1
|
Horenstein AL, Faini AC, Morandi F, Ortolan E, Storti P, Giuliani N, Richardson PG, Malavasi F. Monoclonal anti-CD38 therapy in human myeloma: retrospects and prospects. Front Immunol 2025; 16:1519300. [PMID: 40013150 PMCID: PMC11860881 DOI: 10.3389/fimmu.2025.1519300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025] Open
Abstract
Monoclonal antibody therapy using CD38 as a target remains central to managing human multiple myeloma (MM). CD38 was selected early on as a target for mAb-mediated therapy for MM, driven by findings from an early Cluster of Differentiation (CD) Workshop. The first CD38-targeting antibody to be approved yielded strong trial results, significantly improving survival rates and earning widespread patient acceptance. However, resistance to the therapy later emerged, complicating treatment management. Despite CD38's still central role in MM therapy, too little attention has been paid to its broader roles-not only as a myeloma marker but also as an enzyme and adhesion molecule in physiology. This review, a collaborative effort between basic scientists and clinical experts, explores some of the lesser-known mechanisms of antibody action and interactions with CD38 at key stages of treatment. The review also highlights the relevance of the MM environment, focusing on the importance of the bone marrow (BM) niche. The goal is to identify new agents whose unique properties may enhance tumor eradication. By gaining a deeper understanding of interactions between therapeutic antibodies, myeloma cells, and the tumor microenvironment (TME), it is hoped that previously unrecognized vulnerabilities within the disease may be revealed, paving the way to more effective treatment strategies.
Collapse
Affiliation(s)
- Alberto L. Horenstein
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Angelo C. Faini
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
- Immunogenetics and Transplant Biology, University Hospital “Città della Salute e della Scienza di Torino”, Torino, Italy
| | - Fabio Morandi
- UOSD Laboratorio di Terapie Cellulari, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Erika Ortolan
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
| | - Paola Storti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Nicola Giuliani
- Department of Medicine and Surgery, University of Parma, Parma, Italy
- Department of Medicine and Surgery, University of Parma & Multiple Myeloma Program, AOU, Parma, Italy
| | - Paul G. Richardson
- Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Fabio Malavasi
- Lab of Immunogenetics, Department of Medical Sciences, University of Torino, Torino, Italy
- Fondazione Ricerca Molinette Ets, Torino, Italy
| |
Collapse
|
2
|
Chen W, Wu X, Hu J, Liu X, Guo Z, Wu J, Shao Y, Hao M, Zhang S, Hu W, Wang Y, Zhang M, Zhu M, Wang C, Wu Y, Wang J, Xing D. The translational potential of miR-26 in atherosclerosis and development of agents for its target genes ACC1/2, COL1A1, CPT1A, FBP1, DGAT2, and SMAD7. Cardiovasc Diabetol 2024; 23:21. [PMID: 38195542 PMCID: PMC10777520 DOI: 10.1186/s12933-024-02119-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/02/2024] [Indexed: 01/11/2024] Open
Abstract
Atherosclerosis is one of the leading causes of death worldwide. miR-26 is a potential biomarker of atherosclerosis. Standardized diagnostic tests for miR-26 (MIR26-DX) have been developed, but the fastest progress has been in predicting the efficacy of IFN-α therapy for hepatocellular carcinoma (HCC, phase 3). MiR-26 slows atherosclerosis development by suppressing ACC1/2, ACLY, ACSL3/4, ALDH3A2, ALPL, BMP2, CD36, COL1A1, CPT1A, CTGF, DGAT2, EHHADH, FAS, FBP1, GATA4, GSK3β, G6PC, Gys2, HMGA1, HMGB1, LDLR, LIPC, IL-1β, IL-6, JAG2, KCNJ2, MALT1, β-MHC, NF-κB, PCK1, PLCβ1, PYGL, RUNX2, SCD1, SMAD1/4/5/7, SREBF1, TAB3, TAK1, TCF7L2, and TNF-α expression. Many agents targeting these genes, such as the ACC1/2 inhibitors GS-0976, PF-05221304, and MK-4074; the DGAT2 inhibitors IONIS-DGAT2Rx, PF-06427878, PF-0685571, and PF-07202954; the COL1A1 inhibitor HT-100; the stimulants 68Ga-CBP8 and RCT-01; the CPT1A inhibitors etomoxir, perhexiline, and teglicar; the FBP1 inhibitors CS-917 and MB07803; and the SMAD7 inhibitor mongersen, have been investigated in clinical trials. Interestingly, miR-26 better reduced intima-media thickness (IMT) than PCSK9 or CT-1 knockout. Many PCSK9 inhibitors, including alirocumab, evolocumab, inclisiran, AZD8233, Civi-007, MK-0616, and LIB003, have been investigated in clinical trials. Recombinant CT-1 was also investigated in clinical trials. Therefore, miR-26 is a promising target for agent development. miR-26 promotes foam cell formation by reducing ABCA1 and ARL4C expression. Multiple materials can be used to deliver miR-26, but it is unclear which material is most suitable for mass production and clinical applications. This review focuses on the potential use of miR-26 in treating atherosclerosis to support the development of agents targeting it.
Collapse
Affiliation(s)
- Wujun Chen
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Xiaolin Wu
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Jianxia Hu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xiaolei Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Zhu Guo
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Jianfeng Wu
- Department of Cardiology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Key Laboratory of Heart Failure Prevention & Treatment of Hengyang, Clinical Medicine Research Center of Arteriosclerotic Disease of Hunan Province, Hengyang, 421001, Hunan, China
| | - Yingchun Shao
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Minglu Hao
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Shuangshuang Zhang
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Weichao Hu
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
- Department of Endocrinology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266000, Shandong, China
| | - Yanhong Wang
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Miao Zhang
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
| | - Meng Zhu
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, 266071, Shandong, China
| | - Chao Wang
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Yudong Wu
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Jie Wang
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
| | - Dongming Xing
- Cancer Institute, Department of Orthopaedics, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, 266071, Shandong, China.
- School of Life Sciences, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
3
|
Li X, Wang Y, Yang Q, Song L, Kang L, Hu Z, Wang Z. Microarray-Based CD38 Peptide Probe Screening for Multiple Myeloma Imaging. Mol Pharm 2024; 21:245-254. [PMID: 38096423 DOI: 10.1021/acs.molpharmaceut.3c00808] [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] [Indexed: 01/02/2024]
Abstract
Assessing CD38 expression in vivo has become a significant element in multiple myeloma (MM) therapy, as it can be used to detect lesions and forecast the effectiveness of treatment. Accurate diagnosis requires a multifunctional, high-throughput probe screening platform to develop molecular probes for tumor-targeted multimodal imaging and treatment. Here, we investigated a microarray chip-based strategy for high-throughput screening of peptide probes for CD38. We obtained two new target peptides, CA-1 and CA-2, from a 105 peptide library with a dissociation constant (KD) of 10-7 M. The specificity and affinity of the target peptides were confirmed at the molecular and cellular levels. Peptide probes were labeled with indocyanine green (ICG) dye and 68Ga-DOTA, which were injected into a CD38-positive Ramos tumor-bearing mouse via its tail vein, and small animal fluorescence and positron emission tomography (PET) imaging showed that the peptide probes could show specific enrichment in the tumor tissue. Our study shows that a microchip-based screening of peptide probes can be used as a promising imaging tool for MM diagnosis.
Collapse
Affiliation(s)
- Xuejie Li
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Yuanzhuo Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
| | - Qi Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Lele Song
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Zhiyuan Hu
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing 100190, China
- School of Nanoscience and Technology, Sino-Danish College, University of Chinese Academy of Sciences, Beijing 100049, China
- School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan 430205, China
| | - Zihua Wang
- Fujian Provincial Key Laboratory of Brain Aging and Neurodegenerative Diseases, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian 350122, China
| |
Collapse
|
4
|
Zhao A, Zhou H, Yang J, Li M, Niu T. Epigenetic regulation in hematopoiesis and its implications in the targeted therapy of hematologic malignancies. Signal Transduct Target Ther 2023; 8:71. [PMID: 36797244 PMCID: PMC9935927 DOI: 10.1038/s41392-023-01342-6] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 01/03/2023] [Accepted: 01/19/2023] [Indexed: 02/18/2023] Open
Abstract
Hematologic malignancies are one of the most common cancers, and the incidence has been rising in recent decades. The clinical and molecular features of hematologic malignancies are highly heterogenous, and some hematologic malignancies are incurable, challenging the treatment, and prognosis of the patients. However, hematopoiesis and oncogenesis of hematologic malignancies are profoundly affected by epigenetic regulation. Studies have found that methylation-related mutations, abnormal methylation profiles of DNA, and abnormal histone deacetylase expression are recurrent in leukemia and lymphoma. Furthermore, the hypomethylating agents and histone deacetylase inhibitors are effective to treat acute myeloid leukemia and T-cell lymphomas, indicating that epigenetic regulation is indispensable to hematologic oncogenesis. Epigenetic regulation mainly includes DNA modifications, histone modifications, and noncoding RNA-mediated targeting, and regulates various DNA-based processes. This review presents the role of writers, readers, and erasers of DNA methylation and histone methylation, and acetylation in hematologic malignancies. In addition, this review provides the influence of microRNAs and long noncoding RNAs on hematologic malignancies. Furthermore, the implication of epigenetic regulation in targeted treatment is discussed. This review comprehensively presents the change and function of each epigenetic regulator in normal and oncogenic hematopoiesis and provides innovative epigenetic-targeted treatment in clinical practice.
Collapse
Affiliation(s)
- Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Hui Zhou
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Jinrong Yang
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Meng Li
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, 610041, Chengdu, Sichuan, China.
| |
Collapse
|
5
|
De Luca F, Allegra A, Di Chio C, Previti S, Zappalà M, Ettari R. Monoclonal Antibodies: The Greatest Resource to Treat Multiple Myeloma. Int J Mol Sci 2023; 24:ijms24043136. [PMID: 36834545 PMCID: PMC9959320 DOI: 10.3390/ijms24043136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/09/2023] Open
Abstract
Multiple myeloma (MM) is a currently incurable hematologic cancer. This disease is characterized by immunological alterations of myeloid cells and lymphocytes. The first-line therapy involves the use of classic chemotherapy; however, many patients have a relapsed form that could evolve into a refractory MM. The new therapeutic frontiers involve the use of new monoclonal antibodies (Mab) such as daratumumab, isatuximab, and elotuzumab. In addition to monoclonal antibodies, new immunotherapies based on modern bispecific antibodies and chimeric antigen receptor (CAR) T cell therapy have been investigated. For this reason, immunotherapy represents the greatest hope for the treatment of MM. This review intends to focus the attention on the new approved antibody targets. The most important are: CD38 (daratumumab and isatuximab), SLAM7 (elotuzumab), and BCMA (belantamab mafodotin) for the treatment of MM currently used in clinical practice. Although the disease is still incurable, the future perspective is to find the best therapeutic combination among all available drugs.
Collapse
Affiliation(s)
- Fabiola De Luca
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Alessandro Allegra
- Department of Human Pathology in Adulthood and Childhood, University of Messina, Via Consolare Valeria, 90100 Messina, Italy
| | - Carla Di Chio
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Santo Previti
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Maria Zappalà
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy
- Correspondence: ; Tel.: +39-090-6766554
| |
Collapse
|
6
|
De Novellis D, Fontana R, Giudice V, Serio B, Selleri C. Innovative Anti-CD38 and Anti-BCMA Targeted Therapies in Multiple Myeloma: Mechanisms of Action and Resistance. Int J Mol Sci 2022; 24:645. [PMID: 36614086 PMCID: PMC9820921 DOI: 10.3390/ijms24010645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
CD38 and B-cell maturation antigens (BCMAs) are prevalently expressed on neoplastic plasma cells in multiple myeloma (MM), making them ideal therapeutic targets. Anti-CD38 monoclonal antibodies, such as approved daratumumab and isatuximab, are currently the milestone in MM treatment because they induce plasma cell apoptosis and kill through several mechanisms, including antibody-dependent cellular cytotoxicity or phagocytosis. BCMA is considered an excellent target in MM, and three different therapeutic strategies are either already available in clinical practice or under investigation: antibody-drug conjugates, such as belantamab-mafodotin; bispecific T cell engagers; and chimeric antigen receptor-modified T cell therapies. Despite the impressive clinical efficacy of these new strategies in the treatment of newly diagnosed or multi-refractory MM patients, several mechanisms of resistance have already been described, including antigen downregulation, the impairment of antibody-dependent cell cytotoxicity and phagocytosis, T- and natural killer cell senescence, and exhaustion. In this review, we summarize the current knowledge on the mechanisms of action and resistance of anti-CD38 and anti-BCMA agents and their clinical efficacy and safety.
Collapse
Affiliation(s)
- Danilo De Novellis
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Raffaele Fontana
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Bianca Serio
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081 Baronissi, Italy
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| |
Collapse
|
7
|
Yang Y, Wu Z, Wang M, Wu Z, Sun Z, Liu M, Li G. MicroRNA-429 Regulates Invasion and Migration of Multiple Myeloma Cells via Bmi1/AKT Pathway. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: miR-429-mediated progression of multiple myeloma (MM) was studied through mediating B cell-specific Moloney murine leukemia virus integration site 1 (Bmi1)/protein kinase B (AKT) pathway. Methods: miRNA or siRNA was delivered into MM cell lines to alter cellular
proliferation, apoptosis, invasion and migration. Measurements of miR-429 and Bmi1 levels were performed. AKT and p-AKT expression change was measured after regulating miR-429. The interaction between miR-429 and Bmi1 was analyzed. Results: miR-429 elevation disrupted proliferation,
anti-apoptosis, migration and invasion properties of MM cells, and inactivated AKT pathway. Bmi1 was a targeting partner of miR-429, which was highly expressed in MM. Bmi1 knockdown phenotyped the effects of overexpressed miR-429 on MM cells. AKT agonist SC70 reversed miR-429-regulated inhibition
of MM cell growth. Conclusion: miR-429 suppresses the activation of Bmi1/AKT pathway to down-regulate the malignant functions of MM cells.
Collapse
Affiliation(s)
- YongMing Yang
- Department of Orthopedics, The First Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| | - ZhiFeng Wu
- Department of Orthopedics, The First Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| | - Ming Wang
- Department of Orthopedics, The First Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| | - ZuTong Wu
- Department of Orthopedics, The First Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| | - ZhenZheng Sun
- Department of Pediatrics, The First Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| | - Man Liu
- Department of Operating Room, The first Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| | - GuangBao Li
- Department of Orthopedics, The First Affiliated Hospital of Shaoyang College, ShaoYang City, HuNan Province, 422000, China
| |
Collapse
|
8
|
Vlachová M, Gregorová J, Vychytilová-Faltejsková P, Gabło NA, Radová L, Pospíšilová L, Almáši M, Štork M, Knechtová Z, Minařík J, Popková T, Jelínek T, Hájek R, Pour L, Říhová L, Ševčíková S. Involvement of Small Non-Coding RNA and Cell Antigens in Pathogenesis of Extramedullary Multiple Myeloma. Int J Mol Sci 2022; 23:ijms232314765. [PMID: 36499093 PMCID: PMC9741227 DOI: 10.3390/ijms232314765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 11/21/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Extramedullary multiple myeloma (EMD) is an aggressive disease; malignant plasma cells lose their dependence in the bone marrow microenvironment and migrate into tissues. EMD is a negative prognostic factor of survival. Using flow cytometry and next-generation sequencing, we aimed to identify antigens and microRNAs (miRNAs) involved in EMD pathogenesis. Flow cytometry analysis revealed significant differences in the level of clonal plasma cells between MM and EMD patients, while the expression of CD markers was comparable between these two groups. Further, miR-26a-5p and miR-30e-5p were found to be significantly down-regulated in EMD compared to MM. Based on the expression of miR-26a-5p, we were able to distinguish these two groups of patients with high sensitivity and specificity. In addition, the involvement of deregulated miRNAs in cell cycle regulation, ubiquitin-mediated proteolysis and signaling pathways associated with infections or neurological disorders was observed using GO and KEGG pathways enrichment analysis. Subsequently, a correlation between the expression of analyzed miRNAs and the levels of CD molecules was observed. Finally, clinicopathological characteristics as well as CD antigens associated with the prognosis of MM and EMD patients were identified. Altogether, we identified several molecules possibly involved in the transformation of MM into EMD.
Collapse
Affiliation(s)
- Monika Vlachová
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Jana Gregorová
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Petra Vychytilová-Faltejsková
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
- Centre for Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Natalia Anna Gabło
- Centre for Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Lenka Radová
- Centre for Molecular Medicine, Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Lenka Pospíšilová
- Institute of Biostatistics and Analyses, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Martina Almáši
- Department of Clinical Hematology, University Hospital Brno, 625 00 Brno, Czech Republic
| | - Martin Štork
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 625 00 Brno, Czech Republic
| | - Zdeňka Knechtová
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 625 00 Brno, Czech Republic
| | - Jiří Minařík
- Department of Hemato-Oncology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, 779 00 Olomouc, Czech Republic
| | - Tereza Popková
- Department of Hematooncology, University Hospital Ostrava, Ostrava, and Faculty of Medicine, University Ostrava, 708 00 Ostrava, Czech Republic
| | - Tomáš Jelínek
- Department of Hematooncology, University Hospital Ostrava, Ostrava, and Faculty of Medicine, University Ostrava, 708 00 Ostrava, Czech Republic
| | - Roman Hájek
- Department of Hematooncology, University Hospital Ostrava, Ostrava, and Faculty of Medicine, University Ostrava, 708 00 Ostrava, Czech Republic
| | - Luděk Pour
- Department of Internal Medicine, Hematology and Oncology, University Hospital Brno, 625 00 Brno, Czech Republic
| | - Lucie Říhová
- Department of Clinical Hematology, University Hospital Brno, 625 00 Brno, Czech Republic
| | - Sabina Ševčíková
- Babak Myeloma Group, Department of Pathophysiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
- Correspondence: ; Tel.: +420-549-493-380
| |
Collapse
|
9
|
Molecular Determinants Underlying the Anti-Cancer Efficacy of CD38 Monoclonal Antibodies in Hematological Malignancies. Biomolecules 2022; 12:biom12091261. [PMID: 36139103 PMCID: PMC9496523 DOI: 10.3390/biom12091261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
CD38 was first discovered as a T-cell antigen and has since been found ubiquitously expressed in various hematopoietic cells, including plasma cells, NK cells, B cells, and granulocytes. More importantly, CD38 expression levels on malignant hematopoietic cells are significantly higher than counterpart healthy cells, thus presenting itself as a promising therapeutic target. In fact, for many aggressive hematological cancers, including CLL, DLBCL, T-ALL, and NKTL, CD38 expression is significantly associated with poorer prognosis and a hyperproliferative or metastatic phenotype. Studies have shown that, beyond being a biomarker, CD38 functionally mediates dysregulated survival, adhesion, and migration signaling pathways, as well as promotes an immunosuppressive microenvironment conducive for tumors to thrive. Thus, targeting CD38 is a rational approach to overcoming these malignancies. However, clinical trials have surprisingly shown that daratumumab monotherapy has not been very effective in these other blood malignancies. Furthermore, extensive use of daratumumab in MM is giving rise to a subset of patients now refractory to daratumumab treatment. Thus, it is important to consider factors modulating the determinants of response to CD38 targeting across different blood malignancies, encompassing both the transcriptional and post-transcriptional levels so that we can diversify the strategy to enhance daratumumab therapeutic efficacy, which can ultimately improve patient outcomes.
Collapse
|
10
|
Szudy-Szczyrek A, Ahern S, Krawczyk J, Szczyrek M, Hus M. MiRNA as a Potential Target for Multiple Myeloma Therapy–Current Knowledge and Perspectives. J Pers Med 2022; 12:jpm12091428. [PMID: 36143213 PMCID: PMC9503263 DOI: 10.3390/jpm12091428] [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: 07/22/2022] [Revised: 08/23/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Multiple myeloma (MM) is the second most common hematological malignancy. Despite the huge therapeutic progress thanks to the introduction of novel therapies, MM remains an incurable disease. Extensive research is currently ongoing to find new options. MicroRNAs (miRNAs) are small, non-coding RNA molecules that regulate gene expression at a post-transcriptional level. Aberrant expression of miRNAs in MM is common. Depending on their role in MM development, miRNAs have been reported as oncogenes and tumor suppressors. It was demonstrated that specific miRNA alterations using miRNA mimics or antagomirs can normalize the gene regulatory network and signaling pathways in the microenvironment and MM cells. These properties make miRNAs attractive targets in anti-myeloma therapy. However, only a few miRNA-based drugs have been entered into clinical trials. In this review, we discuss the role of the miRNAs in the pathogenesis of MM, their current status in preclinical/clinical trials, and the mechanisms by which miRNAs can theoretically achieve therapeutic benefit in MM treatment.
Collapse
Affiliation(s)
- Aneta Szudy-Szczyrek
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
- Correspondence: (A.S.-S.); (M.H.)
| | - Sean Ahern
- Department of Haematology, University Hospital Galway, H91 Galway, Ireland
- National University of Ireland, H91 Galway, Ireland
| | - Janusz Krawczyk
- Department of Haematology, University Hospital Galway, H91 Galway, Ireland
- National University of Ireland, H91 Galway, Ireland
| | - Michał Szczyrek
- Chair and Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-950 Lublin, Poland
| | - Marek Hus
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-081 Lublin, Poland
- Correspondence: (A.S.-S.); (M.H.)
| |
Collapse
|
11
|
Souza OF, Popi AF. Role of microRNAs in B-Cell Compartment: Development, Proliferation and Hematological Diseases. Biomedicines 2022; 10:biomedicines10082004. [PMID: 36009551 PMCID: PMC9405569 DOI: 10.3390/biomedicines10082004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/14/2022] [Indexed: 11/24/2022] Open
Abstract
B-cell development is a very orchestrated pathway that involves several molecules, such as transcription factors, cytokines, microRNAs, and also different cells. All these components maintain the ideal microenvironment and control B-cell differentiation. MicroRNAs are small non-coding RNAs that bind to target mRNA to control gene expression. These molecules could circulate in the body in a free form, protein-bounded, or encapsulated into extracellular vesicles, such as exosomes. The comprehension of the role of microRNAs in the B-cell development was possible based on microRNA profile of each B-cell stage and functional studies. Herein, we report the knowledge about microRNAs in the B-cell the differentiation, proliferation, and also in hematological malignancies.
Collapse
|
12
|
Li H, Da D, Yu W, Chen L, Yang S, Zhang B, Wang Y, Li L, Dang C. Tumor suppressor genes are reactivated by miR-26A1 via enhancer reprogramming in NSCLC. Hum Mol Genet 2022; 32:79-92. [PMID: 35921230 PMCID: PMC9838096 DOI: 10.1093/hmg/ddac185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/23/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most malignant epithelial tumors. Studies have suggested that DNA hypermethylation of promoters and abnormal histone modifications could induce tumor suppressor genes (TSGs) downregulation in NSCLC. However, the exact mechanism of TSGs downregulation remains unclear. In this study, we found that there is no difference in the regions of most TSGs promoters in NSCLC. Moreover, we found that there is no DNA methylation difference in the region of VILL promoter in NSCLC compared with adjacent tissue samples by pyrosequencing. We further demonstrated that VILL was markedly reactivated in A549 and H1703 cells infected with miR-26A1 lentivirus while this activation was inhibited by JQ1, an enhancer inhibitor. In addition, we identified that miR-26A1 could function as a tumor suppressor to inhibit proliferation and metastasis of NSCLC cells. Chromatin immunoprecipitation assays revealed that overexpression of miR-26A1 could significantly induce the enrichment of H3K27ac at the enhancer regions in A549 cells. To sum up, our findings revealed that enhancer-mediated TSGs regulation occured in NSCLC, suggesting that miR-26A1 could serve as a key regulator and may be a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Hongling Li
- To whom correspondence should be addressed at: Department of Oncology, Gansu Provincial Hospital, Lanzhou 730000, PR China. Tel: +86-0931-8281563;
| | | | | | - Lu Chen
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200000, PR China
| | - Shuai Yang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200000, PR China
| | - Baolong Zhang
- Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200000, PR China
| | - Yongying Wang
- Department of Oncology, Gansu Provincial Hospital, The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| | - Linyu Li
- Department of Oncology, Gansu Provincial Hospital, The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| | - Chunyan Dang
- Department of Oncology, Gansu Provincial Hospital, The First Clinical Medical College of Gansu University of Chinese Medicine, Lanzhou 730000, PR China
| |
Collapse
|
13
|
Ghosal S, Banerjee S. Investigating the potential molecular players and therapeutic drug molecules in carfilzomib resistant multiple myeloma by comprehensive bioinformatics analysis. Leuk Lymphoma 2022; 63:2545-2556. [DOI: 10.1080/10428194.2022.2087064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
- Somnath Ghosal
- School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| | - Subrata Banerjee
- School of Biological Sciences, Ramakrishna Mission Vivekananda Educational and Research Institute (RKMVERI), Kolkata, India
| |
Collapse
|
14
|
Shin HG, Yang HR, Yoon A, Lee S. Bispecific Antibody-Based Immune-Cell Engagers and Their Emerging Therapeutic Targets in Cancer Immunotherapy. Int J Mol Sci 2022; 23:5686. [PMID: 35628495 PMCID: PMC9146966 DOI: 10.3390/ijms23105686] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/16/2022] [Accepted: 05/17/2022] [Indexed: 12/16/2022] Open
Abstract
Cancer is the second leading cause of death worldwide after cardiovascular diseases. Harnessing the power of immune cells is a promising strategy to improve the antitumor effect of cancer immunotherapy. Recent progress in recombinant DNA technology and antibody engineering has ushered in a new era of bispecific antibody (bsAb)-based immune-cell engagers (ICEs), including T- and natural-killer-cell engagers. Since the first approval of blinatumomab by the United States Food and Drug Administration (US FDA), various bsAb-based ICEs have been developed for the effective treatment of patients with cancer. Simultaneously, several potential therapeutic targets of bsAb-based ICEs have been identified in various cancers. Therefore, this review focused on not only highlighting the action mechanism, design and structure, and status of bsAb-based ICEs in clinical development and their approval by the US FDA for human malignancy treatment, but also on summarizing the currently known and emerging therapeutic targets in cancer. This review provides insights into practical considerations for developing next-generation ICEs.
Collapse
Affiliation(s)
- Ha Gyeong Shin
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Ha Rim Yang
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
| | - Aerin Yoon
- R&D Division, GC Biopharma, Yongin 16924, Korea
| | - Sukmook Lee
- Department of Biopharmaceutical Chemistry, College of Science and Technology, Kookmin University, Seoul 02707, Korea; (H.G.S.); (H.R.Y.)
- Biopharmaceutical Chemistry Major, School of Applied Chemistry, Kookmin University, Seoul 02707, Korea
- Antibody Research Institute, Kookmin University, Seoul 02707, Korea
| |
Collapse
|
15
|
DiLillo AM, Chan KK, Sun XL, Ao G. Glycopolymer-Wrapped Carbon Nanotubes Show Distinct Interaction of Carbohydrates With Lectins. Front Chem 2022; 10:852988. [PMID: 35308788 PMCID: PMC8927622 DOI: 10.3389/fchem.2022.852988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022] Open
Abstract
Glyconanomaterials with unique nanoscale property and carbohydrate functionality show vast potential in biological and biomedical applications. We investigated the interactions of noncovalent complexes of single-wall carbon nanotubes that are wrapped by disaccharide lactose-containing glycopolymers with the specific carbohydrate-binding proteins. The terminal galactose (Gal) of glycopolymers binds to the specific lectin as expected. Interestingly, an increased aggregation of nanotubes was also observed when interacting with a glucose (Glc) specific lectin, likely due to the removal of Glc groups from the surface of nanotubes resulting from the potential binding of the lectin to the Glc in the glycopolymers. This result indicates that the wrapping conformation of glycopolymers on the surface of nanotubes potentially allows improved accessibility of the Glc for specific lectins. Furthermore, it shows that the interaction between Glc groups in the glycopolymers and nanotubes play a key role in stabilizing the nanocomplexes. Overall, our results demonstrate that nanostructures can enable conformation-dependent interactions of glycopolymers and proteins and can potentially lead to the creation of versatile optical sensors for detecting carbohydrate-protein interactions with enhanced specificity and sensitivity.
Collapse
Affiliation(s)
- Ana M. DiLillo
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, Cleveland, OH, United States
| | - Ka Keung Chan
- Department of Chemistry, Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH, United States
| | - Xue-Long Sun
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, Cleveland, OH, United States
- Department of Chemistry, Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, Cleveland, OH, United States
| | - Geyou Ao
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, Cleveland, OH, United States
| |
Collapse
|
16
|
Wu HT, Zhao XY. Regulation of CD38 on Multiple Myeloma and NK Cells by Monoclonal Antibodies. Int J Biol Sci 2022; 18:1974-1988. [PMID: 35342342 PMCID: PMC8935232 DOI: 10.7150/ijbs.68148] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Accepted: 01/19/2022] [Indexed: 11/14/2022] Open
Abstract
CD38 is highly expressed on multiple myeloma (MM) cells and plays a role in regulating tumor generation and development. CD38 monoclonal antibodies (mAbs) have been used as an effective therapy for MM treatment by various mechanisms, including complement-dependent cytotoxic effects, antibody-dependent cell-mediated cytotoxicity, antibody-dependent cellular phagocytosis, programmed cell death, enzymatic modulation, and immunomodulation. Although CD38 mAbs inhibit the proliferation and survival of MM cells, there are substantial side effects on antitumoral NK cells. The NK-mediated immune response needs to be further evaluated to minimize the adverse effects of NK cell loss. The killing effect of CD38 mAbs on CD38high NK cells should be minimized and the potential combination of CD38low/- NK cells and CD38 mAbs should be maximized to better benefit from their therapeutic efficacy against MM. CD38 mAb effects against MM can be maximized by combination therapies with immunomodulatory imide drugs (IMiDs), proteasome inhibitors (PIs), anti-programmed death 1 (PD-1)/programmed death ligand 1 (PD-L1) antibodies, or cellular therapies for the treatment of MM, especially in patients with relapsed or refractory MM (R/R MM) and drug-resistant MM.
Collapse
Affiliation(s)
- Hao-Tian Wu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
| | - Xiang-Yu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, National Clinical Research Center for Hematologic Disease, No. 11 South Street of Xizhimen, Xicheng District, Beijing 100044, China
- Collaborative Innovation Center of Hematology, China
| |
Collapse
|
17
|
Xue L, Jia T, Zhu Y, Zhao L, Mao J. Down-regulation of circ_0058058 suppresses proliferation, angiogenesis and metastasis in multiple myeloma through miR-338-3p/ATG14 pathway. J Orthop Surg Res 2021; 16:723. [PMID: 34930344 PMCID: PMC8686392 DOI: 10.1186/s13018-021-02867-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 12/05/2021] [Indexed: 12/12/2022] Open
Abstract
Background Multiple myeloma (MM) is one of the most frequently diagnosed hematological malignancy. Dysregulation of circular RNAs (circRNAs) has important impacts on MM process. Herein, this work aimed to investigate the role and mechanism of circ_0058058 in MM progression. Methods Levels of genes and proteins were detected by real-time reverse transcription PCR (RT-qPCR) and Western blot. CCK-8 assay, colony formation assay, EdU assay, flow cytometry, tube formation assay, transwell assay and Western blot were utilized to detect the proliferation, apoptosis, angiogenesis and metastasis of MM cells. The target relationship between miR-338-3p and circ_0058058 or ATG14 (autophagy related 14) was verified by dual-luciferase reporter assay and RNA immunoprecipitation (RIP) assay. In vivo experiments were performed using Xenograft assay. Results Circ_0058058 was up-regulated in MM bone marrow aspirates and cells, knockdown of circ_0058058 reduced MM cell proliferation, angiogenesis and metastasis, but induced apoptosis in vitro. In a MM xenograft mouse model, circ_0058058 silencing reduced MM tumor growth and cell proliferation. Mechanistically, circ_0058058 acted as a sponge for miR-338-3p to up-regulate ATG14 expression, which was validated to be a target of miR-338-3p. Rescue assay showed that miR-338-3p inhibition reversed the antitumor effects of circ_0058058 knockdown on MM cell. Moreover, forced expression of miR-338-3p suppressed MM cell malignant phenotype, which was abolished by ATG14 up-regulation. Conclusion Circ_0058058 functions as a sponge for miR-338-3p to elevate ATG14 expression to promote MM cell proliferation, metastasis and angiogenesis, affording a potential therapeutic target for MM prevention.
Collapse
Affiliation(s)
- Lianguo Xue
- Department of Hematology, The First People's Hospital of Lianyungang, No. 182 Tongguan North Road, Haizhou District, Lianyungang City, 222002, Jiangsu Province, China
| | - Tao Jia
- Department of Hematology, The First People's Hospital of Lianyungang, No. 182 Tongguan North Road, Haizhou District, Lianyungang City, 222002, Jiangsu Province, China
| | - Yuanxin Zhu
- Department of Hematology, The First People's Hospital of Lianyungang, No. 182 Tongguan North Road, Haizhou District, Lianyungang City, 222002, Jiangsu Province, China
| | - Lidong Zhao
- Department of Hematology, The First People's Hospital of Lianyungang, No. 182 Tongguan North Road, Haizhou District, Lianyungang City, 222002, Jiangsu Province, China
| | - Jianping Mao
- Department of Hematology, The First People's Hospital of Lianyungang, No. 182 Tongguan North Road, Haizhou District, Lianyungang City, 222002, Jiangsu Province, China.
| |
Collapse
|
18
|
Kalushkova A, Nylund P, Párraga AA, Lennartsson A, Jernberg-Wiklund H. One Omics Approach Does Not Rule Them All: The Metabolome and the Epigenome Join Forces in Haematological Malignancies. EPIGENOMES 2021; 5:epigenomes5040022. [PMID: 34968247 PMCID: PMC8715477 DOI: 10.3390/epigenomes5040022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 02/01/2023] Open
Abstract
Aberrant DNA methylation, dysregulation of chromatin-modifying enzymes, and microRNAs (miRNAs) play a crucial role in haematological malignancies. These epimutations, with an impact on chromatin accessibility and transcriptional output, are often associated with genomic instability and the emergence of drug resistance, disease progression, and poor survival. In order to exert their functions, epigenetic enzymes utilize cellular metabolites as co-factors and are highly dependent on their availability. By affecting the expression of metabolic enzymes, epigenetic modifiers may aid the generation of metabolite signatures that could be utilized as targets and biomarkers in cancer. This interdependency remains often neglected and poorly represented in studies, despite well-established methods to study the cellular metabolome. This review critically summarizes the current knowledge in the field to provide an integral picture of the interplay between epigenomic alterations and the cellular metabolome in haematological malignancies. Our recent findings defining a distinct metabolic signature upon response to enhancer of zeste homolog 2 (EZH2) inhibition in multiple myeloma (MM) highlight how a shift of preferred metabolic pathways may potentiate novel treatments. The suggested link between the epigenome and the metabolome in haematopoietic tumours holds promise for the use of metabolic signatures as possible biomarkers of response to treatment.
Collapse
Affiliation(s)
- Antonia Kalushkova
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
- Correspondence:
| | - Patrick Nylund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Alba Atienza Párraga
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| | - Andreas Lennartsson
- Department of Biosciences and Nutrition, NEO, Karolinska Institutet, 14157 Huddinge, Sweden;
| | - Helena Jernberg-Wiklund
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, 75185 Uppsala, Sweden; (P.N.); (A.A.P.); (H.J.-W.)
| |
Collapse
|
19
|
Jia H, Liu Y, Lv S, Qiao R, Zhang X, Niu F, Shang W, Liu S, Dong J, Zhang Z. LBX2-AS1 as a Novel Diagnostic Biomarker and Therapeutic Target Facilitates Multiple Myeloma Progression by Enhancing mRNA Stability of LBX2. Front Mol Biosci 2021; 8:706570. [PMID: 34552959 PMCID: PMC8450339 DOI: 10.3389/fmolb.2021.706570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 08/04/2021] [Indexed: 02/02/2023] Open
Abstract
Objective: Multiple myeloma (MM) represents a common age-associated malignancy globally. The function and underlying mechanism of antisense lncRNA LBX2-AS1 remain ambiguous in multiple myeloma (MM). Herein, we aimed to observe the biological implication of this lncRNA in MM. Methods: RT-qPCR was employed to examine circulating LBX2-AS1 and LBX2 in 60 paired MM and healthy subjects. Correlation between the two was analyzed by Pearson test. Under transfection with shLBX2-AS1, proliferation and apoptosis were evaluated in MM cells through CCK-8, colony formation and flow cytometry. LBX2 expression was examined in MM cells with shLBX2-AS1 or pcDNA3.1-LBX2 transfection. Following treatment with cycloheximide or actinomycin D, LBX2 expression was examined in pcDNA3.1-LBX2-transfected MM cells at different time points. Rescue assays were then presented. Finally, xenograft tumor models were established. Results: Circulating LBX2-AS1 was up-regulated in MM patients and positively correlated to LBX2 expression. Area under the curve (AUC) of LBX2-AS1 expression was 0.7525. Its up-regulation was also found in MM cells and primarily distributed in cytoplasm. LBX2-AS1 knockdown distinctly weakened proliferative ability and induced apoptosis in MM cells. Overexpressing LBX2-AS1 markedly strengthened LBX2 expression by increasing its mRNA stability. Rescue assays showed that silencing LBX2-AS1 distinctly weakened the pcDNA3.1-LBX2-induced increase in proliferation and decrease in apoptosis for MM cells. Silencing LBX2-AS1 markedly weakened tumor growth. Conclusion: Our data demonstrated that circulating LBX2-AS1 could be an underlying diagnostic marker in MM. Targeting LBX2-AS1 suppressed tumor progression by affecting mRNA stability of LBX2 in MM. Hence, LBX2-AS1 could be a novel therapeutic marker against MM.
Collapse
Affiliation(s)
- Haipeng Jia
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Yan Liu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Sulong Lv
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Ruifang Qiao
- Respiratory Intensive Care Unit, Tai'an City Central Hospital, Tai'an, China
| | - Xiaofen Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Fei Niu
- Department of Emergency, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Wenqing Shang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Shumei Liu
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Jing Dong
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| | - Zhirong Zhang
- Department of Hematology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, China
| |
Collapse
|
20
|
Kode VR, Hinkle KR, Ao G. Interaction of DNA-Complexed Boron Nitride Nanotubes and Cosolvents Impacts Dispersion and Length Characteristics. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:10934-10944. [PMID: 34496213 DOI: 10.1021/acs.langmuir.1c01309] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Processing boron nitride nanotubes (BNNTs) for applications ranging from nanomedicine to electronics generally requires dispersions of nanotubes that are stable in various compounds and solvents. We show that alcohol/water cosolvents, particularly isopropyl alcohol (IPA), are essential for the complexation of BNNTs with DNA under mild bath sonication. The resulting DNA-wrapped BNNT complexes are highly stable during purification and solvent exchange from cosolvents to water, providing potential for the versatile liquid-phase processing of BNNTs. Via molecular dynamics simulations, we demonstrate that IPA assists in the solvation of BNNTs due to its pseudosurfactant nature by verifying that water is replaced in the solvation layer as IPA is added. We quantify the solvation free energy of BNNTs in various IPA/water mixtures and observe a nonmonotonic trend, highlighting the importance of utilizing solvent-nanotube interactions in nanomaterial dispersions. Additionally, we show that nanotube lengths can be characterized by rheology measurements via determining the viscosity of dilute dispersions of DNA-BNNTs. This represents the bulk sample property in the liquid state, as compared to conventional imaging techniques that require surface deposition and drying. Our results also demonstrate that BNNT dispersions exhibit the rheological behavior of dilute Brownian rigid rods, which can be further exploited for the controlled processing and property enhancement of BNNT-enabled assemblies such as films and fibers.
Collapse
Affiliation(s)
- Venkateswara R Kode
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
| | - Kevin R Hinkle
- Department of Chemical and Materials Engineering, University of Dayton, 300 College Park, Dayton, Ohio 45469, United States
| | - Geyou Ao
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
| |
Collapse
|
21
|
Tombolan L, Millino C, Pacchioni B, Cattelan M, Zin A, Bonvini P, Bisogno G. Circulating miR-26a as Potential Prognostic Biomarkers in Pediatric Rhabdomyosarcoma. Front Genet 2020; 11:606274. [PMID: 33362864 PMCID: PMC7758343 DOI: 10.3389/fgene.2020.606274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Rhabdomyosarcoma (RMS) arises from myogenic precursors that fail to complete muscle differentiation and represents the most frequent soft tissue sarcoma in children. Two major histological subtypes are recognized: alveolar RMS, characterized by a more aggressive behavior and a greater proneness to metastasis, and embryonal RMS which accounts for the 80% of cases and carries a better prognosis. Despite the survival of patients with localized tumors has progressively improved, RMS remains a challenging disease especially for metastatic patients and in case of progressive or recurrent disease after front-line therapy. MicroRNAs, a class of small non-coding RNA, have emerged as crucial players in cancer development and progression, and their detection in plasma (circulating miRNAs) represents a promising minimally invasive approach that deserve to be exploited in clinical practice. We evaluated the utility of circulating miRNAs as diagnostic and prognostic biomarkers in children with RMS profiling miRNAs from plasma of a small cohort of RMS patients and healthy donors (HD) using a qPCR Cancer Panel. An assessment of hemolysis status of plasma using miR-451/miR-23a ratio was performed as pre-analytical analysis. Statistical analysis revealed that miRNAs expression pattern clearly distinguished RMS patients from HD (p < 0.05). Interestingly, plasma levels of muscle-specific miR-206 were found to be significantly increased in RMS patients compared to HD, whereas levels of three potential tumor-suppressor miRNAs, miR-26a and miR-30b/30c, were found lower. Reduced levels of circulating miR-26a and miR-30b/c were further measured in an independent larger cohort of patients (validation set) by digital droplet PCR. In particular, we evidenced that miR-26a absolute plasma levels were associated with fusion status and adverse outcome (p < 0.05). Taken together, these findings demonstrate the potential of circulating miRNA as diagnostic and prognostic biomarker in children affected by this malignancy and enforced the key role of miR-26a in pediatric rhabdomyosarcoma.
Collapse
Affiliation(s)
- Lucia Tombolan
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy
| | - Caterina Millino
- Functional Genomics Laboratory, Department of Biology, University of Padua, Padua, Italy
| | - Beniamina Pacchioni
- Functional Genomics Laboratory, Department of Biology, University of Padua, Padua, Italy
| | - Manuela Cattelan
- Department of Statistical Sciences, University of Padua, Padua, Italy
| | - Angelica Zin
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy
| | - Paolo Bonvini
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy
| | - Gianni Bisogno
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy.,Department of Woman's and Children's Health, Hematology and Oncology Unit, University of Padua, Padua, Italy
| |
Collapse
|
22
|
Alfarra H, Weir J, Grieve S, Reiman T. Targeting NK Cell Inhibitory Receptors for Precision Multiple Myeloma Immunotherapy. Front Immunol 2020; 11:575609. [PMID: 33304346 PMCID: PMC7693637 DOI: 10.3389/fimmu.2020.575609] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Innate immune surveillance of cancer involves multiple types of immune cells including the innate lymphoid cells (ILCs). Natural killer (NK) cells are considered the most active ILC subset for tumor elimination because of their ability to target infected and malignant cells without prior sensitization. NK cells are equipped with an array of activating and inhibitory receptors (IRs); hence NK cell activity is controlled by balanced signals between the activating and IRs. Multiple myeloma (MM) is a hematological malignancy that is known for its altered immune landscape. Despite improvements in therapeutic options for MM, this disease remains incurable. An emerging trend to improve clinical outcomes in MM involves harnessing the inherent ability of NK cells to kill malignant cells by recruiting NK cells and enhancing their cytotoxicity toward the malignant MM cells. Following the clinical success of blocking T cell IRs in multiple cancers, targeting NK cell IRs is drawing increasing attention. Relevant NK cell IRs that are attractive candidates for checkpoint blockades include KIRs, NKG2A, LAG-3, TIGIT, PD-1, and TIM-3 receptors. Investigating these NK cell IRs as pathogenic agents and therapeutic targets could lead to promising applications in MM therapy. This review describes the critical role of enhancing NK cell activity in MM and discusses the potential of blocking NK cell IRs as a future MM therapy.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents, Immunological/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Cytotoxicity, Immunologic/drug effects
- Humans
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/therapeutic use
- Immunotherapy, Adoptive/adverse effects
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Killer Cells, Natural/transplantation
- Molecular Targeted Therapy
- Multiple Myeloma/drug therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/metabolism
- Multiple Myeloma/pathology
- Receptors, Natural Killer Cell/antagonists & inhibitors
- Receptors, Natural Killer Cell/metabolism
- Signal Transduction
- Tumor Escape
- Tumor Microenvironment
Collapse
Affiliation(s)
- Helmi Alfarra
- Department of Biology, University of New Brunswick, Saint John, NB, Canada
| | - Jackson Weir
- Department of Biology, University of New Brunswick, Saint John, NB, Canada
| | - Stacy Grieve
- Department of Biology, University of New Brunswick, Saint John, NB, Canada
| | - Tony Reiman
- Department of Biology, University of New Brunswick, Saint John, NB, Canada
- Department of Oncology, Saint John Regional Hospital, Saint John, NB, Canada
- Department of Medicine, Dalhousie University, Saint John, NB, Canada
| |
Collapse
|
23
|
The Circular Life of Human CD38: From Basic Science to Clinics and Back. Molecules 2020; 25:molecules25204844. [PMID: 33096610 PMCID: PMC7587951 DOI: 10.3390/molecules25204844] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/08/2020] [Accepted: 10/15/2020] [Indexed: 11/16/2022] Open
Abstract
Monoclonal antibodies (mAbs) were initially considered as a possible “magic bullet” for in vivo elimination of tumor cells. mAbs represented the first step: however, as they were murine in nature (the earliest experience on the field), they were considered unfit for human applications. This prompted the development of techniques for cloning the variable regions of conventional murine antibodies, genetically mounted on human IgG. The last step in this years-long process was the design for the preparation of fully human reagents. The choice of the target molecule was also problematic, since cancer-specific targets are quite limited in number. To overcome this obstacle in the planning phases of antibody-mediated therapy, attention was focused on a set of normal molecules, whose quantitative distribution may balance a tissue-dependent generalized expression. The results and clinical success obtained with anti-CD20 mAbs revived interest in this type of strategy. Using multiple myeloma (MM) as a tumor model was challenging first of all because the plasma cells and their neoplastic counterpart eluded the efforts of the Workshop on Differentiation Antigens to find a target molecule exclusively expressed by these cells. For this reason, attention was turned to surface molecules which fulfill the requisites of being reasonably good targets, even if not specifically restricted to tumor cells. In 2009, we proposed CD38 as a MM target in virtue of its expression: it is absent on early hematological progenitors, has variable but generalized limited expression by normal cells, but is extremely high in plasma cells and in myeloma. Further, regulation of its expression appeared to be dependent on a variety of factors, including exposure to all-trans retinoic acid (ATRA), a potent and highly specific inducer of CD38 expression in human promyelocytic leukemia cells that are now approved for in vivo use. This review discusses the history of human CD38, from its initial characterization to its targeting in antibody-mediated therapy of human myeloma.
Collapse
|
24
|
Soliman AM, Lin TS, Mahakkanukrauh P, Das S. Role of microRNAs in Diagnosis, Prognosis and Management of Multiple Myeloma. Int J Mol Sci 2020; 21:E7539. [PMID: 33066062 PMCID: PMC7589124 DOI: 10.3390/ijms21207539] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/19/2020] [Accepted: 10/02/2020] [Indexed: 12/18/2022] Open
Abstract
Multiple myeloma (MM) is a cancerous bone disease characterized by malignant transformation of plasma cells in the bone marrow. MM is considered to be the second most common blood malignancy, with 20,000 new cases reported every year in the USA. Extensive research is currently enduring to validate diagnostic and therapeutic means to manage MM. microRNAs (miRNAs) were shown to be dysregulated in MM cases and to have a potential role in either progression or suppression of MM. Therefore, researchers investigated miRNAs levels in MM plasma cells and created tools to test their impact on tumor growth. In the present review, we discuss the most recently discovered miRNAs and their regulation in MM. Furthermore, we emphasized utilizing miRNAs as potential targets in the diagnosis, prognosis and treatment of MM, which can be useful for future clinical management.
Collapse
Affiliation(s)
- Amro M. Soliman
- Department of Biological Sciences—Physiology, Cell and Developmental Biology, University of Alberta, Edmonton, AB T6G 2R3, Canada;
| | - Teoh Seong Lin
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur 56000, Malaysia
| | - Pasuk Mahakkanukrauh
- Department of Anatomy & Excellence in Osteology Research and Training Center (ORTC), Chiang Mai University, Chiang Mai 50200, Thailand;
| | - Srijit Das
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
25
|
Cantwell MA, Chan KK, Sun XL, Ao G. Carbohydrate- and Chain Length-Controlled Complexation of Carbon Nanotubes by Glycopolymers. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:9878-9885. [PMID: 32787060 DOI: 10.1021/acs.langmuir.0c01498] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Stable dispersions of single-wall carbon nanotubes (SWCNTs) by biopolymers in an aqueous environment facilitate their potential biological and biomedical applications. In this report, we investigated a small library of precision synthesized glycopolymers with monosaccharide and disaccharide groups for stabilizing SWCNTs via noncovalent complexation in aqueous conditions. Among the glycopolymers tested, disaccharide lactose-containing glycopolymers demonstrate effective stabilization of SWCNTs in water, which strongly depends on carbohydrate density and polymer chain length as well. The introduction of disaccharide lactose potentially makes glycopolymers less flexible as compared to those containing monosaccharide and facilitates the wrapping conformation of polymers on the surface of SWCNTs while preserving intrinsic photoluminescence of nanotubes in the near-infrared region. This work demonstrates the synergistic effects of the identity of carbohydrate pendant groups and polymer chain length of glycopolymers on stabilizing SWCNTs in water, which has not been achieved previously.
Collapse
Affiliation(s)
- Michael A Cantwell
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
| | - Ka Keung Chan
- Department of Chemistry, Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
| | - Xue-Long Sun
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
- Department of Chemistry, Center for Gene Regulation in Health and Disease (GRHD), Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
| | - Geyou Ao
- Department of Chemical and Biomedical Engineering, Washkewicz College of Engineering, Cleveland State University, 2121 Euclid Avenue, Cleveland, Ohio 44115, United States
| |
Collapse
|