1
|
Yang A, Lu Y, Zhang Z, Wang J. Nanodrug Delivery Systems for Acute Lymphoblastic Leukemia Therapy. Pharmaceuticals (Basel) 2025; 18:639. [PMID: 40430460 PMCID: PMC12114507 DOI: 10.3390/ph18050639] [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: 03/25/2025] [Revised: 04/19/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
Acute lymphoblastic leukemia (ALL) is a malignant tumor caused by abnormal proliferation of B-line or T-line lymphocytes in the bone marrow. Traditional treatments have limitations. Because of their unique advantages, nanodrug delivery systems (NDDSs) show great potential in the treatment of ALL. In this paper, the pathological features of ALL, the limitations of current therapeutic methods, and the definition and composition of NDDSs were reviewed. Research strategies for the use of NDDSs in the treatment of ALL were discussed. In addition, challenges and future development directions of NDDSs in the treatment of ALL were also discussed, aiming to provide reference for the application of NDDSs in the diagnosis and treatment of ALL.
Collapse
Affiliation(s)
- Aiyun Yang
- Translational Medicine Laboratory, Beijing Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China;
| | - Yuanfang Lu
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China; (Y.L.); (Z.Z.)
| | - Zuo Zhang
- Beijing Key Laboratory of Environmental & Viral Oncology, College of Life Science & Bioengineering, Beijing University of Technology, Beijing 100124, China; (Y.L.); (Z.Z.)
| | - Jianhua Wang
- Translational Medicine Laboratory, Beijing Key Laboratory of Child Development and Nutriomics, Capital Institute of Pediatrics, Beijing 100020, China;
| |
Collapse
|
2
|
Abolhasani S, Salehi Khesht AM, Khodakarami A, Masjedi A, Rashidi B, Izadi S, Noukabadi FK, Karpisheh V, Poudeh KT, Jalali P, Salehi Z, Bagherifar R, Hejazian SS, Movassaghpour A, Feizi AAH, Jadidi F. Advancing the therapeutic effectiveness of paclitaxel in chronic lymphocytic leukemia through the simultaneous inhibition of NOTCH1 and SF3B1. Cancer Cell Int 2025; 25:104. [PMID: 40108537 PMCID: PMC11924741 DOI: 10.1186/s12935-025-03702-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Chemoresistance is still a significant obstacle to cancer therapy. Overexpression of the splicing factor 3b subunit 1 (SF3B1) and neurogenic locus notch homolog protein 1 (NOTCH1) factors is typically found in chronic lymphocytic leukemia (CLL), leading to the development of chemotherapy resistance. OBJECTIVE The current investigation aims to evaluate the chemosensitivity of CLL cells by blocking NOTCH1 and SF3B1 using chitosan lactate (CL) nanoparticles (NPs). METHODS We used CL-NPs loaded with anti-NOTCH1 and -SF3B1 small interfering RNAs (siRNAs) in combination with paclitaxel (PTX) to suppress NOTCH1 and SF3B1 in peripheral blood mononuclear cells (PBMCs) and bone marrow mononuclear cells (BMMCs) isolated from CLL cases to assess the impact of this therapeutic strategy on leukemic cell chemosensitivity. Further, the competing endogenous RNA (ceRNA) network that regulates NOTCH1 and -SF3B1 was constructed and enriched. RESULTS Our findings showed that CL-NPs loaded with anti-NOTCH1/-SF3B1 siRNAs-PTX significantly suppressed NOTCH1 and SF3B1 expression in PBMCs and BMMCs isolated from CLL cases in comparison with the untreated samples, leading to increased leukemic cell sensitivity to PTX and decreased the proliferative capacity of leukemic cells. The enrichment analysis highlighted the fundamental pathways where the NOTCH1- and SF3B1-associated ceRNA network exerts its influence in the context of CLL. CONCLUSIONS This study implies the efficacy of combined therapy by CL-NPs loaded with anti-NOTCH1/-SF3B1 siRNAs and PTX as a novel therapeutic strategy for CLL, even though further studies are required to warrant the findings.
Collapse
Affiliation(s)
- Shiva Abolhasani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Atefeh Khodakarami
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Masjedi
- Institute of Experimental Hematology, School of Medicine, Technical University of Munich, 81675, Munich, Germany
- Center for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675, Munich, Germany
| | - Bentolhoda Rashidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Izadi
- Research Center for Integrative Medicine in Aging, Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Vahid Karpisheh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Pooya Jalali
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Centre, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Salehi
- Hematology, Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Rafieh Bagherifar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - AliAkbar Movassaghpour
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
3
|
Li F, Wang H, Ye T, Guo P, Lin X, Hu Y, Wei W, Wang S, Ma G. Recent Advances in Material Technology for Leukemia Treatments. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313955. [PMID: 38547845 DOI: 10.1002/adma.202313955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/11/2024] [Indexed: 04/13/2024]
Abstract
Leukemia is a widespread hematological malignancy characterized by an elevated white blood cell count in both the blood and the bone marrow. Despite notable advancements in leukemia intervention in the clinic, a large proportion of patients, especially acute leukemia patients, fail to achieve long-term remission or complete remission following treatment. Therefore, leukemia therapy necessitates optimization to meet the treatment requirements. In recent years, a multitude of materials have undergone rigorous study to serve as delivery vectors or direct intervention agents to bolster the effectiveness of leukemia therapy. These materials include liposomes, protein-based materials, polymeric materials, cell-derived materials, and inorganic materials. They possess unique characteristics and are applied in a broad array of therapeutic modalities, including chemotherapy, gene therapy, immunotherapy, radiotherapy, hematopoietic stem cell transplantation, and other evolving treatments. Here, an overview of these materials is presented, describing their physicochemical properties, their role in leukemia treatment, and the challenges they face in the context of clinical translation. This review inspires researchers to further develop various materials that can be used to augment the efficacy of multiple therapeutic modalities for novel applications in leukemia treatment.
Collapse
Affiliation(s)
- Feng Li
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huaiji Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tong Ye
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peilin Guo
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xiaoyun Lin
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yuxing Hu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Wei Wei
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuang Wang
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guanghui Ma
- State Key Laboratory of Biochemical Engineering, Institute of Process Engineering, Chinese Academy of Sciences, Beijing, 100190, China
- Key Laboratory of Biopharmaceutical Preparation and Delivery, Chinese Academy of Sciences, Beijing, 100190, China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|
4
|
Heidarizadi S, Rashidi Z, Jalili C, Gholami M. Overview of biological effects of melatonin on testis: A review. Andrologia 2022; 54:e14597. [PMID: 36168927 DOI: 10.1111/and.14597] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/31/2022] [Accepted: 09/05/2022] [Indexed: 11/29/2022] Open
Abstract
Infertility is a major global health issue and male factors account for half of all infertility cases. One of the causes of male infertility is the loss of spermatogonial stem cells, which may occur because of chemotherapy, radiotherapy or genetic defects. In numerous animal species, the evidence suggests the pineal gland and melatonin secretion in their reproductive activities are involved. Recently, considerable attention has pointed to the usage of melatonin in the treatment of diseases. Melatonin is associated with the regulation of circadian and seasonal rhythmic functions, immune system functions, retinal physiology, spermatogenesis and inhibition of tumour growth in different species. Several studies demonstrated that melatonin acts as an anti-apoptotic, anti-inflammatory, anticancer and antioxidant agent. Melatonin can also protect testicles and spermatogonia against oxidative damage, chemotherapy drugs, environmental radiation, toxic substances, hyperthermia, ischemia/reperfusion, diabetes-induced testicular damage, metal-induced testicular toxicity, improve sperm quality and it affects the testosterone secretion pathway by affecting Leydig cells. Therefore, the objective of this study is to investigate the biological effects of melatonin as a natural antioxidant on testicles and their disorders.
Collapse
Affiliation(s)
- Somayeh Heidarizadi
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Rashidi
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Cyrus Jalili
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran.,Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammadreza Gholami
- Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
5
|
Jiang S, Li X, Zhang F, Mao J, Cao M, Zhang X, Huang S, Duan X, Shen J. Manganese Dioxide-Based Nanocarrier Delivers Paclitaxel to Enhance Chemotherapy against Orthotopic Glioma through Hypoxia Relief. SMALL METHODS 2022; 6:e2101531. [PMID: 35587180 DOI: 10.1002/smtd.202101531] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Chemotherapy plays an important role in treating cancers in clinic. Hypoxia-mediated chemoresistance remains a major hurdle for effective tumor chemotherapy. Herein, a new class of tLyP-1-modified dopamine (DOPA)-β-cyclodextrin (CD)-coated paclitaxel (PTX)- and manganese dioxide (MnO2 )-loaded nanoparticles (tLyP-1-CD-DOPA-MnO2 @PTX) is developed to enhance glioma chemotherapy. The nanomedicine delivered to the tumor site decomposes in response to the weak acidity and high hydrogen peroxide in the tumor microenvironment (TME), resulting in collapse of the system to release PTX and generates Mn2+ and O2 . In a rat model of intracranial glioma, tLyP-1-CD-DOPA-MnO2 @PTX can efficiently pass through the blood-brain-barrier to accumulate in tumor sites. The hypoxia in TME can be relieved via O2 generated by MnO2 and the reactive oxygen species produced by Mn2+ can kill tumor cells. The tLyP-1-CD-DOPA-MnO2 @PTX nanoparticles exert a remarkable antitumor effect by promoting apoptosis and inhibiting proliferation of tumor cells in addition to enabling real-time tumor monitoring with magnetic resonance imaging. This MnO2 -based theranostic medicine will offer a novel strategy to simultaneously enhance chemotherapy and achieve real-time imaging of therapeutic process in glioma treatment.
Collapse
Affiliation(s)
- Shuqi Jiang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Department of Radiology, the First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
| | - Xiaohu Li
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Fang Zhang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Jiaji Mao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Minghui Cao
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Xinna Zhang
- Department of Radiology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Siming Huang
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xiaohui Duan
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| | - Jun Shen
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangzhou, 510120, China
| |
Collapse
|
6
|
Aboelwafa H, Ramadan R, El-Kott A, Abdelhamid F. The protective effect of melatonin supplementation against taxol-induced testicular cytotoxicity in adult rats. Braz J Med Biol Res 2022; 55:e11614. [PMID: 35137851 PMCID: PMC8851920 DOI: 10.1590/1414-431x2021e11614] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 09/30/2021] [Indexed: 01/22/2023] Open
Affiliation(s)
| | | | - A.F. El-Kott
- King Khalid University, Saudi Arabia; College of Science, Damanhour University, Egypt
| | | |
Collapse
|
7
|
Raza F, Zafar H, Khan MW, Ullah A, Khan AU, Baseer A, Fareed R, Sohail M. Recent advances in the targeted delivery of paclitaxel nanomedicine for cancer therapy. MATERIALS ADVANCES 2022; 3:2268-2290. [DOI: 10.1039/d1ma00961c] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Cancer cases have reached an all-time high in the current era.
Collapse
Affiliation(s)
- Faisal Raza
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | - Hajra Zafar
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, P. R. China
| | | | - Aftab Ullah
- Department of Pharmacy, Shantou University Medical College, Shantou, 515041, P. R. China
| | | | - Abdul Baseer
- Department of Pharmacy, Abasyn University, Peshawar, Pakistan
| | - Rameesha Fareed
- Riphah Institute of Pharmaceutical Sciences, Riphah International University Islamabad, Pakistan
| | - Muhammad Sohail
- School of Pharmacy, Yantai University, Shandong, 264005, China
| |
Collapse
|
8
|
Hao J, Wang J, Pan H, Sang Y, Wang D, Wang Z, Ai J, Lin B, Chen L. pH-redox responsive polymer-doxorubicin prodrug micelles studied by molecular dynamics, dissipative particle dynamics simulations and experiments. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
9
|
Khing TM, Choi WS, Kim DM, Po WW, Thein W, Shin CY, Sohn UD. The effect of paclitaxel on apoptosis, autophagy and mitotic catastrophe in AGS cells. Sci Rep 2021; 11:23490. [PMID: 34873207 PMCID: PMC8648765 DOI: 10.1038/s41598-021-02503-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 11/17/2021] [Indexed: 12/20/2022] Open
Abstract
Paclitaxel is an anti-microtubule agent that has been shown to induce cell death in gastric cancer. However, the detailed mechanism of action is unclear. In this study, we reveal that the paclitaxel-induced cell death mechanism involves mitotic catastrophe, autophagy and apoptosis in AGS cells. Paclitaxel induced intrinsic apoptosis by activating caspase-3, caspase-9 and PARP. In addition, the significant increase in autophagy marker LC3B-II, together with Atg5, class III PI3K and Beclin-1, and the down-regulation of p62 following paclitaxel treatment verified that paclitaxel induced autophagy. Further experiments showed that paclitaxel caused mitotic catastrophe, cell cycle arrest of the accumulated multinucleated giant cells at the G2/M phase and induction of cell death in 24 h. Within 48 h, the arrested multinucleated cells escaped mitosis by decreasing cell division regulatory proteins and triggered cell death. Cells treated with paclitaxel for 48 h were grown in fresh medium for 24 h and checked for CDC2, CDC25C and lamin B1 protein expressions. These proteins had decreased significantly, indicating that the remaining cells became senescent. In conclusion, it is suggested that paclitaxel-induced mitotic catastrophe is an integral part of the cell death mechanism, in addition to apoptosis and autophagy, in AGS cells.
Collapse
Affiliation(s)
- Tin Myo Khing
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Won Seok Choi
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Dong Min Kim
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Wah Wah Po
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Wynn Thein
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Chang Yell Shin
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Uy Dong Sohn
- Laboratory of Signaling and Pharmacological Activity, Department of Pharmacology, College of Pharmacy, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
10
|
Recent advances in peptide-targeted micelleplexes: Current developments and future perspectives. Int J Pharm 2021; 597:120362. [PMID: 33556489 DOI: 10.1016/j.ijpharm.2021.120362] [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: 12/12/2020] [Revised: 01/27/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022]
Abstract
The decoding of the human genome revolutionized the understanding of how genetics influence the interplay between health and disease, in a multidisciplinary perspective. Thus, the development of exogenous nucleic acids-based therapies has increased to overcome hereditary or acquired genetic-associated diseases. Gene drug delivery using non-viral systems, for instance micelleplexes, have been recognized as promising options for gene-target therapies. Micelleplexes are core-shell structures, at a nanometric scale, designed using amphiphilic block copolymers. These can self-assemble in an aqueous medium, leading to the formation of a hydrophilic and positively charged corona - that can transport nucleic acids, - and a hydrophobic core - which can transport poor water-soluble drugs. However, the performance of these types of carriers usually is hindered by several in vivo barriers. Fortunately, due to a significant amount of research, strategies to overcome these shortcomings emerged. With a wide range of structural features, good stability against proteolytic degradation, affordable characteristic, easy synthesis, low immunogenicity, among other advantages, peptides have increasingly gained popularity as target ligands for non-viral carriers. Hence, this review addresses the use of peptides with micelleplexes illustrating, through the analysis of in vitro and in vivo studies, the potential and future perspectives of this combination.
Collapse
|
11
|
Lu Y, Zhang P, Zhang Q, Yang C, Qian Y, Suo J, Tao X, Zhu J. Duloxetine Attenuates Paclitaxel-Induced Peripheral Nerve Injury by Inhibiting p53-Related Pathways. J Pharmacol Exp Ther 2020; 373:453-462. [PMID: 32238452 DOI: 10.1124/jpet.120.265082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 03/18/2020] [Indexed: 03/08/2025] Open
Abstract
Paclitaxel (PTX) is an antineoplastic drug extracted from the Taxus species, and peripheral neuropathy is a common side effect. Paclitaxel-induced peripheral neuropathy (PIPN) seriously affects patient quality of life. Currently, the mechanism of PIPN is still unknown, and few treatments are recognized clinically. Duloxetine is recommended as the only potential treatment of chemotherapy-induced peripheral neuropathy (CIPN) by the American Society of Clinical Oncology. However, this guidance lacks a theoretical basis and experimental evidence. Our study suggested that duloxetine could improve PIPN and provide neuroprotection. We explored the potential mechanisms of duloxetine on PIPN. As a result, duloxetine acts by inhibiting poly ADP-ribose polymerase cleavage (PARP) and tumor suppressor gene p53 activation and regulating apoptosis regulator the Bcl2 family to reverse PTX-induced oxidative stress and apoptosis. Taken together, the present study shows that using duloxetine to attenuate PTX-induced peripheral nerve injury and peripheral pain may provide new clinical therapeutic targets for CIPN. SIGNIFICANCE STATEMENT: This study reported that duloxetine significantly alleviates neuropathic pain induced by paclitaxel and is related to poly ADP-ribose polymerase (PARP), tumor suppressor gene p53, and apoptosis regulator the Bcl2 family. Our findings thus not only provide important guidance to support duloxetine to become the first standard chemotherapy-induced peripheral neuropathy (CIPN) drug but also will find potential new targets and positive control for new CIPN drug development.
Collapse
Affiliation(s)
- Yuting Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Peng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Qiuyan Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Chao Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Yangyan Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Jinshuai Suo
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Xinxia Tao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| | - Jing Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Department of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China (Y.L., P.Z., Q.Z., C.Y., Y.Q., J.S., X.T., J.Z.); Departments of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland (J.Z.)
| |
Collapse
|
12
|
|
13
|
Li Y, Du L, Wu C, Yu B, Zhang H, An F. Peptide Sequence-Dominated Enzyme-Responsive Nanoplatform for Anticancer Drug Delivery. Curr Top Med Chem 2019; 19:74-97. [PMID: 30686257 DOI: 10.2174/1568026619666190125144621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/06/2018] [Accepted: 11/23/2018] [Indexed: 02/08/2023]
Abstract
Enzymatic dysregulation in tumor and intracellular microenvironments has made this property
a tremendously promising responsive element for efficient diagnostics, carrier targeting, and drug
release. When combined with nanotechnology, enzyme-responsive drug delivery systems (DDSs) have
achieved substantial advancements. In the first part of this tutorial review, changes in tumor and intracellular
microenvironmental factors, particularly the enzymatic index, are described. Subsequently, the
peptide sequences of various enzyme-triggered nanomaterials are summarized for their uses in various
drug delivery applications. Then, some other enzyme responsive nanostructures are discussed. Finally,
the future opportunities and challenges are discussed. In brief, this review can provide inspiration and
impetus for exploiting more promising internal enzyme stimuli-responsive nanoDDSs for targeted tumor
diagnosis and treatment.
Collapse
Affiliation(s)
- Yanan Li
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Liping Du
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Chunsheng Wu
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Hui Zhang
- First Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Feifei An
- Institute of Medical Engineering, Department of Biophysics, School of Basic Medical Science, Health Science Center, Xi’an Jiaotong University, No.76 Yanta West Road, Xi'an, Shaanxi 710061, China
| |
Collapse
|
14
|
Harini L, Srivastava S, Gnanakumar GP, Karthikeyan B, Ross C, Krishnakumar V, Kannan VR, Sundar K, Kathiresan T. An ingenious non-spherical mesoporous silica nanoparticle cargo with curcumin induces mitochondria-mediated apoptosis in breast cancer (MCF-7) cells. Oncotarget 2019; 10:1193-1208. [PMID: 30838091 PMCID: PMC6383822 DOI: 10.18632/oncotarget.26623] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 01/12/2019] [Indexed: 12/18/2022] Open
Abstract
Curcumin delivery to cancer cells is challenging due to its hydrophobic nature, low bio distribution and low availability. Many nano vehicles suffer from low stability and toxicity, and hence the prerequisite of a non-toxic nano vehicle with effective drug delivery is still being delved. The present study investigates the delivery efficiency of curcumin with non-spherical mesoporous silica nanoparticles (MSNAs). Their mechanism of drug delivery and signalling proteins activated to induce apoptosis was further explored in MCF-7 cells. A non-spherical MSN was synthesised, functionalised with PEI (MSNAP) and analysed its intracellular behaviour. Our result indicates that MSNAP was non-toxic until 20 µg/mL and likely localizes in cytoplasmic vesicles. On contrast, well-known MCM-41P induced autophagosome formation, indicating cellular toxicity. Curcumin was loaded on MSNAP and its effectiveness in inducing cell death was studied in MCF-7 and in MCF-7R cells. Curcumin loading on MSNAP induces better cell death with 30 µM curcumin, better than unbounded curcumin. Western blot analysis suggest, curcumin induce apoptosis through the activation of caspase 9, 6, 12, PARP, CHOP and PTEN. The cell survival protein Akt1 was downregulated by curcumin with and without the nanostructure. Interestingly, cleaved caspase 9 was activated in higher amount in nano-conjugated curcumin compared to the free curcumin. But other ER resident protein like IRE1α, PERK and GRP78 were downregulated indicating curcumin disturbs ER homeostasis. Further, electron microscopic analysis reveled that nanocurcumin induced apoptosis by disrupting mitochondria and nucleus. Our results with doxorubicin resistant MCF-7 cell lines confirm nanodelivery of doxorubicin and curcumin sensitised cells effectively at lesser concentration. Further docking studies of curcumin indicate it interacts with the apoptotic proteins through hydrogen bonding formation and with higher binding energy.
Collapse
Affiliation(s)
| | - Sweta Srivastava
- Department of Translation Medicine, St. Johns National Academy of Health Sciences, Bangalore, Karnataka, India
| | | | - Bose Karthikeyan
- Department of Biotechnology, Kalasalingam University, Krishnankoil, Tamil Nadu, India
- Oregon Health and Science University, Knight Cardiovascular Institute (KCVI), Portland, Oregon, USA
| | - Cecil Ross
- Department of Medicine, St. Johns National Academy of Health Sciences, Bangalore, Karnataka, India
| | | | - Velu Rajesh Kannan
- Department of Microbiology, Bharathidasan University, Tiruchirappalli, Tamil Nadu, India
| | - Krishnan Sundar
- Department of Biotechnology, Kalasalingam University, Krishnankoil, Tamil Nadu, India
- International Research Centre, Kalasalingam University, Krishnankoil, Tamil Nadu, India
| | - Thandavarayan Kathiresan
- Department of Biotechnology, Kalasalingam University, Krishnankoil, Tamil Nadu, India
- International Research Centre, Kalasalingam University, Krishnankoil, Tamil Nadu, India
| |
Collapse
|
15
|
Basic principles of drug delivery systems - the case of paclitaxel. Adv Colloid Interface Sci 2019; 263:95-130. [PMID: 30530177 DOI: 10.1016/j.cis.2018.11.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 11/15/2018] [Accepted: 11/15/2018] [Indexed: 01/15/2023]
Abstract
Cancer is the second cause of death worldwide, exceeded only by cardiovascular diseases. The prevalent treatment currently used against metastatic cancer is chemotherapy. Among the most studied drugs that inhibit neoplastic cells from acquiring unlimited replicative ability (a hallmark of cancer) are the taxanes. They operate via a unique molecular mechanism affecting mitosis. In this review, we show this mechanism for one of them, paclitaxel, and for other (non-taxanes) anti-mitotic drugs. However, the use of paclitaxel is seriously limited (its bioavailability is <10%) due to several long-standing challenges: its poor water solubility (0.3 μg/mL), its being a substrate for the efflux multidrug transporter P-gp, and, in the case of oral delivery, its first-pass metabolism by certain enzymes. Adequate delivery methods are therefore required to enhance the anti-tumor activity of paclitaxel. Thus, we have also reviewed drug delivery strategies in light of the various physical, chemical, and enzymatic obstacles facing the (especially oral) delivery of drugs in general and paclitaxel in particular. Among the powerful and versatile platforms that have been developed and achieved unprecedented opportunities as drug carriers, microemulsions might have great potential for this aim. This is due to properties such as thermodynamic stability (leading to long shelf-life), increased drug solubilization, and ease of preparation and administration. In this review, we define microemulsions and nanoemulsions, analyze their pertinent properties, and review the results of several drug delivery carriers based on these systems.
Collapse
|
16
|
Pautu V, Leonetti D, Lepeltier E, Clere N, Passirani C. Nanomedicine as a potent strategy in melanoma tumor microenvironment. Pharmacol Res 2017; 126:31-53. [PMID: 28223185 DOI: 10.1016/j.phrs.2017.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 12/19/2022]
Abstract
Melanoma originated from melanocytes is the most aggressive type of skin cancer. Despite considerable progresses in clinical treatment with the discovery of BRAF or MEK inhibitors and monoclonal antibodies, the durability of response to treatment is often limited to the development of acquired resistance and systemic toxicity. The limited success of conventional treatment highlights the importance of understanding the role of melanoma tumor microenvironment in tumor developement and drug resistance. Nanoparticles represent a promising strategy for the development of new cancer treatments able to improve the bioavailability of drugs and increase their penetration by targeting specifically tumors cells and/or tumor environment. In this review, we will discuss the main influence of tumor microenvironment in melanoma growth and treatment outcome. Furthermore, third generation loaded nanotechnologies represent an exciting tool for detection, treatment, and escape from possible mechanism of resistance mediated by tumor microenvironment, and will be highlighted in this review.
Collapse
Affiliation(s)
- Vincent Pautu
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | | | - Elise Lepeltier
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | - Nicolas Clere
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France
| | - Catherine Passirani
- MINT, UNIV Angers, INSERM, CNRS, Université Bretagne Loire, IBS-CHU, 4 rue Larrey, F-49933 Angers, France.
| |
Collapse
|