western blotting were used to clarify the mechanism by which LILRB4 mediated bone damage in multiple myeloma. RESULTS We comprehensively analyzed the expression of LILRB4 in various tumor tissue arrays, and found that LILRB4 was highly expressed in multiple myeloma samples. The patient's imaging data showed that the higher the expression level of LILRB4, the more serious the bone lesion in patients with multiple myeloma. The conditioned medium from LILRB4-WT not -KO cells could significantly promote the differentiation and maturation of osteoclasts. Xenograft, syngeneic and patient derived xenograft models furtherly confirmed that LILRB4 could mediate bone lesion of multiple myeloma. Next, cytokine array was performed to identify the differentially expressed cytokines, and RELT was identified and regulated by LILRB4. The overexpression or exogenous RELT could regenerate the bone damage in LILRB4-KO cells in vitro and in vivo. The deletion of LILRB4, anti-LILRB4 alone or in combination with bortezomib could significantly delay the progression of bone lesion of multiple myeloma. CONCLUSIONS Our findings indicated that LILRB4 promoted the bone lesion by promoting the differentiation and mature of osteoclasts through secreting RELT, and blocking LILRB4 singling pathway could inhibit the bone lesion.
Collapse
Affiliation(s)
- Hongying Wang
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Lei Wang
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Huiwen Luan
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Jing Xiao
- Department of Hematology, Yantaishan Hospital, Yantai, Shandong, 264003, P.R. China
| | - Zhiling Zhao
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Pengfei Yu
- Department of Biopharmaceutical, School of Pharmacy, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Mi Deng
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA
- Peking University International Cancer Institute, Peking University, CN 38 Xueyuan Rd. Haidian Dis., Beijing, 100191, P.R. China
| | - Yifan Liu
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Shuhao Ji
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Junjie Ma
- Department of Hematology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, 264009, P.R. China
| | - Yan Zhou
- Department of Gastrointestinalstrointestinal Surgery, Yantaishan Hospital, Yantai, Shandong, 264003, P.R. China
| | - Jiashen Zhang
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Shandong Agricultural University, Taian, Shandong, 271018, P.R. China
| | - Xianhui Meng
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Juan Zhang
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Xinyu Zhao
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Chunling Li
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Fangmin Li
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Dapeng Wang
- Department of Pathophysiology, Bengbu Medical College, Anhui, 233000, P.R. China
| | - Shujuan Wei
- R&D Center, Luye Pharma Group, Yantai, Shandong, 264005, P.R. China
| | - Lijun Hui
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Siman Nie
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Changzhu Jin
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China
| | - Zhiqiang An
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Ningyan Zhang
- Texas Therapeutics Institute, Brown Foundation Institute of Molecular Medicine, University of Texas Health Science Center, Houston, TX, 77030, USA
| | - Yaopeng Wang
- Department of Thoracic Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, 266011, P.R. China.
| | - Cheng Cheng Zhang
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390, USA.
| | - Zunling Li
- Department of Biochemistry and Molecular Biology, Shandong Tumour Immunotherapy Research Innovation Team, Binzhou Medical University, Yantai, Shandong, 264003, P.R. China.
| |
Collapse
|
5
|
Murphy K, Shi Q, Hoover DR, Adimora AA, Alcaide ML, Brockmann S, Daubert E, Duggal P, Merenstein D, Dionne JA, Sheth AN, Keller MJ, Herold BC, Anastos K, Aouizerat B. Genetic predictors for bacterial vaginosis in women living with and at risk for HIV infection. Am J Reprod Immunol 2024; 91:e13845. [PMID: 38720636 PMCID: PMC11410097 DOI: 10.1111/aji.13845] [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: 08/31/2023] [Revised: 03/24/2024] [Accepted: 04/03/2024] [Indexed: 08/29/2024] Open
Abstract
PROBLEM Bacterial vaginosis (BV) disproportionally impacts Black and Hispanic women, placing them at risk for HIV, sexually transmitted infections and preterm birth. It is unknown whether there are differences by genetic ancestry in BV risk or whether polymorphisms associated with BV risk differ by ancestry. METHODS Women's Interagency HIV Study (WIHS) participants with longitudinal Nugent scores were dichotomized as having (n = 319, Nugent 7-10) or not having BV (n = 367, Nugent 0-3). Genetic ancestry was defined by clustering of principal components from ancestry informative markers and further stratified by BV status. 627 single nucleotide polymorphisms (SNPs) across 41 genes important in mucosal defense were identified in the WIHS GWAS. A logistic regression analysis was adjusted for nongenetic predictors of BV and self-reported race/ethnicity to assess associations between genetic ancestry and genotype. RESULTS Self-reported race and genetic ancestry were associated with BV risk after adjustment for behavioral factors. Polymorphisms in mucosal defense genes including syndecans, cytokines and toll-like receptors (TLRs) were associated with BV in all ancestral groups. CONCLUSIONS The common association of syndecan, cytokine and TLR genes and the importance of immune function and inflammatory pathways in BV, suggests these should be targeted for further research on BV pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Kerry Murphy
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Quihu Shi
- School of Health Sciences and Practice, New York Medical College, Valhalla, New York, USA
| | - Donald R Hoover
- Department of Statistics and Institute for Health, Health Care Policy and Aging Research Rutgers the State University of New Jersey, Piscataway, New Jersey, USA
| | - Adaora A Adimora
- Department of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Maria L Alcaide
- Department of Medicine, Obstetrics & Gynecology and Public Health, University of Miami Miller School of Medicine, Miami, USA
| | - Susan Brockmann
- Health Sciences Center, State University of New York, Brooklyn, New York, USA
| | - Elizabeth Daubert
- Cook County Health/Hektoen Institute of Medicine, Chicago, Illinois, USA
| | - Priya Duggal
- Department of Epidemiology and International Health, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Jodie A Dionne
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Anandi N Sheth
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marla J Keller
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Betsy C Herold
- Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, USA
- Pediatrics, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kathryn Anastos
- Departments of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
- Obstetrics & Gynecology and Women's Health, Albert Einstein College of Medicine, Bronx, New York, USA
- Epidemiology & Population Health, Albert Einstein College of Medicine, Bronx, New York, USA
| | | |
Collapse
|
6
|
Riccardi F, Tangredi C, Dal Bo M, Toffoli G. Targeted therapy for multiple myeloma: an overview on CD138-based strategies. Front Oncol 2024; 14:1370854. [PMID: 38655136 PMCID: PMC11035824 DOI: 10.3389/fonc.2024.1370854] [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: 01/15/2024] [Accepted: 03/28/2024] [Indexed: 04/26/2024] Open
Abstract
Multiple myeloma (MM) is an incurable hematological disease characterized by the uncontrolled growth of plasma cells primarily in the bone marrow. Although its treatment consists of the administration of combined therapy regimens mainly based on immunomodulators and proteosome inhibitors, MM remains incurable, and most patients suffer from relapsed/refractory disease with poor prognosis and survival. The robust results achieved by immunotherapy targeting MM-associated antigens CD38 and CD319 (also known as SLAMF7) have drawn attention to the development of new immune-based strategies and different innovative compounds in the treatment of MM, including new monoclonal antibodies, antibody-drug conjugates, recombinant proteins, synthetic peptides, and adaptive cellular therapies. In this context, Syndecan1 (CD138 or SDC1), a transmembrane heparan sulfate proteoglycan that is upregulated in malignant plasma cells, has gained increasing attention in the panorama of MM target antigens, since its key role in MM tumorigenesis, progression and aggressiveness has been largely reported. Here, our aim is to provide an overview of the most important aspects of MM disease and to investigate the molecular functions of CD138 in physiologic and malignant cell states. In addition, we will shed light on the CD138-based therapeutic approaches currently being tested in preclinical and/or clinical phases in MM and discuss their properties, mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Federico Riccardi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Carmela Tangredi
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Michele Dal Bo
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| | - Giuseppe Toffoli
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
7
|
Li G, Ma L, Feng C, Yin H, Bao J, Wu D, Zhang Z, Li X, Li Z, Yang C, Wang H, Fang F, Hu X, Li M, Xu L, Xu Y, Liang H, Yang T, Wang J, Pan J. MZ1, a BRD4 inhibitor, exerted its anti-cancer effects by suppressing SDC1 in glioblastoma. BMC Cancer 2024; 24:220. [PMID: 38365636 PMCID: PMC10870565 DOI: 10.1186/s12885-024-11966-8] [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: 12/10/2023] [Accepted: 02/06/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Glioblastoma (GBM) is a relatively prevalent primary tumor of the central nervous system in children, characterized by its high malignancy and mortality rates, along with the intricate challenges of achieving complete surgical resection. Recently, an increasing number of studies have focused on the crucial role of super-enhancers (SEs) in the occurrence and development of GBM. This study embarks on the task of evaluating the effectiveness of MZ1, an inhibitor of BRD4 meticulously designed to specifically target SEs, within the intricate framework of GBM. METHODS The clinical data of GBM patients was sourced from the Chinese Glioma Genome Atlas (CGGA) and the Gene Expression Profiling Interactive Analysis 2 (GEPIA2), and the gene expression data of tumor cell lines was derived from the Cancer Cell Line Encyclopedia (CCLE). The impact of MZ1 on GBM was assessed through CCK-8, colony formation assays, EdU incorporation analysis, flow cytometry, and xenograft mouse models. The underlying mechanism was investigated through RNA-seq and ChIP-seq analyses. RESULTS In this investigation, we made a noteworthy observation that MZ1 exhibited a substantial reduction in the proliferation of GBM cells by effectively degrading BRD4. Additionally, MZ1 displayed a notable capability in inducing significant cell cycle arrest and apoptosis in GBM cells. These findings were in line with our in vitro outcomes. Notably, MZ1 administration resulted in a remarkable decrease in tumor size within the xenograft model with diminished toxicity. Furthermore, on a mechanistic level, the administration of MZ1 resulted in a significant suppression of pivotal genes closely associated with cell cycle regulation and epithelial-mesenchymal transition (EMT). Interestingly, our analysis of RNA-seq and ChIP-seq data unveiled the discovery of a novel prospective oncogene, SDC1, which assumed a pivotal role in the tumorigenesis and progression of GBM. CONCLUSION In summary, our findings revealed that MZ1 effectively disrupted the aberrant transcriptional regulation of oncogenes in GBM by degradation of BRD4. This positions MZ1 as a promising candidate in the realm of therapeutic options for GBM treatment.
Collapse
Affiliation(s)
- Gen Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Liya Ma
- College of Basic Medicine and Forensic Medicine, Henan University of Science and Technology, Luoyang, 471023, P.R. China
| | - Chenxi Feng
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Hongli Yin
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Jianping Bao
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Di Wu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Zimu Zhang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Xiaolu Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Zhiheng Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Chun Yang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Hairong Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Fang Fang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Xiaohan Hu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Lixiao Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Yunyun Xu
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China
| | - Hansi Liang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, P.R. China
| | - Tianquan Yang
- Department of Neurosurgery, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China.
| | - Jianwei Wang
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China.
| | - Jian Pan
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, P.R. China.
| |
Collapse
|
8
|
Ughade PA, Shrivastava D. Unveiling the Role of Endometrial CD-138: A Comprehensive Review on Its Significance in Infertility and Early Pregnancy. Cureus 2024; 16:e54782. [PMID: 38529432 PMCID: PMC10961243 DOI: 10.7759/cureus.54782] [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: 12/24/2023] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
This review comprehensively examines the role of endometrial CD-138 (syndecan-1) in the context of infertility and early pregnancy. The endometrium, a dynamic tissue responsive to hormonal cues, plays a central role in fertility, and understanding the molecular intricacies governing its function is crucial. CD-138, a cell surface proteoglycan, emerges as a critical player expressed by various endometrial cell types. Our exploration encompasses a brief overview of the endometrium, introducing CD-138 as a significant molecular entity. The rationale for the review underscores the importance of elucidating endometrial factors in fertility and addresses existing knowledge gaps related to CD-138. Throughout the review, we unravel the multifaceted nature of CD-138 and its involvement in infertility, highlighting its potential as a diagnostic marker. Furthermore, insights into CD-138's role during early pregnancy, including trophoblast-endothelial interactions, are discussed. In conclusion, the findings underscore the clinical implications of CD-138, suggesting its utility in diagnostics and offering prospects for targeted therapeutic interventions. The identified knowledge gaps propel future research directions, promising to deepen our understanding of this enigmatic molecule and its transformative potential in reproductive medicine.
Collapse
Affiliation(s)
- Prachi A Ughade
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| | - Deepti Shrivastava
- Obstetrics and Gynaecology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education & Research, Wardha, IND
| |
Collapse
|
9
|
Wemlinger SM, Cambier JC. Therapeutic tactics for targeting B lymphocytes in autoimmunity and cancer. Eur J Immunol 2024; 54:e2249947. [PMID: 37816494 DOI: 10.1002/eji.202249947] [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: 01/13/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
B lymphocytes have become a very popular therapeutic target in a number of autoimmune indications due to their newly appreciated roles, and approachability, in these diseases. Many of the therapies now applied in autoimmunity were initially developed to deplete malignant B cells. These strategies have also been found to benefit patients suffering from such autoimmune diseases as multiple sclerosis, type I diabetes, systemic lupus erythematosus, and rheumatoid arthritis, to name a few. These observations have supported the expansion of research addressing the mechanistic contributions of B cells in these diseases, as well as blossoming of therapeutics that target them. This review seeks to summarize cutting-edge modalities for targeting B cells, including monoclonal antibodies, bispecific antibodies, antibody-drug conjugates, chimeric antigen receptor-T cells, and small molecule inhibitors. Efforts to refine B-cell targeted therapy to eliminate only pathogenic autoreactive cells will be addressed as well as the potential for future B-cell-based cellular therapeutics. Finally, we also address approaches that seek to silence B-cell function without depletion.
Collapse
Affiliation(s)
- Scott M Wemlinger
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
10
|
Ray U, Orlowski RZ. Antibody-Drug Conjugates for Multiple Myeloma: Just the Beginning, or the Beginning of the End? Pharmaceuticals (Basel) 2023; 16:ph16040590. [PMID: 37111346 PMCID: PMC10145905 DOI: 10.3390/ph16040590] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/17/2023] [Accepted: 03/27/2023] [Indexed: 04/29/2023] Open
Abstract
Multiple myeloma is a malignancy of immunoglobulin-secreting plasma cells that is now often treated in the newly diagnosed and relapsed and/or refractory settings with monoclonal antibodies targeting lineage-specific markers used either alone or in rationally designed combination regimens. Among these are the anti-CD38 antibodies daratumumab and isatuximab, and the anti-Signaling lymphocytic activation molecule family member 7 antibody elotuzumab, all of which are used in their unconjugated formats. Single-chain variable fragments from antibodies also form a key element of the chimeric antigen receptors (CARs) in the B-cell maturation antigen (BCMA)-targeted CAR T-cell products idecabtagene vicleucel and ciltacabtagene autoleucel, which are approved in the advanced setting. Most recently, the bispecific anti-BCMA and T-cell-engaging antibody teclistamab has become available, again for patients with relapsed/refractory disease. Another format into which antibodies can be converted to exert anti-tumor efficacy is as antibody-drug conjugates (ADCs), and belantamab mafodotin, which also targets BCMA, represented the first such agent that gained a foothold in myeloma. Negative results from a recent Phase III study have prompted the initiation of a process for withdrawal of its marketing authorization. However, belantamab remains a drug with some promise, and many other ADCs targeting either BCMA or other plasma cell surface markers are in development and showing potential. This contribution will provide an overview of some of the current data supporting the possibility that ADCs will remain a part of our chemotherapeutic armamentarium against myeloma moving forward, and also highlight areas for future development.
Collapse
Affiliation(s)
- Upasana Ray
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
| | - Robert Z Orlowski
- Department of Lymphoma/Myeloma, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX 77030-4009, USA
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 429, Houston, TX 77030-4009, USA
| |
Collapse
|
11
|
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: 4.0] [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
|
12
|
Yang H, Wang L. Heparan sulfate proteoglycans in cancer: Pathogenesis and therapeutic potential. Adv Cancer Res 2023; 157:251-291. [PMID: 36725112 DOI: 10.1016/bs.acr.2022.08.001] [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] [Indexed: 02/04/2023]
Abstract
The heparan sulfate proteoglycans (HSPGs) are glycoproteins that consist of a proteoglycan "core" protein and covalently attached heparan sulfate (HS) chain. HSPGs are ubiquitously expressed in mammalian cells on the cell surface and in the extracellular matrix (ECM) and secretory vesicles. Within HSPGs, the protein cores determine when and where HSPG expression takes place, and the HS chains mediate most of HSPG's biological roles through binding various protein ligands, including cytokines, chemokines, growth factors and receptors, morphogens, proteases, protease inhibitors, and ECM proteins. Through these interactions, HSPGs modulate cell proliferation, adhesion, migration, invasion, and angiogenesis to display essential functions in physiology and pathology. Under physiological conditions, the expression and localization of HSPGs are finely regulated to orchestrate their physiological functions, and this is disrupted in cancer. The HSPG dysregulation elicits multiple oncogenic signaling, including growth factor signaling, ECM and Integrin signaling, chemokine and immune signaling, cancer stem cell, cell differentiation, apoptosis, and senescence, to prompt cell transformation, proliferation, tumor invasion and metastasis, tumor angiogenesis and inflammation, and immunotolerance. These oncogenic roles make HSPGs an attractive pharmacological target for anti-cancer therapy. Several therapeutic strategies have been under development, including anti-HSPG antibodies, peptides and HS mimetics, synthetic xylosides, and heparinase inhibitors, and shown promising anti-cancer efficacy. Therefore, much progress has been made in this line of study. However, it needs to bear in mind that the roles of HSPGs in cancer can be either oncogenic or tumor-suppressive, depending on the HSPG and the cancer cell type with the underlying mechanisms that remain obscure. Further studies need to address these to fill the knowledge gap and rationalize more efficient therapeutic targeting.
Collapse
Affiliation(s)
- Hua Yang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Lianchun Wang
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Bryd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States.
| |
Collapse
|
13
|
Park HE, Oh H, Baek JH. Interleukin-34-regulated T-cell responses in rheumatoid arthritis. Front Med (Lausanne) 2022; 9:1078350. [PMID: 36530919 PMCID: PMC9747768 DOI: 10.3389/fmed.2022.1078350] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 11/14/2022] [Indexed: 09/10/2023] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a multifaceted etiology, which primarily affects and results in the deterioration of the synovium of patients. While the exact etiology of RA is still largely unknown, there is growing interest in the cytokine interleukin-34 (IL-34) as a driver or modulator of RA pathogenesis on the grounds that IL-34 is drastically increased in the serum and synovium of RA patients. Several studies have so far revealed the relationship between IL-34 levels and RA disease progression. Nevertheless, the significance and role of IL-34 in RA have remained ambiguous, as illustrated by two most recent studies, which reported contrasting effects of genetic IL-34 deletion in RA. Of note, IL-34 is a macrophage growth factor and is increasingly perceived as a master regulator of T-cell responses in RA via macrophage-dependent as well as T cell-intrinsic mechanisms. In this regard, several studies have demonstrated that IL-34 potentiates helper T-cell (Th) responses in RA, whereas studies also suggested that IL-34 alleviates synovial inflammation, potentially by inducing regulatory T-cells (Treg). Herein, we provide an overview of the current understanding of IL-34 involvement in RA and outline IL-34-mediated mechanisms in regulating T-cell responses in RA.
Collapse
Affiliation(s)
| | | | - Jea-Hyun Baek
- School of Life Science, Handong Global University, Pohang, South Korea
| |
Collapse
|
14
|
Cho SF, Yeh TJ, Anderson KC, Tai YT. Bispecific antibodies in multiple myeloma treatment: A journey in progress. Front Oncol 2022; 12:1032775. [PMID: 36330495 PMCID: PMC9623099 DOI: 10.3389/fonc.2022.1032775] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/03/2022] [Indexed: 07/29/2023] Open
Abstract
The incorporation of novel agents and monoclonal antibody-based therapies into the treatment of multiple myeloma (MM) has significantly improved long-term patient survival. However, the disease is still largely incurable, with high-risk patients suffering shorter survival times, partly due to weakened immune systems. Bispecific molecules, including bispecific antibodies (BisAbs) and bispecific T-cell engagers (BiTEs), encourage immune cells to lyse MM cells by simultaneously binding antigens on MM cells and immune effector cells, bringing those cells into close proximity. BisAbs that target B-cell maturation antigen (BCMA) and GPRC5D have shown impressive clinical activity, and the results of early-phase clinical trials targeting FcRH5 in patients with relapsed/refractory MM (RRMM) are also promising. Furthermore, the safety profile of these agents is favorable, including mainly low-grade cytokine release syndrome (CRS). These off-the-shelf bispecific molecules will likely become an essential part of the MM treatment paradigm. Here, we summarize and highlight various bispecific immunotherapies under development in MM treatment, as well as the utility of combining them with current standard-of-care treatments and new strategies. With the advancement of novel combination treatment approaches, these bispecific molecules may lead the way to a cure for MM.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tsung-Jang Yeh
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Kenneth C. Anderson
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Yu-Tzu Tai
- LeBow Institute for Myeloma Therapeutics and Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
15
|
Meyer A, Sienes R, Zanotti B, van Raemdonck K, Palasiewicz K, Mass DP, Volin MV, Shahrara S. Dysregulation of IL-34 ligation to SDC-1 mitigates collagen-induced arthritis. Cell Mol Immunol 2022; 19:1070-1072. [PMID: 35676327 PMCID: PMC9424265 DOI: 10.1038/s41423-022-00886-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 05/24/2022] [Indexed: 11/08/2022] Open
Affiliation(s)
- Anja Meyer
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Ryan Sienes
- Jesse Brown VA Medical Center, Chicago, IL, USA
| | - Brian Zanotti
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Katrien van Raemdonck
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Karol Palasiewicz
- Jesse Brown VA Medical Center, Chicago, IL, USA
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA
| | - Daniel P Mass
- Department of Orthopedic Surgery, The University of Illinois at Chicago, Chicago, IL, USA
| | - Michael V Volin
- Department of Microbiology and Immunology, Midwestern University, Downers Grove, IL, USA
| | - Shiva Shahrara
- Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Medicine, Division of Rheumatology, The University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
16
|
Mayer M, Nachtsheim L, Hoffmann F, von Eggeling F, Guntinas-Lichius O, Prinz J, Klußmann JP, Quaas A, Arolt C, Wolber P. CD138 Is Expressed in Different Entities of Salivary Gland Cancer and Their Lymph Node Metastases and Therefore Represents a Potential Therapeutic Target. Int J Mol Sci 2022; 23:ijms23169037. [PMID: 36012301 PMCID: PMC9409357 DOI: 10.3390/ijms23169037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 12/12/2022] Open
Abstract
Advanced salivary gland carcinomas (SGC) often lack therapeutic options. Agents targeting CD138 have recently shown promising results in clinical trials for multiple myeloma and a preclinical trial for triple-negative breast cancer. Immunohistochemistry for CD138 was performed for all patients who had undergone primary surgery for SGC with curative intent. Findings were validated using matrix-assisted laser desorption/ionization mass spectrometry (MALDI-MS) imaging. Overall, 111 primary SGC and 13 lymph node metastases from salivary duct carcinomas (SaDu) were evaluated. CD138 expression was found in 60% of all SGC with differing expression across entities (p < 0.01). A mean of 25.2% of the tumor cells in mucoepidermoid carcinoma (MuEp) were positive, followed by epithelial-myoepithelial carcinoma (20.9%), acinic cell carcinoma (16.0%), and SaDu (15.2%). High-/intermediate-grade MuEp showed CD138 expression in a mean of 34.8% of tumor cells. For SaDu, lymph node metastases showed CD138 expression in a mean of 31.2% of tumor cells which correlated with CD138 expression in their primaries (p = 0.01; Spearman’s ρ = 0.71). MALDI-MS imaging confirmed the presence of the CD138 protein in SGC. No significant association was found between clinicopathological data, including progression-free survival (p = 0.50) and CD138 expression. CD138 is expressed in the cell membrane of different entities of SGC and SaDu lymph node metastases and therefore represents a potential target for CD138 targeting drugs.
Collapse
Affiliation(s)
- Marcel Mayer
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
- Correspondence: ; Tel.: +49-22147877840
| | - Lisa Nachtsheim
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Franziska Hoffmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
- Department of Otorhinolaryngology, MALDI Imaging and Innovative Biophotonics, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Ferdinand von Eggeling
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
- Department of Otorhinolaryngology, MALDI Imaging and Innovative Biophotonics, Jena University Hospital, Friedrich-Schiller-University, 07743 Jena, Germany
| | - Orlando Guntinas-Lichius
- Department of Otorhinolaryngology, Head and Neck Surgery, Jena University Hospital, Friedrich-Schiller-University, 07747 Jena, Germany
| | - Johanna Prinz
- Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, University of Cologne, 50937 Cologne, Germany
| | - Jens Peter Klußmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Alexander Quaas
- Medical Faculty, Institute of Pathology, University of Cologne, 50937 Cologne, Germany
| | - Christoph Arolt
- Medical Faculty, Institute of Pathology, University of Cologne, 50937 Cologne, Germany
| | - Philipp Wolber
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
17
|
Noborn F, Nilsson J, Larson G. Site-specific glycosylation of proteoglycans: a revisited frontier in proteoglycan research. Matrix Biol 2022; 111:289-306. [PMID: 35840015 DOI: 10.1016/j.matbio.2022.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 06/11/2022] [Accepted: 07/11/2022] [Indexed: 11/29/2022]
Abstract
Proteoglycans (PGs), a class of carbohydrate-modified proteins, are present in essentially all metazoan organisms investigated to date. PGs are composed of glycosaminoglycan (GAG) chains attached to various core proteins and are important for embryogenesis and normal homeostasis. PGs exert many of their functions via their GAG chains and understanding the details of GAG-ligand interactions has been an essential part of PG research. Although PGs are also involved in many diseases, the number of GAG-related drugs used in the clinic is yet very limited, indicating a lack of detailed structure-function understanding. Structural analysis of PGs has traditionally been obtained by first separating the GAG chains from the core proteins, after which the two components are analyzed separately. While this strategy greatly facilitates the analysis, it precludes site-specific information and introduces either a "GAG" or a "core protein" perspective on the data interpretation. Mass-spectrometric (MS) glycoproteomic approaches have recently been introduced, providing site-specific information on PGs. Such methods have revealed a previously unknown structural complexity of the GAG linkage regions and resulted in identification of several novel CSPGs and HSPGs in humans and in model organisms, thereby expanding our view on PG complexity. In light of these findings, we discuss here if the use of such MS-based techniques, in combination with various functional assays, can also be used to expand our functional understanding of PGs. We have also summarized the site-specific information of all human PGs known to date, providing a theoretical framework for future studies on site-specific functional analysis of PGs in human pathophysiology.
Collapse
Affiliation(s)
- Fredrik Noborn
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Department of Laboratory Medicine, Sundsvall County Hospital, Sweden.
| | - Jonas Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden; Proteomics Core Facility, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Göran Larson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
18
|
Yang Z, Chen S, Ying H, Yao W. Targeting syndecan-1: new opportunities in cancer therapy. Am J Physiol Cell Physiol 2022; 323:C29-C45. [PMID: 35584326 PMCID: PMC9236862 DOI: 10.1152/ajpcell.00024.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 12/02/2022]
Abstract
Syndecan-1 (SDC1, CD138) is one of the heparan sulfate proteoglycans and is essential for maintaining normal cell morphology, interacting with the extracellular and intracellular protein repertoire, as well as mediating signaling transduction upon environmental stimuli. The critical role of SDC1 in promoting tumorigenesis and metastasis has been increasingly recognized in various cancer types, implying a promising potential of utilizing SDC1 as a novel target for cancer therapy. This review summarizes the current knowledge on SDC1 structure and functions, including its role in tumor biology. We also discuss the highlights and limitations of current SDC1-targeted therapies as well as the obstacles in developing new therapeutic methods, offering our perspective on the future directions to target SDC1 for cancer treatment.
Collapse
Affiliation(s)
- Zecheng Yang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Shuaitong Chen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- UTHealth Graduate School of Biomedical Sciences, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Haoqiang Ying
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Wantong Yao
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
19
|
Aghanejad A, Bonab SF, Sepehri M, Haghighi FS, Tarighatnia A, Kreiter C, Nader ND, Tohidkia MR. A review on targeting tumor microenvironment: The main paradigm shift in the mAb-based immunotherapy of solid tumors. Int J Biol Macromol 2022; 207:592-610. [PMID: 35296439 DOI: 10.1016/j.ijbiomac.2022.03.057] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 03/09/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022]
Abstract
Monoclonal antibodies (mAbs) as biological macromolecules have been remarked the large and growing pipline of the pharmaceutical market and also the most promising tool in modern medicine for cancer therapy. These therapeutic entities, which consist of whole mAbs, armed mAbs (i.e., antibody-toxin conjugates, antibody-drug conjugates, and antibody-radionuclide conjugates), and antibody fragments, mostly target tumor cells. However, due to intrinsic heterogeneity of cancer diseases, tumor cells targeting mAb have been encountered with difficulties in their unpredictable efficacy as well as variability in remission and durable clinical benefits among cancer patients. To address these pitfalls, the area has undergone two major evolutions with the intent of minimizing anti-drug responses and addressing limitations experienced with tumor cell-targeted therapies. As a novel hallmark of cancer, the tumor microenvironment (TME) is becoming the great importance of attention to develop innovative strategies based on therapeutic mAbs. Here, we underscore innovative strategies targeting TME by mAbs which destroy tumor cells indirectly through targeting vasculature system (e.g., anti-angiogenesis), immune system modulation (i.e., stimulation, suppression, and depletion), the targeting and blocking of stroma-based growth signals (e.g., cancer-associated fibroblasts), and targeting cancer stem cells, as well as, their effector mechanisms, clinical uses, and relevant mechanisms of resistance.
Collapse
Affiliation(s)
- Ayuob Aghanejad
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Farashi Bonab
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Sepehri
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Fatemeh Sadat Haghighi
- Yazd Diabetes Research Center, Shahid Sadoghi University of Medical Sciences, Yazd, Iran
| | - Ali Tarighatnia
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Christopher Kreiter
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Nader D Nader
- Department of Anesthesiology, University at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
20
|
Clara JA, Childs RW. Harnessing natural killer cells for the treatment of multiple myeloma. Semin Oncol 2022; 49:69-85. [DOI: 10.1053/j.seminoncol.2022.01.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 01/08/2022] [Indexed: 12/11/2022]
|
21
|
Cho SF, Xing L, Anderson KC, Tai YT. Promising Antigens for the New Frontier of Targeted Immunotherapy in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13236136. [PMID: 34885245 PMCID: PMC8657018 DOI: 10.3390/cancers13236136] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/29/2021] [Accepted: 12/03/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Defining the specificity and biological sequalae induced by receptors differentiated expressed in multiple myeloma cells are critical for the development of effective immunotherapies based on monoclonal antibodies. Ongoing studies continue to discover new antigens with superior tumor selectivity and defined function in regulating the pathophysiology of myeloma cells directly or indirectly in the immunosuppressive bone marrow microenvironment. Meanwhile, it is urgent to identify mechanisms of immune resistance and design more potent immunotherapies, alone and/or with best combination partners to further prolong anti-MM immunity. Abstract The incorporation of novel agents in recent treatments in multiple myeloma (MM) has improved the clinical outcome of patients. Specifically, the approval of monoclonal antibody (MoAb) against CD38 (daratumumab) and SLAMF7 (elotuzumab) in relapsed and refractory MM (RRMM) represents an important milestone in the development of targeted immunotherapy in MM. These MoAb-based agents significantly induce cytotoxicity of MM cells via multiple effector-dependent mechanisms and can further induce immunomodulation to repair a dysfunctional tumor immune microenvironment. Recently, targeting B cell maturation antigen (BCMA), an even MM-specific antigen, has shown high therapeutic activities by chimeric antigen receptor T cells (CAR T), antibody-drug conjugate (ADC), bispecific T-cell engager (BiTE), as well as bispecific antibody (BiAb), with some already approved for heavily pretreated RRMM patients. New antigens, such as orphan G protein-coupled receptor class C group 5 member D (GPRC5D) and FcRH5, were identified and rapidly moved to ongoing clinical studies. We here summarized the pathobiological function of key MM antigens and the status of the corresponding immunotherapies. The potential challenges and emerging treatment strategies are also discussed.
Collapse
Affiliation(s)
- Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Division of Hematology & Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Lijie Xing
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China;
| | - Kenneth C. Anderson
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, 450 Brookline Avenue, Boston, MA 02215, USA; (S.-F.C.); (K.C.A.)
- Correspondence: ; Tel.: +1-617-632-3875; Fax: +1-617-632-2140
| |
Collapse
|
22
|
Marques C, Reis CA, Vivès RR, Magalhães A. Heparan Sulfate Biosynthesis and Sulfation Profiles as Modulators of Cancer Signalling and Progression. Front Oncol 2021; 11:778752. [PMID: 34858858 PMCID: PMC8632541 DOI: 10.3389/fonc.2021.778752] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 10/15/2021] [Indexed: 12/17/2022] Open
Abstract
Heparan Sulfate Proteoglycans (HSPGs) are important cell surface and Extracellular Matrix (ECM) maestros involved in the orchestration of multiple cellular events in physiology and pathology. These glycoconjugates bind to various bioactive proteins via their Heparan Sulfate (HS) chains, but also through the protein backbone, and function as scaffolds for protein-protein interactions, modulating extracellular ligand gradients, cell signalling networks and cell-cell/cell-ECM interactions. The structural features of HS chains, including length and sulfation patterns, are crucial for the biological roles displayed by HSPGs, as these features determine HS chains binding affinities and selectivity. The large HS structural diversity results from a tightly controlled biosynthetic pathway that is differently regulated in different organs, stages of development and pathologies, including cancer. This review addresses the regulatory mechanisms underlying HS biosynthesis, with a particular focus on the catalytic activity of the enzymes responsible for HS glycan sequences and sulfation motifs, namely D-Glucuronyl C5-Epimerase, N- and O-Sulfotransferases. Moreover, we provide insights on the impact of different HS structural epitopes over HSPG-protein interactions and cell signalling, as well as on the effects of deregulated expression of HS modifying enzymes in the development and progression of cancer. Finally, we discuss the clinical potential of HS biosynthetic enzymes as novel targets for therapy, and highlight the importance of developing new HS-based tools for better patients' stratification and cancer treatment.
Collapse
Affiliation(s)
- Catarina Marques
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal.,Programa Doutoral em Biologia Molecular e Celular (MCbiology), Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Celso A Reis
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal.,Faculdade de Medicina da Universidade do Porto (FMUP), Porto, Portugal
| | | | - Ana Magalhães
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal.,Instituto de Patologia e Imunologia Molecular da Universidade do Porto (IPATIMUP), Porto, Portugal.,Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
23
|
Junctional Adhesion Molecule-C expression specifies a CD138low/neg multiple myeloma cell population in mice and humans. Blood Adv 2021; 6:2195-2206. [PMID: 34861679 PMCID: PMC9006287 DOI: 10.1182/bloodadvances.2021004354] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 10/04/2021] [Indexed: 11/20/2022] Open
Abstract
JAM-C identifies a distinct MM cell population in bone marrow of patients and mice. Targeting JAM-C ameliorates MM progression and offers potential therapeutic options that might complement standard treatment regimens.
Deregulation such as overexpression of adhesion molecules influences cancer progression and survival. Metastasis of malignant cells from their primary tumor site to distant organs is the most common reason for cancer-related deaths. Junctional adhesion molecule-C (JAM-C), a member of the immunoglobulin-like JAM family, can homodimerize and aid cancer cell migration and metastasis. Here we show that this molecule is dynamically expressed on multiple myeloma (MM) cells in the bone marrow and co-localizes with blood vessels within the bone marrow of patients and mice. In addition, upregulation of JAM-C inversely correlates with the downregulation of the canonical plasma cell marker CD138 (syndecan-1), whose surface expression has recently been found to dynamically regulate a switch between MM growth in situ and MM dissemination. Moreover, targeting JAM-C in a syngeneic in vivo MM model ameliorates MM progression and improves outcome. Overall, our data demonstrate that JAM-C might serve not only as an additional novel diagnostic biomarker but also as a therapeutic target in MM disease.
Collapse
|
24
|
Wei W, Zhang Y, Zhang D, Liu Q, An S, Chen Y, Huang G, Liu J. Annotating BCMA Expression in Multiple Myelomas. Mol Pharm 2021; 19:3492-3501. [PMID: 34843261 DOI: 10.1021/acs.molpharmaceut.1c00628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
B cell maturation antigen (BCMA) is a promising theranostic target for multiple myeloma (MM). BCMA-targeted therapeutics, such as antibody-drug conjugates and chimeric antigen receptor T-cell immunotherapies, are rapidly reshaping the treatment landscape of MM. Along with the progress, a critical challenge is to noninvasively visualize the dynamic change of BCMA for a better-personalized prescription of the above-mentioned therapeutics. We aim to develop immuno-positron emission tomography (immunoPET) imaging strategies to visualize BCMA expression and realize target-specific diagnosis of MM in the work. A series of BCMA-targeting nanobodies were produced and two of them were successfully labeled with gallium-68 (68Ga). MM models were established using MM.1S cell line and NOD-Prkdcem26Cd52Il2rgem26Cd22/Nju mice. The diagnostic efficacies of the developed probes (i.e., [68Ga]Ga-NOTA-MMBC2 and [68Ga]Ga-NOTA-MMBC3) were investigated in disseminated MM models by immunoPET imaging, region of interest analysis on PET images, biodistribution study, and histopathological staining study. [68Ga]Ga-NOTA-MMBC2 and [68Ga]Ga-NOTA-MMBC3 were developed with radiochemical purities of >99%. ImmunoPET imaging with either [68Ga]Ga-NOTA-MMBC2 or [68Ga]Ga-NOTA-MMBC3 precisely visualized BCMA expression and delineated MM lesions throughout the bone marrows. Moreover, [68Ga]Ga-NOTA-MMBC3 immunoPET successfully detected remnant MM after treatment with daratumumab, a prescription medicine used to treat MM. The immunoPET imaging data correlated well with the biodistribution and immunohistochemistry staining results. The work successfully developed two state-of-the-art BCMA-targeted radiotracers for annotating BCMA expression and diagnosing MM. Translational studies interpreting the diagnostic efficacies of the immunoPET radiotracers are warranted.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - You Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Di Zhang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Qiufang Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Shuxian An
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yumei Chen
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.,Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Jianjun Liu
- Department of Nuclear Medicine, Institute of Clinical Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| |
Collapse
|
25
|
Fuertes MB, Domaica CI, Zwirner NW. Leveraging NKG2D Ligands in Immuno-Oncology. Front Immunol 2021; 12:713158. [PMID: 34394116 PMCID: PMC8358801 DOI: 10.3389/fimmu.2021.713158] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/02/2021] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) revolutionized the field of immuno-oncology and opened new avenues towards the development of novel assets to achieve durable immune control of cancer. Yet, the presence of tumor immune evasion mechanisms represents a challenge for the development of efficient treatment options. Therefore, combination therapies are taking the center of the stage in immuno-oncology. Such combination therapies should boost anti-tumor immune responses and/or target tumor immune escape mechanisms, especially those created by major players in the tumor microenvironment (TME) such as tumor-associated macrophages (TAM). Natural killer (NK) cells were recently positioned at the forefront of many immunotherapy strategies, and several new approaches are being designed to fully exploit NK cell antitumor potential. One of the most relevant NK cell-activating receptors is NKG2D, a receptor that recognizes 8 different NKG2D ligands (NKG2DL), including MICA and MICB. MICA and MICB are poorly expressed on normal cells but become upregulated on the surface of damaged, transformed or infected cells as a result of post-transcriptional or post-translational mechanisms and intracellular pathways. Their engagement of NKG2D triggers NK cell effector functions. Also, MICA/B are polymorphic and such polymorphism affects functional responses through regulation of their cell-surface expression, intracellular trafficking, shedding of soluble immunosuppressive isoforms, or the affinity of NKG2D interaction. Although immunotherapeutic approaches that target the NKG2D-NKG2DL axis are under investigation, several tumor immune escape mechanisms account for reduced cell surface expression of NKG2DL and contribute to tumor immune escape. Also, NKG2DL polymorphism determines functional NKG2D-dependent responses, thus representing an additional challenge for leveraging NKG2DL in immuno-oncology. In this review, we discuss strategies to boost MICA/B expression and/or inhibit their shedding and propose that combination strategies that target MICA/B with antibodies and strategies aimed at promoting their upregulation on tumor cells or at reprograming TAM into pro-inflammatory macrophages and remodeling of the TME, emerge as frontrunners in immuno-oncology because they may unleash the antitumor effector functions of NK cells and cytotoxic CD8 T cells (CTL). Pursuing several of these pipelines might lead to innovative modalities of immunotherapy for the treatment of a wide range of cancer patients.
Collapse
Affiliation(s)
- Mercedes Beatriz Fuertes
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Carolina Inés Domaica
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina
| | - Norberto Walter Zwirner
- Laboratorio de Fisiopatología de la Inmunidad Innata, Instituto de Biología y Medicina Experimental (IBYME-CONICET), Buenos Aires, Argentina.,Facultad de Ciencias Exactas y Naturales, Departamento de Química Biológica, Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
26
|
Xing L, Wang S, Liu J, Yu T, Chen H, Wen K, Li Y, Lin L, Hsieh PA, Cho SF, An G, Qiu L, Kinneer K, Munshi N, Anderson KC, Tai YT. BCMA-Specific ADC MEDI2228 and Daratumumab Induce Synergistic Myeloma Cytotoxicity via IFN-Driven Immune Responses and Enhanced CD38 Expression. Clin Cancer Res 2021; 27:5376-5388. [PMID: 34301753 DOI: 10.1158/1078-0432.ccr-21-1621] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/11/2021] [Accepted: 07/22/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Efforts are required to improve the potency and durability of CD38- and BCMA-based immunotherapies in human multiple myeloma. We here delineated the molecular and cellular mechanisms underlying novel immunomodulatory effects triggered by BCMA pyrrolobenzodiazepine (PBD) antibody drug conjugate (ADC) MEDI2228 which can augment efficacy of these immunotherapies. EXPERIMENTAL DESIGN MEDI2228-induced transcriptional and protein changes were investigated to define significantly impacted genes and signaling cascades in multiple myeloma cells. Mechanisms whereby MEDI2228 combination therapies can enhance cytotoxicity or overcome drug resistance in multiple myeloma cell lines and patient multiple myeloma cells were defined using in vitro models of tumor in the bone marrow (BM) microenvironment, as well as in human natural killer (NK)-reconstituted NOD/SCID gamma (NSG) mice bearing MM1S tumors. RESULTS MEDI2228 enriched IFN I signaling and enhanced expression of IFN-stimulated genes in multiple myeloma cell lines following the induction of DNA damage-ATM/ATR-CHK1/2 pathways. It activated cGAS-STING-TBK1-IRF3 and STAT1-IRF1-signaling cascades and increased CD38 expression in multiple myeloma cells but did not increase CD38 expression in BCMA-negative NK effector cells. It overcame CD38 downregulation on multiple myeloma cells triggered by IL6 and patient BM stromal cell-culture supernatant via activation of STAT1-IRF1, even in immunomodulatory drug (IMiD)- and bortezomib-resistant multiple myeloma cells. In vitro and in vivo upregulation of NKG2D ligands and CD38 in MEDI2228-treated multiple myeloma cells was further associated with synergistic daratumumab (Dara) CD38 MoAb-triggered NK-mediated cytotoxicity of both cell lines and autologous drug-resistant patient multiple myeloma cells. CONCLUSIONS These results provide the basis for clinical evaluation of combination MEDI2228 with Dara to further improve patient outcome in multiple myeloma.
Collapse
Affiliation(s)
- Lijie Xing
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, China
| | - Su Wang
- Department of Biomedical Informatics, Blavatnik Institute, Harvard Medical School, Boston, Massachusetts
| | - Jiye Liu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Tengteng Yu
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Hailin Chen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Kenneth Wen
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Yuyin Li
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
- School of Food Engineering and Biotechnology, Tianjin University of Science and Technology, Tianjin, China
- Key Lab of Industrial Fermentation Microbiology of the Ministry of Education
- State Key Laboratory of Food Nutrition and Safety, Tianjin University of Science and Technology, Tianjin China
| | - Liang Lin
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Phillip A Hsieh
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Shih-Feng Cho
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Gang An
- Division of Hematology-Oncology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Lugui Qiu
- State Key Laboratory of Experimental Hematology, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | | | - Nikhil Munshi
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| | - Yu-Tzu Tai
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston Massachusetts
| |
Collapse
|
27
|
B cells as target for immunotherapy in rheumatic diseases - current status. Immunol Lett 2021; 236:12-19. [PMID: 34077805 DOI: 10.1016/j.imlet.2021.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/16/2021] [Accepted: 05/25/2021] [Indexed: 01/16/2023]
Abstract
This mini-review is a short overview of different therapeutical strategies targeting B cells in systemic autoimmune rheumatic diseases, mainly: rheumatoid arthritis (RA), systemic lupus erythematosus (SLE) and primary Sjogren Syndrome (pSS). Many strategies and their rationale are discussed in this review: B cells' depletion (anti-CD20, anti-CD22), long-lived plasma cells depletion (anti-CD19, anti-CD27, anti-CD38 and anti-CD138), changing activation of B cells (anti-BAFF) and inhibiting proteasomes in plasma cells (bortezomib). The past successful therapies and less successful are shown, and the possible reasons for failures are discussed.
Collapse
|
28
|
Syndecan-1 (CD138), Carcinomas and EMT. Int J Mol Sci 2021; 22:ijms22084227. [PMID: 33921767 PMCID: PMC8072910 DOI: 10.3390/ijms22084227] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Cell surface proteoglycans are known to be important regulators of many aspects of cell behavior. The principal family of transmembrane proteoglycans is the syndecans, of which there are four in mammals. Syndecan-1 is mostly restricted to epithelia, and bears heparan sulfate chains that are capable of interacting with a large array of polypeptides, including extracellular matrix components and potent mediators of proliferation, adhesion and migration. For this reason, it has been studied extensively with respect to carcinomas and tumor progression. Frequently, but not always, syndecan-1 levels decrease as tumor grade, stage and invasiveness and dedifferentiation increase. This parallels experiments that show depletion of syndecan-1 can be accompanied by loss of cadherin-mediated adhesion. However, in some tumors, levels of syndecan-1 increase, but the characterization of its distribution is relevant. There can be loss of membrane staining, but acquisition of cytoplasmic and/or nuclear staining that is abnormal. Moreover, the appearance of syndecan-1 in the tumor stroma, either associated with its cellular component or the collagenous matrix, is nearly always a sign of poor prognosis. Given its relevance to myeloma progression, syndecan-1-directed antibody—toxin conjugates are being tested in clinical and preclinical trials, and may have future relevance to some carcinomas.
Collapse
|
29
|
Sperling AS, Anderson KC. Facts and Hopes in Multiple Myeloma Immunotherapy. Clin Cancer Res 2021; 27:4468-4477. [PMID: 33771856 DOI: 10.1158/1078-0432.ccr-20-3600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/09/2021] [Accepted: 03/11/2021] [Indexed: 12/30/2022]
Abstract
Among the hallmarks of cancer is the ability of neoplastic cells to evade and suppress immune surveillance to allow their growth and evolution. Nowhere is this as apparent as in multiple myeloma, a cancer of antibody-producing plasma cells, where a complex interplay between neoplastic cells and the immune microenvironment is required for the development and progression of disease. Decades of research has led to the discovery of a number of therapeutic agents, from cytotoxic drugs to genetically engineered cells that mediate their antimyeloma effects at least partially through altering these immune interactions. In this review, we discuss the history of immunotherapy and current practices in multiple myeloma, as well as the advances that promise to one day offer a cure for this deadly disease.
Collapse
Affiliation(s)
- Adam S Sperling
- Division of Hematology, Brigham and Women's Hospital, Boston, Massachusetts. .,Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Kenneth C Anderson
- Jerome Lipper Multiple Myeloma Center, LeBow Institute for Myeloma Therapeutics, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|