1
|
Li M, Wu W, Gu M, Su C, Wang X, Pan D, Xu Y, Wang L, Chen C, Yang M, Yan J. Purification-Free Bortezomib-Drug Conjugates Optimize Drug Economy and Cancer Therapeutic Synergy. ACS APPLIED MATERIALS & INTERFACES 2025; 17:23691-23706. [PMID: 40203453 DOI: 10.1021/acsami.5c02160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
As a promising candidate in overcoming resistance, providing synergy, and developing treatments, conjugated combination drugs mostly prevail over drug cocktails in establishing prodrugs and precisely codelivering multiple drugs for combination chemotherapies. However, current drug-drug conjugation methods (e.g., esterification, amidation, etherification, etc.) do not allow quantitative drug conversion and require necessary purification of crude products, resulting in a limited economy of initial drugs. Meanwhile, practical stimulus concentration in vivo usually fails to efficiently activate parent drug release from drug conjugates in target sites, which diminishes their efficacy. Herein, we report a click conjugation strategy based on boronic acid-cis diol complexation, realizing a fast (<30 min), quantitative, and purification-free conjugation of bortezomib (BTZ) and azacytidine (AZA) or capecitabine or doxifluridine. Notably, the BTZ-AZA conjugate spontaneously self-assembles into nanomedicine and exhibits enhanced synergistic efficacy. Furthermore, BTZ and AZA could be conjugated into a polyprodrug with controlled size and composition, and different organelle uptakes augment the synergy of BTZ-AZA conjugate by approximately 1000-fold versus free BTZ toward A549 adenocarcinoma cells (IC50: 0.55 nM versus 536.7 nM). This click strategy would expand the vision for developing smart combination drugs.
Collapse
Affiliation(s)
- Meng Li
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Wei Wu
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Min Gu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Chen Su
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Xinyu Wang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Yuping Xu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Lizhen Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Chongyang Chen
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Min Yang
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| | - Junjie Yan
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing 211166, P. R. China
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, P. R. China
| |
Collapse
|
2
|
Shafiei FS, Abroun S. Recent advancements in nanomedicine as a revolutionary approach to treating multiple myeloma. Life Sci 2024; 356:122989. [PMID: 39197575 DOI: 10.1016/j.lfs.2024.122989] [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: 05/21/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024]
Abstract
Multiple myeloma, the second most common hematological malignancy, remains incurable with a 5-year survival rate of approximately 50 % and recurrence rates near 100 %, despite significant attempts to develop effective medicines. Therefore, there is a pressing demand in the medical field for innovative and more efficient treatments for MM. Currently, the standard approach for treating MM involves administering high-dose chemotherapy, which frequently correlates with improved results; however, one major limiting factor is the significant side effects of these medications. Furthermore, the strategies used to deliver medications to tumors limit their efficacy, whether by rapid clearance from circulation or an insufficient concentration in cancer cells. Cancer treatment has shifted from cytotoxic, nonspecific chemotherapy regimens to molecularly targeted, rationally developed drugs with improved efficacy and fewer side effects. Nanomedicines may provide an effective alternative way to avoid these limits by delivering drugs into the complicated bone marrow microenvironment and efficiently reaching myeloma cells. Putting drugs into nanoparticles can make their pharmacokinetic and pharmacodynamic profiles much better. This can increase the drug's effectiveness in tumors, extend its time in circulation in the blood, and lower its off-target toxicity. In this review, we introduce several criteria for the rational design of nanomedicine to achieve the best anti-tumoral therapeutic results. Next, we discuss recent advances in nanomedicine for MM therapy.
Collapse
Affiliation(s)
- Fatemeh Sadat Shafiei
- Department Medical Laboratory Sciences, School of Paramedical Sciences, Zanjan University of Medical Sciences, Zanjan, Iran; Department of Biotechnology, Iranian Research Organization for Science & Technology (IROST), Tehran, Iran
| | - Saeid Abroun
- Department of Hematology, Faculty of Medical Science, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
3
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-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: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
4
|
Yan H, Gao S, Xu A, Zuo L, Zhang J, Zhao Y, Cheng Q, Yin X, Sun C, Hu Y. MALAT1 regulates network of microRNA-15a/16-VEGFA to promote tumorigenesis and angiogenesis in multiple myeloma. Carcinogenesis 2023; 44:760-772. [PMID: 37549238 DOI: 10.1093/carcin/bgad053] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 06/15/2023] [Accepted: 07/12/2023] [Indexed: 08/09/2023] Open
Abstract
MALAT1 is one of the most hopeful members implicated in angiogenesis in a variety of non-malignant diseases. In multiple myeloma (MM), MALAT1 is recognized as the most highly expressed long non-coding RNA. However, the functional roles of MALAT1 in angiogenesis and the responsible mechanisms have not yet been explored. Herein, we discovered a novel regulatory network dependent on MALAT1 in relation to MM tumorigenesis and angiogenesis. We observed that MALAT1 was upregulated in MM and significantly associated with poor overall survival. MALAT1 knockdown suppressed MM cell proliferation and promoted apoptosis, while restricting endothelial cells angiogenesis. Moreover, MALAT1 directly targeted microRNA-15a/16, and microRNA-15a/16 suppression partly reverted the effects of MALAT1 deletion on MM cells in vitro as well as tumor growth and angiogenesis in vivo. In addition, further study indicated that MALAT1 functioned as a competing endogenous RNA for microRNA-15a/16 to regulate vascular endothelial growth factor A (VEGFA) expression. Our results suggest that MALAT1 plays an important role in the regulatory axis of microRNA-15a/16-VEGFA to promote tumorigenicity and angiogenesis in MM. Consequently, MALAT1 could serve as a novel promising biomarker and a potential antiangiogenic target against MM.
Collapse
Affiliation(s)
- Han Yan
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Su Gao
- Department of Geriatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Institute of Gerontology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Aoshuang Xu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liping Zuo
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiasi Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuhong Zhao
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qianwen Cheng
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuejiao Yin
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunyan Sun
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Collaborative Innovation Center of Hematology, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Zhang F, Yang Q, Tang S, Jiang S, Zhao Q, Li J, Xu C, Liu J, Fu Y. CD38-targeted and erythrocyte membrane camouflaged nanodrug delivery system for photothermal and chemotherapy in multiple myeloma. Int J Pharm 2023; 643:123241. [PMID: 37479101 DOI: 10.1016/j.ijpharm.2023.123241] [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: 03/11/2023] [Revised: 06/19/2023] [Accepted: 07/15/2023] [Indexed: 07/23/2023]
Abstract
Multiple myeloma (MM) is a malignant and incurable disease. Chemotherapy is currently the primary treatment option for MM. However, chemotherapeutic drugs can interrupt treatment because of serious side effects. Therefore, development of novel therapeutics for MM is essential. In this study, we designed and constructed an innovative nanoparticle-based drug delivery system, P-R@Ni3P-BTZ, and investigated its feasibility, effectiveness, and safety both in vitro and in vivo. P-R@Ni3P-BTZ is a nanocomposite that consists of two parts: (1) the drug carrier (Ni3P), which integrates photothermal therapy (PTT) with chemotherapy by loading bortezomib (BTZ); and (2) the shell (P-R), a CD38 targeting peptide P-modified red blood cell membrane nanovesicles. In vitro and in vivo, it was proven that P-R@Ni3P-BTZ exhibits remarkable antitumor effects by actively targeting CD38 + MM cells. P-R@Ni3P-BTZ significantly induces the accumulation of intracellular reactive oxygen species (ROS) and increases the apoptosis of MM cells, which underlies the primary mechanism of its antitumor effects. In addition, P-R@Ni3P exhibits good biocompatibility and biosafety, both in vitro and in vivo. Overall, P-R@Ni3P-BTZ is a specific and efficient MM therapeutic method.
Collapse
Affiliation(s)
- Fangrong Zhang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Qin Yang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Sishi Tang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Siyi Jiang
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Cong Xu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China
| | - Jing Liu
- Department of Hematology, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China.
| | - Yunfeng Fu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha 410013, People's Republic of China.
| |
Collapse
|
6
|
Li J, Wang Q, Han Y, Jiang L, Lu S, Wang B, Qian W, Zhu M, Huang H, Qian P. Development and application of nanomaterials, nanotechnology and nanomedicine for treating hematological malignancies. J Hematol Oncol 2023; 16:65. [PMID: 37353849 PMCID: PMC10290401 DOI: 10.1186/s13045-023-01460-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Accepted: 05/30/2023] [Indexed: 06/25/2023] Open
Abstract
Hematologic malignancies (HMs) pose a serious threat to patients' health and life, and the five-year overall survival of HMs remains low. The lack of understanding of the pathogenesis and the complex clinical symptoms brings immense challenges to the diagnosis and treatment of HMs. Traditional therapeutic strategies for HMs include radiotherapy, chemotherapy, targeted therapy and hematopoietic stem cell transplantation. Although immunotherapy and cell therapy have made considerable progress in the last decade, nearly half of patients still relapse or suffer from drug resistance. Recently, studies have emerged that nanomaterials, nanotechnology and nanomedicine show great promise in cancer therapy by enhancing drug targeting, reducing toxicity and side effects and boosting the immune response to promote durable immunological memory. In this review, we summarized the strategies of recently developed nanomaterials, nanotechnology and nanomedicines against HMs and then proposed emerging strategies for the future designment of nanomedicines to treat HMs based on urgent clinical needs and technological progress.
Collapse
Affiliation(s)
- Jinxin Li
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Qiwei Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Yingli Han
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Lingli Jiang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Siqi Lu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Beini Wang
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Wenchang Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Meng Zhu
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - He Huang
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Pengxu Qian
- Center for Stem Cell and Regenerative Medicine and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
| |
Collapse
|
7
|
Detappe A, Nguyen HVT, Jiang Y, Agius MP, Wang W, Mathieu C, Su NK, Kristufek SL, Lundberg DJ, Bhagchandani S, Ghobrial IM, Ghoroghchian PP, Johnson JA. Molecular bottlebrush prodrugs as mono- and triplex combination therapies for multiple myeloma. NATURE NANOTECHNOLOGY 2023; 18:184-192. [PMID: 36702954 PMCID: PMC10032145 DOI: 10.1038/s41565-022-01310-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 12/06/2022] [Indexed: 05/19/2023]
Abstract
Cancer therapies often have narrow therapeutic indexes and involve potentially suboptimal combinations due to the dissimilar physical properties of drug molecules. Nanomedicine platforms could address these challenges, but it remains unclear whether synergistic free-drug ratios translate to nanocarriers and whether nanocarriers with multiple drugs outperform mixtures of single-drug nanocarriers at the same dose. Here we report a bottlebrush prodrug (BPD) platform designed to answer these questions in the context of multiple myeloma therapy. We show that proteasome inhibitor (bortezomib)-based BPD monotherapy slows tumour progression in vivo and that mixtures of bortezomib, pomalidomide and dexamethasone BPDs exhibit in vitro synergistic, additive or antagonistic patterns distinct from their corresponding free-drug counterparts. BPDs carrying a statistical mixture of three drugs in a synergistic ratio outperform the free-drug combination at the same ratio as well as a mixture of single-drug BPDs in the same ratio. Our results address unanswered questions in the field of nanomedicine, offering design principles for combination nanomedicines and strategies for improving current front-line monotherapies and combination therapies for multiple myeloma.
Collapse
Affiliation(s)
- Alexandre Detappe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Institut de Cancérologie Strasbourg Europe, Strasbourg, France
| | - Hung V-T Nguyen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Window Therapeutics, Boston, MA, USA
| | - Yivan Jiang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Window Therapeutics, Boston, MA, USA
| | - Michael P Agius
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Wencong Wang
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Clelia Mathieu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Nang K Su
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Samantha L Kristufek
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - David J Lundberg
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sachin Bhagchandani
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - P Peter Ghoroghchian
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| | - Jeremiah A Johnson
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
8
|
Ghasemi M, Liang S, Luu QM, Kempson I. The MTT Assay: A Method for Error Minimization and Interpretation in Measuring Cytotoxicity and Estimating Cell Viability. Methods Mol Biol 2023; 2644:15-33. [PMID: 37142913 DOI: 10.1007/978-1-0716-3052-5_2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
The MTT assay is extensively used, most often to infer a measure of cytotoxicity of treatments to cells. As with any assay though, there are a number of limitations. The method described here is designed with consideration of how the MTT assay fundamentally works to account for, or at least identify, confounding factors in measurements. It also provides a decision-making framework to best interpret and complement the MTT assay to apply it as either a measure of metabolic activity or cell viability.
Collapse
Affiliation(s)
- Mahshid Ghasemi
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Sisi Liang
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Quang Minh Luu
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - Ivan Kempson
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia.
| |
Collapse
|
9
|
Liu Q, Ran R, Song M, Li X, Wu Z, Dai G, Xia R. LncRNA HCP5 acts as a miR-128-3p sponge to promote the progression of multiple myeloma through activating Wnt/β-catenin/cyclin D1 signaling via PLAGL2. Cell Biol Toxicol 2022; 38:979-993. [PMID: 34331612 DOI: 10.1007/s10565-021-09628-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Accepted: 06/24/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Although long non-coding RNA (lncRNA) HCP plays essential roles in human cancers, its function and mechanism in multiple myeloma (MM) have not crystallized. METHODS HCP5 level in MM was assessed through qRT-PCR. A series of functional investigations were conducted to evaluate the influences of HCP5 on proliferation and apoptosis. Bioinformatics analysis and RIP/RNA pull-down assays were carried out to determine the relationships among HCP5, miR-128-3p, and PLAGL2. Relative protein level was determined through Western blot. A xenograft tumor model was applied for validating the roles of HCP5/miR-128-3p/PLAGL2 axis in vivo. RESULTS HCP5 was significantly increased in MM. HCP5 knockdown effectively thwarted the proliferative rate and cell cycle of MM cell lines and suppressed tumor growth. HCP5 regulated PLAGL2 expression by sponging miR-128-3p. PLAGL2 overexpression effectively rescued cells from influences by sh-HCP5 on cell proliferative and apoptotic rates. Additionally, HCP5 knockdown significantly inhibited Wnt/β-catenin/cyclin D1 signaling, and these effects were eliminated by PLAGL2 overexpression. CONCLUSION Our study revealed that HCP5/miR-128-3p/PLAGL2 is closely correlated to MM development by modulating Wnt/β-catenin/cyclin D1 signaling. HCP5 promoted cell proliferation and tumor formation of MM cells by activating the Wnt/β-catenin/CCND1 signaling pathway by sponging miR-128-3p to increase PLAGL2 expression.
Collapse
Affiliation(s)
- Qinhua Liu
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ruonan Ran
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Mingyue Song
- Department of Hematology, the Chaohu Hospital Affiliated to Anhui Medical University, Chaohu, Anhui, China
| | - Xiaodan Li
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Zhengsheng Wu
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Guanrong Dai
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China
| | - Ruixiang Xia
- Department of Hematology, the First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei, 230022, Anhui, China.
| |
Collapse
|
10
|
Yang P, Qu Y, Wang M, Chu B, Chen W, Zheng Y, Niu T, Qian Z. Pathogenesis and treatment of multiple myeloma. MedComm (Beijing) 2022; 3:e146. [PMID: 35665368 PMCID: PMC9162151 DOI: 10.1002/mco2.146] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 04/27/2022] [Accepted: 04/27/2022] [Indexed: 02/05/2023] Open
Abstract
Multiple myeloma (MM) is the second‐ranking malignancy in hematological tumors. The pathogenesis of MM is complex with high heterogeneity, and the development of the disease is a multistep process. Chromosomal translocations, aneuploidy, genetic mutations, and epigenetic aberrations are essential in disease initiation and progression. The correlation between MM cells and the bone marrow microenvironment is associated with the survival, progression, migration, and drug resistance of MM cells. In recent decades, there has been a significant change in the paradigm for the management of MM. With the development of proteasome inhibitors, immunomodulatory drugs, monoclonal antibodies, chimeric antigen receptor T‐cell therapies, and novel agents, the survival of MM patients has been significantly improved. In addition, nanotechnology acts as both a nanocarrier and a treatment tool for MM. The properties and responsive conditions of nanomedicine can be tailored to reach different goals. Nanomedicine with a precise targeting property has offered great potential for drug delivery and assisted in tumor immunotherapy. In this review, we summarize the pathogenesis and current treatment options of MM, then overview recent advances in nanomedicine‐based systems, aiming to provide more insights into the treatment of MM.
Collapse
Affiliation(s)
- Peipei Yang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Ying Qu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Mengyao Wang
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Bingyang Chu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Wen Chen
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Yuhuan Zheng
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Ting Niu
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| | - Zhiyong Qian
- Department of Hematology and Institute of Hematology, State Key Laboratory of Biotherapy and Cancer Center West China Hospital Sichuan University Chengdu Sichuan China
| |
Collapse
|
11
|
Mehrarya M, Gharehchelou B, Kabarkouhi Z, Ataei S, Esfahani FN, Wintrasiri MN, Mozafari MR. Functionalized Nanostructured Bioactive Carriers: Nanoliposomes, Quantum Dots, Tocosome and Theranostic Approach. Curr Drug Deliv 2022; 19:1001-1011. [PMID: 35331111 DOI: 10.2174/1567201819666220324092933] [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] [Received: 08/24/2021] [Revised: 12/20/2021] [Accepted: 01/19/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Lipidic nanocarriers have great potential for the encapsulation and delivery of numerous bioactive compounds. They have demonstrated significant benefits over traditional disease management and conventional therapy. The benefits associated with the particular properties of lipidic nanocarriers include site-specific drug deposition, improved pharmacokinetics and pharmacodynamics, enhanced internalization and intracellular transport, biodegradability, and decreased biodistribution. These properties result in the alleviation of the harmful consequences of conventional treatment protocols. Scope and approach: The administration of various bioactive molecules has been extensively investigated using nanostructured lipid carriers. In this article, theranostic applications of novel formulations of lipidic nanocarriers combined or complexed with quantum dots, certain polymers such as chitosan, and metallic nanoparticles (particularly gold) are reviewed. These formulations have demonstrated better controlled release features, improved drug loading capability, as well as a lower burst release rate. As a recent innovation in the field of drug delivery, tocosomes and their unique advantages are also explained in the final section of this entry. KEY FINDINGS AND CONCLUSIONS Theranostic medicine requires nanocarriers with improved target-specific accumulation and bio-distribution. Towards this end, lipid-based nanocarrier systems and tocosomes combined with unique properties of quantum dots, biocompatible polymers, and metallic nanoparticles seem to be ideal candidates to be considered for safe and efficient drug delivery.
Collapse
Affiliation(s)
- Mehrnoush Mehrarya
- Department of Applied Biology and Chemical Technology, Hong Kong Polytechnic University, Hong Kong, China
| | | | - Zeinab Kabarkouhi
- Laser and Plasma Research Institute, Shahid Beheshti University, and Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Sara Ataei
- Department of Clinical Pharmacy, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Fahime Nasr Esfahani
- Department of Clinical Pharmacy, School of Pharmacy, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Milint Neleptchenko Wintrasiri
- Supreme NanoBiotics Co. Ltd. and Supreme Pharmatech Co. Ltd., 399/90-95 Moo 13 Kingkaew Rd. Soi 25/1, T. Rachateva, A. Bangplee, Samutprakan 10540, Thailand
| | - M R Mozafari
- Supreme NanoBiotics Co. Ltd. and Supreme Pharmatech Co. Ltd., 399/90-95 Moo 13 Kingkaew Rd. Soi 25/1, T. Rachateva, A. Bangplee, Samutprakan 10540, Thailand
| |
Collapse
|
12
|
Gu W, Qu R, Meng F, Cornelissen JJLM, Zhong Z. Polymeric nanomedicines targeting hematological malignancies. J Control Release 2021; 337:571-588. [PMID: 34364920 DOI: 10.1016/j.jconrel.2021.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/31/2021] [Accepted: 08/02/2021] [Indexed: 10/20/2022]
Abstract
Hematological malignancies (HMs) typically persisting in the blood, lymphoma, and/or bone marrow invalidate surgery and local treatments clinically used for solid tumors. The presence and drug resistance nature of cancer stem cells (CSCs) further lends HMs hard to cure. The development of new treatments like molecular targeted drugs and antibodies has improved the clinical outcomes for HMs but only to a certain extent, due to issues of low bioavailability, moderate response, occurrence of drug resistance, and/or dose-limiting toxicities. In the past years, polymeric nanomedicines targeting HMs including refractory and relapsed lymphoma, leukemia and multiple myeloma have emerged as a promising chemotherapeutic approach that is shown capable of overcoming drug resistance, delivering drugs not only to cancer cells but also CSCs, and increasing therapeutic index by lessening drug-associated adverse effects. In addition, polymeric nanomedicines have shown to potentiate next-generation anticancer modalities such as therapeutic proteins and nucleic acids in effectively treating HMs. In this review, we highlight recent advance in targeted polymeric nanoformulations that are coated with varying ligands (e.g. cancer cell membrane proteins, antibodies, transferrin, hyaluronic acid, aptamer, peptide, and folate) and loaded with different therapeutic agents (e.g. chemotherapeutics, molecular targeted drugs, therapeutic antibodies, nucleic acid drugs, and apoptotic proteins) for directing to distinct targets (e.g. CD19, CD20, CD22, CD30, CD38, CD44, CD64, CXCR, FLT3, VLA-4, and bone marrow microenvironment) in HMs. The advantages and potential challenges of different designs are discussed.
Collapse
Affiliation(s)
- Wenxing Gu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China; Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, the Netherlands
| | - Ruobing Qu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| | - Jeroen J L M Cornelissen
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE Enschede, the Netherlands.
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, PR China.
| |
Collapse
|
13
|
Wu L, Xia L, Jiang H, Hu Y, Li L, Xu L, Xia R. Long non‑coding RNA DANCR represses the viability, migration and invasion of multiple myeloma cells by sponging miR‑135b‑5p to target KLF9. Mol Med Rep 2021; 24:649. [PMID: 34278459 DOI: 10.3892/mmr.2021.12288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 03/13/2021] [Indexed: 11/06/2022] Open
Abstract
Multiple myeloma (MM) is a malignancy of plasma cells that leads to marrow failure and bone lesions. Numerous studies have verified the link between long non‑coding RNAs (lncRNAs) and MM. The present study aimed to examine the role and underlying mechanism of differentiation antagonizing non‑protein coding RNA (DANCR) in MM cells. The relative expression levels of DANCR, microRNA (miR)‑135b‑5p and Krüppel‑like factor 9 (KLF9) were examined using reverse transcription‑quantitative PCR. Cell viability was assessed using the MTT assay, while relative cell migration and invasion were evaluated using Transwell assays. Moreover, the dual‑luciferase reporter assay was used to examine the interplay between DANCR, miR‑135b‑5p and KLF9. Western blotting was performed to determine the expression level of KLF9. It was found that lncRNA DANCR and KLF9 were downregulated, while miR‑135b‑5p was upregulated in the serum of patients with MM and in MM cells compared with the controls. Overexpressing DANCR or knocking down miR‑135b‑5p reduced the viability of the MM cells, as well as restrained MM cells from migrating and invading. Furthermore, DANCR directly targeted miR‑135b‑5p and was negatively correlated with miR‑135b‑5p. It was also found that KLF9 was targeted by miR‑135b‑5p and was inversely correlated with miR‑135b‑5p expression. The impact of lncRNA DANCR‑mediated suppression on cell viability, invasion and migration was partially abolished by short hairpin RNA KLF9 or miR‑135b‑5p mimics transfection in MM cells. Thus, it was suggested that lncRNA DANCR repressed the viability, migration and invasion of MM cells by sponging miR‑135b‑5p to target KLF9.
Collapse
Affiliation(s)
- Linlin Wu
- Department of Hematology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Liang Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Huimin Jiang
- Department of Hematology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Yue Hu
- Department of Hematology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Lingling Li
- Department of Hematology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Li Xu
- Department of Hematology, Anhui No. 2 Provincial People's Hospital, Hefei, Anhui 230041, P.R. China
| | - Ruixiang Xia
- Department of Hematology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| |
Collapse
|
14
|
Federico C, Alhallak K, Sun J, Duncan K, Azab F, Sudlow GP, de la Puente P, Muz B, Kapoor V, Zhang L, Yuan F, Markovic M, Kotsybar J, Wasden K, Guenthner N, Gurley S, King J, Kohnen D, Salama NN, Thotala D, Hallahan DE, Vij R, DiPersio JF, Achilefu S, Azab AK. Tumor microenvironment-targeted nanoparticles loaded with bortezomib and ROCK inhibitor improve efficacy in multiple myeloma. Nat Commun 2020; 11:6037. [PMID: 33247158 PMCID: PMC7699624 DOI: 10.1038/s41467-020-19932-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 10/23/2020] [Indexed: 12/21/2022] Open
Abstract
Drug resistance and dose-limiting toxicities are significant barriers for treatment of multiple myeloma (MM). Bone marrow microenvironment (BMME) plays a major role in drug resistance in MM. Drug delivery with targeted nanoparticles have been shown to improve specificity and efficacy and reduce toxicity. We aim to improve treatments for MM by (1) using nanoparticle delivery to enhance efficacy and reduce toxicity; (2) targeting the tumor-associated endothelium for specific delivery of the cargo to the tumor area, and (3) synchronizing the delivery of chemotherapy (bortezomib; BTZ) and BMME-disrupting agents (ROCK inhibitor) to overcome BMME-induced drug resistance. We find that targeting the BMME with P-selectin glycoprotein ligand-1 (PSGL-1)-targeted BTZ and ROCK inhibitor-loaded liposomes is more effective than free drugs, non-targeted liposomes, and single-agent controls and reduces severe BTZ-associated side effects. These results support the use of PSGL-1-targeted multi-drug and even non-targeted liposomal BTZ formulations for the enhancement of patient outcome in MM.
Collapse
Affiliation(s)
- Cinzia Federico
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Kinan Alhallak
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Jennifer Sun
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Kathleen Duncan
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Feda Azab
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gail P Sudlow
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Pilar de la Puente
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbara Muz
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vaishali Kapoor
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Luna Zhang
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
| | - Fangzheng Yuan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
| | - Matea Markovic
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
| | - Joseph Kotsybar
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
| | - Katherine Wasden
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicole Guenthner
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shannon Gurley
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Justin King
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daniel Kohnen
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Noha N Salama
- Department of Pharmaceutical and Administrative Sciences, St. Louis College of Pharmacy, St. Louis, MO, USA
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Dinesh Thotala
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dennis E Hallahan
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA
| | - Ravi Vij
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - John F DiPersio
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Samuel Achilefu
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Abdel Kareem Azab
- Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University, St. Louis, MO, USA.
| |
Collapse
|
15
|
Iannazzo D, Ettari R, Giofrè S, Eid AH, Bitto A. Recent Advances in Nanotherapeutics for Multiple Myeloma. Cancers (Basel) 2020; 12:cancers12113144. [PMID: 33120945 PMCID: PMC7693822 DOI: 10.3390/cancers12113144] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 10/26/2020] [Indexed: 12/13/2022] Open
Abstract
Simple Summary Nanotherapeutics are useful tools to improve the deliverability of drugs, especially anti-cancer drugs that need to target specific cells. Several approaches have been studied for multiple myeloma, considering that immune cells are not easy to target with the available drugs. These pharmacological agents are administered in various combinations using Thalidomide (or Lenalidomide, Pomalidomide), corticosteroids (Dexamethasone), proteasome inhibitors (Bortezomib, Carfilzomib, Ixazomib), deacetylase inhibitors (Panobinostat), and monoclonal antibodies (Elotuzumab, Daratumumab). As all drugs these agents might have serious side effects and in addition, the reliance on stochastic events to deliver drugs to tumors reduces their effectiveness either through rapid clearance from blood or inadequate concentration in cancer cells. To address these issues liposomes, micelles, polymeric nanoparticles, inorganic nanoparticles, and carbon-based nanomaterials have been successfully tested in vivo and can be considered as useful tools to improve delivery of active pharmaceuticals that show poor bioavailability or poor internalization into myeloma cells. Abstract Anticancer therapies cannot be included in a one-size-fits-all scenario; it is imperative to adapt therapies to the tumor molecular profile and most importantly to develop target-specific therapeutics. Nanotherapeutics can combine molecular imaging with molecular therapy in order to provide the maximum benefit to patients in terms of disease prevention, identification, and treatment. Nanotechnology applied to therapy provides numerous advantages in diagnostics and in drug delivery, especially for those malignant cells that are difficult to target or for drugs with poor bioavailability, such as those used for multiple myeloma (MM). This review summarizes the recent advances in the development of nanoparticle-based systems for the treatment of MM, taking into account the methods used for their functionalization, biocompatibility, and anticancer activity.
Collapse
Affiliation(s)
- Daniela Iannazzo
- Department of Engineering, University of Messina, 98166 Messina, Italy;
| | - Roberta Ettari
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, 98165 Messina, Italy; (R.E.); (S.G.)
| | - Salvatore Giofrè
- Department of Chemical, Biological, Pharmaceutical and Environmental Chemistry, University of Messina, 98165 Messina, Italy; (R.E.); (S.G.)
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, 2713 Doha, Qatar;
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, 2713 Doha, Qatar
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, 11-0236 Beirut, Lebanon
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
- Correspondence:
| |
Collapse
|
16
|
Mittelheisser V, Banerjee M, Pivot X, Charbonnière LJ, Goetz J, Detappe A. Leveraging Immunotherapy with Nanomedicine. ADVANCED THERAPEUTICS 2020. [DOI: 10.1002/adtp.202000134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Vincent Mittelheisser
- Centre Paul Strauss Strasbourg 67000 France
- INSERM UMR_S1109 Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg 67000 France
| | - Mainak Banerjee
- Centre Paul Strauss Strasbourg 67000 France
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| | - Xavier Pivot
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
| | - Loïc J. Charbonnière
- Université de Strasbourg Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| | - Jacky Goetz
- INSERM UMR_S1109 Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Fédération de Médecine Translationnelle de Strasbourg (FMTS) Strasbourg 67000 France
| | - Alexandre Detappe
- Centre Paul Strauss Strasbourg 67000 France
- Université de Strasbourg Strasbourg 67000 France
- Institut de Cancérologie Strasbourg Europe Strasbourg 67000 France
- Institut Pluridisciplinaire Hubert Curien CNRS UMR‐7178 Strasbourg 67087 France
| |
Collapse
|
17
|
Li XY, Li S, Lu GQ, Wang DP, Liu KL, Qian XH, Xue WH, Meng FH. Design, synthesis and biological evaluation of novel (E)-N-phenyl-4-(pyridine-acylhydrazone) benzamide derivatives as potential antitumor agents for the treatment of multiple myeloma (MM). Bioorg Chem 2020; 103:104189. [PMID: 32890996 DOI: 10.1016/j.bioorg.2020.104189] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/06/2020] [Accepted: 08/08/2020] [Indexed: 12/12/2022]
Abstract
A series of novel (E)-N-phenyl-4-(pyridine-acylhydrazone) benzamide derivatives were designed, synthesized, and evaluated for their anti-proliferative activity against two different human cancer cell lines and one human normal cell line. Compound 8b had the best anti-proliferative activity (IC50 = 0.12 ± 0.09 μM, RPMI8226 cells) than the other compounds. And compound 8b had lower toxicity than imatinib. Flow cytometry analysis showed that compound 8b could arrest the cell cycle at the G0/G1 phase, and induce apoptosis of RPMI8226 cells by promoting mitochondrial ROS release, thereby effectively inhibiting cell proliferation. Our findings provided a promising lead compound 8b for further structural optimization and will be instructive for the discovery of more potent antitumor drugs with high selectivity and low toxicity.
Collapse
Affiliation(s)
- Xin-Yang Li
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China; Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang 110122, China
| | - Shuai Li
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Guo-Qing Lu
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - De-Pu Wang
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Kai-Li Liu
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Xin-Hua Qian
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Wen-Han Xue
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China
| | - Fan-Hao Meng
- School of Pharmacy, China Medical University, 77 Puhe Road, North New Area, Shenyang 110122, China.
| |
Collapse
|
18
|
Liu N, Feng S, Li H, Chen X, Bai S, Liu Y. Long non-coding RNA MALAT1 facilitates the tumorigenesis, invasion and glycolysis of multiple myeloma via miR-1271-5p/SOX13 axis. J Cancer Res Clin Oncol 2020; 146:367-379. [PMID: 31953613 PMCID: PMC6985203 DOI: 10.1007/s00432-020-03127-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022]
Abstract
Background Long non-coding RNAs (lncRNAs) play crucial roles in the regulation and treatment of multiple myeloma (MM). The objective of this research was to study the functional mechanism of metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) in MM. Methods MALAT1, microRNA-1271-5p (miR-1271-5p), and SRY-Box 13 (SOX13) levels were examined by quantitative real-time polymerase chain reaction (qRT-PCR). Cell viability, apoptosis, and invasion were respectively assayed using 3-(4, 5-dimethylthiazol-2-y1)-2, 5-diphenyl tetrazolium bromide (MTT), flow cytometry, and transwell assay. Glycolysis was evaluated by glucose consumption, lactate production, ATP/ADP ratio, and the detection of related enzymes. Associated proteins were measured using Western blot. Target relation was verified via dual-luciferase reporter assay. Xenograft tumor assay was implemented to study the influence of MALAT1 on MM in vivo. Results The up-regulation of MALAT1 and the down-regulation of miR-1271-5p were found in MM serums and cells. MALAT1 knockdown suppressed cell viability, invasion, and glycolysis while expedited cell apoptosis in MM cells. MALAT1 directly targeted miR-1271-5p and miR-1271-5p depression reverted the effects of MALAT1 knockdown on MM cells. SOX13 was a target of miR-1271-5p and SOX13 overexpression weakened the effects of miR-1271-5p on MM. MALAT1 indirectly modulated SOX13 expression through targeting miR-1271-5p. MALAT1 down-regulation inhibited MM growth by miR-1271-5p/SOX13 axis in vivo. Conclusion LncRNA MALAT1 expedited MM tumorigenesis, invasion, and glycolysis via miR-1271-5p/SOX13 axis. MALAT1 might contribute to the therapy of MM as a promising indicator.
Collapse
Affiliation(s)
- Na Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Song Feng
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Huanhuan Li
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Xiaoguang Chen
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Songting Bai
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China
| | - Yufeng Liu
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Erqi District, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
19
|
Gu W, An J, Meng H, Yu N, Zhong Y, Meng F, Xu Y, Cornelissen JJLM, Zhong Z. CD44-Specific A6 Short Peptide Boosts Targetability and Anticancer Efficacy of Polymersomal Epirubicin to Orthotopic Human Multiple Myeloma. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904742. [PMID: 31560141 DOI: 10.1002/adma.201904742] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 09/11/2019] [Indexed: 06/10/2023]
Abstract
Chemotherapy is widely used in the clinic though its benefits are controversial owing to low cancer specificity. Nanovehicles capable of selectively transporting drugs to cancer cells have been energetically pursued to remodel cancer treatment. However, no active targeting nanomedicines have succeeded in clinical translation to date, partly due to either modest targetability or complex fabrication. CD44-specific A6 short peptide (KPSSPPEE) functionalized polymersomal epirubicin (A6-PS-EPI), which boosts targetability and anticancer efficacy toward human multiple myeloma (MM) in vivo, is described. A6-PS-EPI encapsulating 11 wt% EPI is small (≈55 nm), robust, reduction-responsive, and easy to fabricate. Of note, A6 decoration markedly augments the uptake and anticancer activity of PS-EPI in CD44-overexpressing LP-1 MM cells. A6-PS-EPI displays remarkable targeting ability to orthotopic LP-1 MM, causing depleted bone damage and striking survival benefits compared to nontargeted PS-EPI. Overall, A6-PS-EPI, as a simple and intelligent nanotherapeutic, demonstrates high potential for clinical translation.
Collapse
Affiliation(s)
- Wenxing Gu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Jingnan An
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Hao Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Na Yu
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yinan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| | - Yang Xu
- Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Institute of Blood and Marrow Transplantation of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, 215123, P. R. China
| | - Jeroen J L M Cornelissen
- Department of Biomolecular Nanotechnology, MESA+ Institute for Nanotechnology, University of Twente, 7500 AE, Enschede, The Netherlands
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
20
|
Pashirova TN, Zhukova NA, Lukashenko SS, Valeeva FG, Burilova EA, Sapunova AS, Voloshina AD, Mirgorodskaya AB, Zakharova LY, Sinyashin OG, Mamedov VA. Multi-targeted approach by 2-benzimidazolylquinoxalines-loaded cationic arginine liposomes against сervical cancer cells in vitro. Colloids Surf B Biointerfaces 2019; 178:317-328. [PMID: 30884347 DOI: 10.1016/j.colsurfb.2019.03.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 02/28/2019] [Accepted: 03/10/2019] [Indexed: 01/04/2023]
Abstract
Multi-targeted approaches for inhibition of сervical cancer cells in vitro were developed by implementing two different strategies and drug combination for creation of new therapeutic target agents and for nanotechnological-enhancement of intracellular delivery. New 2-benzimidazolylquinoxalines derivatives were synthesized and characterized by combining two different pharmacophores - benzimidazole and quinoxaline rings directly bonded in their structures. Spectrophotometric technique for determination of content of compounds in various media was developed to evaluate their solubility in water and micellar solutions of surfactants. The bioavailability of poorly water-soluble 2-benzimidazolylquinoxalines was improved by PEGylated liposomes as antitumor drug delivery carriers. 2-benzimidazolylquinoxalines-loaded PEGylated liposomes, with size close to 100 nm and negative zeta potential ranging from -13 mV to -27 mV, were time-stable at room temperature. The design of liposomal formulations for improving cellular uptake and in vitro antitumor efficacy was performed by modification of liposome surface with the new arginine surfactant. The cell viability of 2-benzimidazolylquinoxalines-loaded arginine liposomes on human cancer M-Hela cells was 16% at the concentration 0.15 mg/ml. Moreover, these liposomes showed a lower toxicity (40%) against normal human Gang liver cells both at the lowest and highest tested concentrations.
Collapse
Affiliation(s)
- Tatiana N Pashirova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation.
| | - Nataliya A Zhukova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Svetlana S Lukashenko
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Farida G Valeeva
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Evgenia A Burilova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Anastasia S Sapunova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Alexandra D Voloshina
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Alla B Mirgorodskaya
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Lucia Y Zakharova
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation; Kazan National Research Technological University, Karl Marx St., 68, Kazan, 420015, Russian Federation.
| | - Oleg G Sinyashin
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| | - Vakhid A Mamedov
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center of Russian Academy of Sciences, Arbuzov St., 8, Kazan, 420088, Russian Federation
| |
Collapse
|