1
|
Rana R, Devi SN, Bhardwaj AK, Yashavarddhan MH, Bohra D, Ganguly NK. Exosomes as nature's nano carriers: Promising drug delivery tools and targeted therapy for glioma. Biomed Pharmacother 2025; 182:117754. [PMID: 39731936 DOI: 10.1016/j.biopha.2024.117754] [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: 09/09/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/30/2024] Open
Abstract
Exosomes, minute vesicles originating from diverse cell types, exhibit considerable potential as carriers for drug delivery in glioma therapy. These naturally occurring nanocarriers facilitate the transfer of proteins, RNAs, and lipids between cells, offering advantages such as biocompatibility, efficient cellular absorption, and the capability to traverse the blood-brain barrier (BBB). In the realm of cancer, particularly gliomas, exosomes play pivotal roles in modulating tumor growth, regulating immunity, and combating drug resistance. Moreover, exosomes serve as valuable biomarkers for diagnosing diseases and assessing prognosis. This review aims to elucidate the therapeutic and diagnostic promise of exosomes in glioma treatment, highlighting the innovative advances in exosome engineering that enable precise drug loading and targeting. By circumventing challenges associated with current glioma treatments, exosome-mediated drug delivery strategies can enhance the efficacy of chemotherapy drugs like temozolomide and overcome drug resistance mechanisms. This review underscores the multifaceted roles of exosomes in glioma pathogenesis and therapy, underscoring their potential as natural nanocarriers for targeted therapy and heralding a new era of hope for glioma treatment.
Collapse
Affiliation(s)
- Rashmi Rana
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India.
| | | | - Amit Kumar Bhardwaj
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - M H Yashavarddhan
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Deepika Bohra
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| | - Nirmal Kumar Ganguly
- Department of Biotechnology and Research, Sir Ganga Ram Hospital, New Delhi 110060, India
| |
Collapse
|
2
|
Liu HM, Zhang Y. Folic acid-decorated astrocytes-derived exosomes enhanced the effect of temozolomide against glioma. Kaohsiung J Med Sci 2024; 40:435-444. [PMID: 38482902 DOI: 10.1002/kjm2.12819] [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: 09/27/2023] [Revised: 01/19/2024] [Accepted: 02/06/2024] [Indexed: 05/12/2024] Open
Abstract
A direct strategy to achieve specific treatments and reduce side effects is through cell type-specific drug delivery. Exosomes (Exos) can be modified with folic acid (FA) to prepare drug delivery systems targeting tumor cells that highly express FA receptors. This study aimed to produce an exo drug delivery system with FA decoration and temozolomide (TMZ) loading to improve the sustained TMZ release and targeting. We used DSPE-PEG2000-FA to modify exos derived from astrocyte U-87 to prepare FA-modified exos (Astro-exo-FA). TMZ was encapsulated into Astro-exo-FA or Astro-exo through electroporation to produce TMZ@Astro-exo and TMZ@Astro-exo-FA. In vitro drug release was examined using the dialysis bag method. Through cell experiments in vitro and mouse glioma models in vivo, the effect of TMZ@Astro-exo-FA on U-87 cells was determined. Exo properties were not affected by FA modification and TMZ loading. The drug release rate of TMZ@Astro-exo-FA was slower. TMZ@Astro-exo-FA uptake by U-87 cells was higher compared to TMZ@Astro-exo, indicating that TMZ@Astro-exo-FA has a stronger targeting toward U-87 cells. TMZ@Astro-exo-FA remarkably reduced U-87 cell proliferation, migration, and invasion compared with TMZ@Astro-exo and free TMZ. Treatment with TMZ@Astro-exo-FA reduced the side effects of TMZ (minimal change in body weight), prolonged survival, and inhibited tumor growth in mouse glioma models, and its efficacy was stronger than that of TMZ@Astro-exo and free TMZ. TMZ@Astro-exo-FA could enhance the effect of TMZ against glioma, providing novel ideas for drug targeting delivery and exploring exos as drug carriers against glioma.
Collapse
Affiliation(s)
- Hong-Ming Liu
- Shandong Center For Food and Drug Evaluation & Inspection, Jinan, China
- Zibo Food and Drug Inspection and Research Institute, Zibo, China
| | - Ye Zhang
- Department of Pharmaceutical Science, Zibo Vocational Institute, Zibo, China
| |
Collapse
|
3
|
Li C, Wang L, Li Z, Li Z, Zhang K, Cao L, Wang Z, Shen C, Chen L. Repolarizing Tumor-Associated Macrophages and inducing immunogenic cell Death: A targeted liposomal strategy to boost cancer immunotherapy. Int J Pharm 2024; 651:123729. [PMID: 38142016 DOI: 10.1016/j.ijpharm.2023.123729] [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: 09/18/2023] [Revised: 11/26/2023] [Accepted: 12/20/2023] [Indexed: 12/25/2023]
Abstract
Cancer immunotherapy has shown promise in treating various malignancies. However, the presence of an immunosuppressive tumor microenvironment (TME) triggered by M2 tumor-associated macrophages (TAMs) and the limited tumor cell antigenicity have hindered its broader application. To address these challenges, we developed DOX/R837@ManL, a liposome loaded with imiquimod (R837) and doxorubicin (DOX), modified with mannose-polyethylene glycol (Man-PEG). DOX/R837@ManL employed a mannose receptor (MRC1)-mediated targeting strategy, allowing it to accumulate selectively at M2 Tumor associated macrophages (TAMs) and tumor sites. R837, an immune adjuvant, promoted the conversion of immunosuppressive M2 TAMs into immunostimulatory M1 TAMs, and reshaped the immunosuppressive TME. Simultaneously, DOX release induced immunogenic cell death (ICD) in tumor cells and enhanced tumor cell antigenicity by promoting dendritic cells (DCs) maturation. Through targeted delivery, the synergistic action of R837 and DOX activated innate immunity and coordinated adaptive immunity, enhancing immunotherapy efficacy. In vivo experiments have demonstrated that DOX/R837@ManL effectively eliminated primary tumors and lung metastases, while also preventing tumor recurrence post-surgery. These findings highlighted the potential of DOX/R837@ManL as a promising strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Cong Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lihong Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zhihang Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zehao Li
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Kexin Zhang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lianrui Cao
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Zeyu Wang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Chao Shen
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China
| | - Lijiang Chen
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, China.
| |
Collapse
|
4
|
Wang C, Yang Y, Cao Y, Liu K, Shi H, Guo X, Liu W, Hao R, Song H, Zhao R. Nanocarriers for the delivery of antibiotics into cells against intracellular bacterial infection. Biomater Sci 2023; 11:432-444. [PMID: 36503914 DOI: 10.1039/d2bm01489k] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The barrier function of host cells enables intracellular bacteria to evade the lethality of the host immune system and antibiotics, thereby causing chronic and recurrent infections that seriously threaten human health. Currently, the main clinical strategy for the treatment of intracellular bacterial infections involves the use of long-term and high-dose antibiotics. However, insufficient intracellular delivery of antibiotics along with various resistance mechanisms not only weakens the efficacy of current therapies but also causes serious adverse drug reactions, further increasing the disease and economic burden. Improving the delivery efficiency, intracellular accumulation, and action time of antibiotics remains the most economical and effective way to treat intracellular bacterial infections. The rapid development of nanotechnology provides a strategy to efficiently deliver antibiotics against intracellular bacterial infections into cells. In this review, we summarize the types of common intracellular pathogens, the difficulties faced by antibiotics in the treatment of intracellular bacterial infections, and the research progress of several types of representative nanocarriers for the delivery of antibiotics against intracellular bacterial infections that have emerged in recent years. This review is expected to provide a reference for further elucidating the intracellular transport mechanism of nanocarrier-drug complexes, designing safer and more effective nanocarriers and establishing new strategies against intracellular bacterial infection.
Collapse
Affiliation(s)
- Chao Wang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yi Yang
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Kaixin Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Hua Shi
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Xudong Guo
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Wanying Liu
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Hongbin Song
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| | - Rongtao Zhao
- Chinese PLA Center for Disease Control and Prevention, Beijing, 100071, China.
| |
Collapse
|
5
|
Zhou ZW, Long HZ, Xu SG, Li FJ, Cheng Y, Luo HY, Gao LC. Therapeutic Effects of Natural Products on Cervical Cancer: Based on Inflammatory Pathways. Front Pharmacol 2022; 13:899208. [PMID: 35645817 PMCID: PMC9136176 DOI: 10.3389/fphar.2022.899208] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 04/26/2022] [Indexed: 12/09/2022] Open
Abstract
Inflammation is a protective response of the body to an irritant. When an inflammatory response occurs, immune cells are recruited to the injury, eliminating the irritation. The excessive inflammatory response can cause harm to the organism. Inflammation has been found to contribute to cervical cancer if there is a problem with the regulation of inflammatory response. Cervical cancer is one of the most common malignant tumors globally, and the incidence tends to be younger. The harm of cervical cancer cannot be ignored. The standard treatments for cervical cancer include surgery, radiotherapy and chemotherapy. However, the prognosis for this treatment is poor, so it is urgent to find a safer and more effective treatment. Natural products are considered excellent candidates for the treatment of cervical cancer. In this review, we first describe the mechanisms by which inflammation induces cervical cancer. Subsequently, we highlight natural products that can treat cervical cancer through inflammatory pathways. We also introduce natural products for the treatment of cervical cancer in clinical trials. Finally, methods to improve the anticancer properties of natural products were added, and the development status of natural products was discussed.
Collapse
Affiliation(s)
- Zi-Wei Zhou
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Hui-Zhi Long
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Shuo-Guo Xu
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Feng-Jiao Li
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Yan Cheng
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Hong-Yu Luo
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| | - Li-Chen Gao
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Changsha, China
| |
Collapse
|
6
|
Shakerzadeh E, Zborowski KK, Chigo Anota E, Nguyen MT. Pristine and alkali and alkaline earth metals encapsulated B
36
N
36
nanoclusters as prospective delivery agents and detectors for 5‐fluorouracil anticancer drug. Appl Organomet Chem 2022. [DOI: 10.1002/aoc.6721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Ehsan Shakerzadeh
- Chemistry Department, Faculty of Science Shahid Chamran University of Ahvaz Ahvaz Iran
| | - Krzysztof K. Zborowski
- Department of General Chemistry, Faculty of Chemistry Jagiellonian University Kraków Poland
| | - Ernesto Chigo Anota
- Benemérita Universidad Autónoma de Puebla, Facultad de Ingeniería Química, Ciudad Universitaria, San Manuel Puebla México
| | - Minh Tho Nguyen
- Institute for Computational Science and Technology (ICST) Ho Chi Minh City Vietnam
| |
Collapse
|
7
|
Liu Q, Song P, Zhang W, Wang Z, Yang K, Luo J, Zhu L, Gui L, Tao Y, Ge F. Acid-Sensitive Nanoparticles Based on Molybdenum Disulfide for Photothermal-Chemo Therapy. ACS Biomater Sci Eng 2022; 8:1706-1716. [PMID: 35291764 DOI: 10.1021/acsbiomaterials.1c01390] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The combination of multiple treatments has recently been investigated for tumor treatment. In this study, molybdenum disulfide (MoS2) with excellent photothermal conversion performance was used as the core, and manganese dioxide (MnO2), which responds to the tumor microenvironment, was loaded on its surface by liquid deposition to form a mesoporous core-shell structure. Then, the chemotherapeutic drug Adriamycin (DOX) was loaded into the hole. To further enhance its water solubility and stability, the surface of MnO2 was modified with mPEG-NH2 to prepare the combined antitumor nanocomposite MoS2@DOX/MnO2-PEG (MDMP). The results showed that MDMP had a diameter of about 236 nm, its photothermal conversion efficiency was 33.7%, and the loading and release rates of DOX were 13 and 65%, respectively. During in vivo and in vitro studies, MDMP showed excellent antitumor activity. Under the combined treatment, the tumor cell viability rate was only 11.8%. This nanocomposite exhibits considerable potential for chemo-photothermal combined antitumor therapy.
Collapse
Affiliation(s)
- Qin Liu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Ping Song
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Weiwei Zhang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Zhenyu Wang
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Kai Yang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection & School for Radiological and Interdisciplinary Sciences (RAD-X), Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou 215123, Jiangsu, China
| | - Jianquan Luo
- Institute of Process Engineering, University of Chinese Academy of Sciences, Beijing 100049, People's Republic of China
| | - Longbao Zhu
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Lin Gui
- Department of Microbiology and Immunology, Wannan Medical College, Wuhu, Anhui 241002, Peoples Republic of China
| | - Yugui Tao
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| | - Fei Ge
- School of Biological and Food Engineering, Anhui Polytechnic University, Wuhu, Anhui 241000, People's Republic of China
| |
Collapse
|
8
|
Liu M, Chen X, Chen H, Wu X, Fan W, Chen J. Nanotechnology-Based Drug Delivery System for Anticancer Active Ingredients from Traditional Chinese Medicines: A Review. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:2011-2032. [DOI: 10.1142/s0192415x22500860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The variable dosage forms of most traditional Chinese medicines (TCMs) could be disadvantaged by low selectivity, poor biological distribution, limited bioavailability with low efficacy, and some adverse effects. These issues limit the control of clinical pharmacodynamics of the antitumor active components. With the progress of science and technology, many new polymer materials and new technologies have emerged, such as nanotechnology, cyclodextrin inclusion, solid dispersion, microcapsule and microsphere technologies. These new technologies provide a good basis for exploring novel TCM dosage forms for overcoming the shortcomings. The increased numbers of new technologies have been used to study TCM dosage forms with remarkable achievements. In this review paper, we will provide a systematic overview of the new dosage forms of nano-formulations and co-medications in relation to nano-delivery systems in an attempt to provide useful references for practical application of active antitumor ingredients from the TCMs.
Collapse
Affiliation(s)
- Mengmeng Liu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P. R. China
| | - Xinmei Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P. R. China
| | - Hang Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P. R. China
| | - Xin Wu
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P. R. China
- Shanghai Wei Er Lab, Shanghai 200137, P. R. China
| | - Wei Fan
- Seventh People’s Hospital of Shanghai, University of Traditional Chinese Medicine, Shanghai 200137, P. R. China
| | - Jianming Chen
- Department of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P. R. China
- Shanghai Wei Er Lab, Shanghai 200137, P. R. China
| |
Collapse
|
9
|
Meng Q, Zhong S, He S, Gao Y, Cui X. Synthesis and characterization of curcumin-loaded pH/reduction dual-responsive folic acid modified carboxymethyl cellulose-based microcapsules for targeted drug delivery. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.09.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
10
|
Handa M, Beg S, Shukla R, Barkat MA, Choudhry H, Singh KK. Recent advances in lipid-engineered multifunctional nanophytomedicines for cancer targeting. J Control Release 2021; 340:48-59. [PMID: 34695523 DOI: 10.1016/j.jconrel.2021.10.025] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 12/12/2022]
Abstract
Cancer is a leading cause of death in many countries around the world. However, the efficacy of current treatments available for variety of cancers is considered to be suboptimal due to the pathophysiological challenges associated with the disease which limits the efficacy of the anticancer drugs. Moreover, the vulnerability towards off-target effects and high toxicity also limits the use of drugs for the treatment of cancers. Besides, the biopharmaceutical challenges like poor water solubility and permeability of the drugs, along with the absence of active targeting capability further decreases the utility of drugs in cancer therapy. As a result of these deficiencies, the current therapeutic strategies face noncompliance to patients for providing meaningful benefits after administration. With the advancements in nanotechnology, there has been a paradigm shift in the modalities for cancer treatment with the help of phytomedicine-based nanosized drug delivery systems coupled with variegated surface-engineering strategies for targeted drug delivery. Among these delivery systems, lipid-based nanoparticles are considered as one of the highly biocompatible, efficient and effective systems extensively explored for anticancer drug delivery. These include diverse range of systems including liposomes, nanoemulsions, solid lipid nanoparticles, nanostructured lipidic carriers and supramolecular carriers, which alters pharmacokinetic and biodistribution of the drugs for active targeting to the desired site of action by overcoming the biopharmaceutical challenges associated with anticancer drug delivery. The present review endeavours to provide a comprehensive account on the recent advances in the application of lipid-based nanostructured systems for improving the pharmacotherapeutic performance of phytomedicines for cancer targeting application.
Collapse
Affiliation(s)
- Mayank Handa
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India
| | - Sarwar Beg
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK; Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard University, New Delhi 110062, India.
| | - Rahul Shukla
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research-Raebareli, Lucknow 226002, U.P., India.
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, Al Jamiah, Hafr Al Batin 39524, Saudi Arabia
| | - Hani Choudhry
- Department of Biochemistry, Cancer Metabolism & Epigenetic Unit, Faculty of Science, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Kamalinder K Singh
- School of Pharmacy and Biomedical Sciences, Faculty of Clinical and Biomedical Sciences, University of Central Lancashire, Preston PR1 2HE, UK.
| |
Collapse
|
11
|
Ju G, Zhu Y, Du T, Cao W, Lin J, Li C, Xu D, Wang Z. MiR-197 Inhibitor Loaded AbCD133@MSNs@GNR Affects the Development of Prostate Cancer Through Targeting ITGAV. Front Cell Dev Biol 2021; 9:646884. [PMID: 34195187 PMCID: PMC8238009 DOI: 10.3389/fcell.2021.646884] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 04/27/2021] [Indexed: 12/25/2022] Open
Abstract
Prostate cancer is one of the most severe male malignant tumors, which ranks second in mortality rate among all tumors. Traditional methods of treatment for prostate cancer produce obvious side effects and a high recurrence rate. Cancer stem cells are considered to be a group of cells that determine the proliferation, metastasis, and drug resistance of tumor. Prostate cancer therapy based on microRNAs and prostate cancer stem cells (PCSCs) has been a research hot spot in this field. Previous studies have reported that miR-197 plays an important role in the occurrence and development of prostate cancer, but the molecular mechanism of miR-197 on the development of prostate cancer has not been reported yet. In this study, we verified that miR-197 is significantly overexpressed in prostate cancer tissues and prostate cancer cells. Then, we verified that miR-197 expression affects the proliferation, invasion, and metastasis of prostate cancer cells by regulating integrin subunit alpha V (ITGAV) expression through STAT5 pathway, and the results indicated that the miR-197 inhibitor can be a prostate cancer suppressor. Then we synthesized the AbCD133@GNR@MSNs@miR-197 inhibitor drug carrier, in which 35.42 μg of the miR-197 inhibitor could be loaded in 1 mg of AbCD133@GNR@MSNs. The AbCD133@GNR@MSNs@miR-197 inhibitor demonstrated good photothermal properties and photothermal controlled-release properties. The modified CD133 antibodies on the surface of the nano drug carrier helped more drug carriers to enter the PCSCs. The pharmacodynamic effects of the AbCD133@GNR@MSNs@miR-197 inhibitor on PCSCs in vivo and in vitro were studied under near-infrared radiation. The results showed that the AbCD133@GNR@MSNs@miR-197 inhibitor prepared in this study could not only significantly suppress the development of PCSCs through ITGAV/STAT5 pathway but also significantly suppress the growth of PCSC solid tumors. In short, our study verified that miR-197 regulates the development of PCSCs through STAT5 pathway by targeting ITGAV, and the AbCD133@MSNs@GNR@miR-197 inhibitor could be a potential suppressor used in prostate cancer treatment. In short, our study found that miR-197 affected the development of prostate cancer by regulating ITGAV. The AbCD133@GNR@MSNs@miR-197 inhibitor prepared in this study could suppress the development and growth of PCSCs in vitro and in solid tumors not only by targeting the ITGAV but also through photothermal therapy. Our study not only provides a theoretical basis for the clinical treatment of prostate cancer but also provides a research scheme of drug loading and microRNA-based photothermal controlled therapy for prostate cancer.
Collapse
Affiliation(s)
- Guanqun Ju
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yingjian Zhu
- Department of Urology, Shanghai Jiangqiao Hospital, Shanghai General Hospital Jiading Branch, Shanghai, China
| | - Tao Du
- Department of Urology, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wanli Cao
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianhai Lin
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Chun Li
- Key Laboratory of Functional Genomic and Molecular Diagnosis of Gansu Province, Lanzhou, China
| | - Dongliang Xu
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China.,Urology Centre, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhijun Wang
- Department of Urology, Changzheng Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|