1
|
Gralewska P, Gajek A, Marczak A, Rogalska A. Targeted Nanocarrier-Based Drug Delivery Strategies for Improving the Therapeutic Efficacy of PARP Inhibitors against Ovarian Cancer. Int J Mol Sci 2024; 25:8304. [PMID: 39125873 PMCID: PMC11312858 DOI: 10.3390/ijms25158304] [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: 06/19/2024] [Revised: 07/20/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
The current focus of ovarian cancer (OC) research is the improvement of treatment options through maximising drug effectiveness. OC remains the fifth leading cause of cancer-induced mortality in women worldwide. In recent years, nanotechnology has revolutionised drug delivery systems. Nanoparticles may be utilised as carriers in gene therapy or to overcome the problem of drug resistance in tumours by limiting the number of free drugs in circulation and thereby minimising undesired adverse effects. Cell surface receptors, such as human epidermal growth factor 2 (HER2), folic acid (FA) receptors, CD44 (also referred to as homing cell adhesion molecule, HCAM), and vascular endothelial growth factor (VEGF) are highly expressed in ovarian cancer cells. Generation of active targeting nanoparticles involves modification with ligands that recognise cell surface receptors and thereby promote internalisation by cancer cells. Several poly(ADP-ribose) polymerase (PARP) inhibitors (PARPi) are currently used for the treatment of high-grade serous ovarian carcinomas (HGSOC) or platinum-sensitive relapsed OC. However, PARP resistance and poor drug bioavailability are common challenges, highlighting the urgent need to develop novel, effective strategies for ovarian cancer treatment. This review evaluates the utility of nanoparticles in ovarian cancer therapy, with a specific focus on targeted approaches and the use of PARPi nanocarriers to optimise treatment outcomes.
Collapse
Affiliation(s)
| | | | | | - Aneta Rogalska
- Department of Medical Biophysics, Institute of Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90–236 Lodz, Poland; (P.G.); (A.G.); (A.M.)
| |
Collapse
|
2
|
Liu J, Zhang W, Jin S, Zhang H, Xu Y, Xiong P, Qin X, Jia B. Plant-derived inducers in tumor differentiation therapy:A systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 130:155749. [PMID: 38763009 DOI: 10.1016/j.phymed.2024.155749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 02/27/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
BACKGROUND Differentiation therapy, a highly regarded treatment method in tumor research, aims to induce tumor cells to differentiate back to normal cells, deviating from the malignant pathway and returning to a benign state. Its development relies on the continuous discovery of efficient and low-toxic differentiation inducers, including plant-derived active components that offer significant biological utilization and therapeutic potential. For this reason, the exploration of plant-derived inducers, particularly in their application in differentiation therapy, holds great promise in advancing cancer treatment strategies toward more effective and safer alternatives. PURPOSE This paper aims to provide a valuable reference for researchers seeking to identify natural, efficient, and low-toxic differentiation inducers from plants and highlights a promising research direction for the application of differentiation therapy in malignant tumor treatment. METHODS For the collection of pertinent information, an extensive search was conducted across diverse literature and electronic databases, including PubMed, ScienceDirect, Wiley, ACS, CNKI, Springer, Taylor & Francis, Web of Science, Google Scholar, and Baidu Scholar. This comprehensive approach aimed to retrieve and include all relevant literature from 1985 to 2023. Primary keywords such as "Natural medicinal plant," "Differentiation therapy," and "Differentiation inducer" were utilized, supplemented by secondary search terms including "Cancer," "Tumor," "Herbal medicine," "Induced differentiation," and "Cancer treatment." RESULTS This study systematically evaluated the application of plant-derived inducers in tumor-induced differentiation therapy. Through extensive literature review, specific plant components with confirmed differentiation-inducing properties were identified. Furthermore, potential molecular mechanisms underlying this process were outlined, shedding light on the future development of differentiation therapy in cancer treatment. CONCLUSION Plant-derived active components exhibit substantial biological utility and therapeutic potential. Delving deeper into the research on these components as differentiation inducers holds promise for the selection of novel cancer drugs and the unveiling of novel pathways for cancer treatment. These results emphasize the importance of continued exploration and in-depth research into natural, efficient, and low-toxic differentiation inducers from plants, which could significantly advance cancer treatment strategies. Moreover, the highlighted research direction underscores the relevance of differentiation therapy in the context of malignant tumor treatment, indicating its potential as a safer and more effective alternative in cancer therapy.
Collapse
Affiliation(s)
- Junyu Liu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Wei Zhang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Shenrui Jin
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Hua Zhang
- Nanbu Hospital of County Chinese Medicine, Nanchong, Sichuan, 637399, China
| | - Yi Xu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 102488, China
| | - Peiyu Xiong
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China
| | - Xuhua Qin
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| | - Bo Jia
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 611137, China.
| |
Collapse
|
3
|
Wang L, Liu J, Wang X, Li X, Zhang X, Yuan L, Wu Y, Liu M. Effect of the combined binding of topotecan and catechin/protocatechuic acid to a pH-sensitive DNA tetrahedron on release and cytotoxicity: Spectroscopic and calorimetric studies. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2024; 314:124179. [PMID: 38522375 DOI: 10.1016/j.saa.2024.124179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/16/2024] [Accepted: 03/18/2024] [Indexed: 03/26/2024]
Abstract
The therapeutic efficacy of chemotherapy drugs can be effectively improved through the dual effects of their combination with natural polyphenols and the delivery of targeted DNA nanostructures. In this work, the interactions of topotecan (TPT), (+)-catechin (CAT), or protocatechuic acid (PCA) with a pH-sensitive DNA tetrahedron (MUC1-TD) in the binary and ternary systems at pHs 5.0 and 7.4 were investigated by fluorescence spectroscopy and calorimetry. The intercalative binding mode of TPT/CAT/PC to MUC1-TD was confirmed, and their affinity was ranked in the order of PCA > CAT > TPT. The effects of the pH-sensitivity of MUC1-TD and different molecular structures of CAT and PCA on the loading, release, and cytotoxicity of TPT were discussed. The weakened interaction under acidic conditions and the co-loading of CAT/PCA, especially PCA, improved the release of TPT loaded by MUC1-TD. The targeting of MUC1-TD and the synergistic effect with CAT/PCA, especially CAT, enhanced the cytotoxicity of TPT on A549 cells. For L02 cells, the protective effect of CAT/PCA reduced the damage caused by TPT. The single or combined TPT loaded by MUC1-TD was mainly concentrated in the nucleus of A549 cells. This work will provide key information for the combined application of TPT and CAT/PCA loaded by DNA nanostructures to improve chemotherapy efficacy and reduce side effects.
Collapse
Affiliation(s)
- Lu Wang
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China
| | - Jie Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Xiangtai Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Xinyu Li
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China
| | - Xinpeng Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Lixia Yuan
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Yushu Wu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China
| | - Min Liu
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, China; Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng 252059, China.
| |
Collapse
|
4
|
Oryani MA, Nosrati S, Javid H, Mehri A, Hashemzadeh A, Karimi-Shahri M. Targeted cancer treatment using folate-conjugated sponge-like ZIF-8 nanoparticles: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1377-1404. [PMID: 37715816 DOI: 10.1007/s00210-023-02707-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/02/2023] [Indexed: 09/18/2023]
Abstract
ZIF-8 (zeolitic imidazolate framework-8) is a potential drug delivery system because of its unique properties, which include a large surface area, a large pore capacity, a large loading capacity, and outstanding stability under physiological conditions. ZIF-8 nanoparticles may be readily functionalized with targeting ligands for the identification and absorption of particular cancer cells, enhancing the efficacy of chemotherapeutic medicines and reducing adverse effects. ZIF-8 is also pH-responsive, allowing medication release in the acidic milieu of cancer cells. Because of its tunable structure, it can be easily functionalized to design cancer-specific targeted medicines. The delivery of ZIF-8 to cancer cells can be facilitated by folic acid-conjugation. Hence, it can bind to overexpressed folate receptors on the surface of cancer cells, which holds the promise of reducing unwanted deliveries. As a result of its importance in cancer treatment, the folate-conjugated ZIF-8 was the major focus of this review.
Collapse
Affiliation(s)
- Mahsa Akbari Oryani
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shamim Nosrati
- Department of Clinical Biochemistry, Faculty of Medicine, Azad Shahroud University, Shahroud, Iran
| | - Hossein Javid
- Department of Medical Laboratory Sciences, Varastegan Institute for Medical Sciences, Mashhad, Iran.
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Ali Mehri
- Endoscopic and Minimally Invasive Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alireza Hashemzadeh
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehdi Karimi-Shahri
- Department of Pathology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Pathology, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
5
|
Kumbhar PR, Kumar P, Lasure A, Velayutham R, Mandal D. An updated landscape on nanotechnology-based drug delivery, immunotherapy, vaccinations, imaging, and biomarker detections for cancers: recent trends and future directions with clinical success. DISCOVER NANO 2023; 18:156. [PMID: 38112935 PMCID: PMC10730792 DOI: 10.1186/s11671-023-03913-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 10/20/2023] [Indexed: 12/21/2023]
Abstract
The recent development of nanotechnology-based formulations improved the diagnostics and therapies for various diseases including cancer where lack of specificity, high cytotoxicity with various side effects, poor biocompatibility, and increasing cases of multi-drug resistance are the major limitations of existing chemotherapy. Nanoparticle-based drug delivery enhances the stability and bioavailability of many drugs, thereby increasing tissue penetration and targeted delivery with improved efficacy against the tumour cells. Easy surface functionalization and encapsulation properties allow various antigens and tumour cell lysates to be delivered in the form of nanovaccines with improved immune response. The nanoparticles (NPs) due to their smaller size and associated optical, physical, and mechanical properties have evolved as biosensors with high sensitivity and specificity for the detection of various markers including nucleic acids, protein/antigens, small metabolites, etc. This review gives, initially, a concise update on drug delivery using different nanoscale platforms like liposomes, dendrimers, polymeric & various metallic NPs, hydrogels, microneedles, nanofibres, nanoemulsions, etc. Drug delivery with recent technologies like quantum dots (QDs), carbon nanotubes (CNTs), protein, and upconverting NPs was updated, thereafter. We also summarized the recent progress in vaccination strategy, immunotherapy involving immune checkpoint inhibitors, and biomarker detection for various cancers based on nanoplatforms. At last, we gave a detailed picture of the current nanomedicines in clinical trials and their possible success along with the existing approved ones. In short, this review provides an updated complete landscape of applications of wide NP-based drug delivery, vaccinations, immunotherapy, biomarker detection & imaging for various cancers with a predicted future of nanomedicines that are in clinical trials.
Collapse
Affiliation(s)
- Pragati Ramesh Kumbhar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Hajipur, Hajipur, 844102, India
| | - Prakash Kumar
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Hajipur, Hajipur, 844102, India
| | - Aarti Lasure
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Hajipur, Hajipur, 844102, India
| | | | - Debabrata Mandal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research- Hajipur, Hajipur, 844102, India.
| |
Collapse
|
6
|
Harakeh S, Saber SH, Al-Raddadi R, Alamri T, Al-Jaouni S, Qari M, Qari Y, Haque S, Zawawi A, Ali SS, Elmageed ZYA, Mousa S. Novel curcumin nanoformulation induces apoptosis, and reduces migration and angiogenesis in liver cancer cells. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:361-370. [PMID: 37524306 DOI: 10.1080/21691401.2023.2238756] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 05/05/2023] [Accepted: 06/16/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Curcumin has been used in the treatment of several diseases; however, its low pharmacologic profile reduces its therapeutic use. Towards improving its biological activity, nanoformulations have emerged. Thus, we aimed to determine whether curcumin nanoparticles (Cur-NPs) coated with PEG/chitosan improve the treatment of liver cancer (LC) cells and underpin the molecular mechanisms underlying their anti-cancer activity. METHODS Cur-NPs were synthesised in the form of Cur-PLGA-PEG/chitosan NPs. The effect of Cur-NPs was assessed in HepG2 and Huh 7 LC cells and THLE-2 normal liver cells. RESULTS The size of synthesised Cur-NPS was determined in the standard range of 141.2 ± 47.5 nm. Compared to THLE-2 cells, LC cells treated with Cur-NPs exerted cytotoxicity at 6.25 µg/mL after 48h. Treatment of HepG-2 cells with 2.5 µg/mL of Cur-NPs inhibited cell migration and this inhibition was augmented at 10 µg/mL (p < 0.001). Treatment of chicken embryo with 5 µg/mL Cur-NPs reduced angiogenesis (p < 0.001) of 4-day-old embryos. The nanoformulation upregulated Bax and p53 and downregulated Bcl-2 in a concentration-dependent manner and subsequently induce apoptosis in HepG-2 cells. CONCLUSION Treatment of LC cells with Cur-NPs decreased cell proliferation, migration, and angiogenesis, and induced cell death by promoting the proapoptotic pathway.
Collapse
Affiliation(s)
- Steve Harakeh
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Saber H Saber
- Laboratory of Molecular Cell Biology, Department of Zoology, Faculty of Science, Assiut University, Assiut, Egypt
| | - Rajaa Al-Raddadi
- Department of Community Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Turki Alamri
- Family and Community Medicine Department, Faculty of Medicine in Rabigh, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Soad Al-Jaouni
- Yousef Abdul Latif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Hematology/Pediatric Oncology, King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammed Qari
- Department of Hematology/Pediatric Oncology, King Abdulaziz University Hospital, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Yousef Qari
- Department of Medicine, King Abdulaziz University Hospital, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
| | - Ayat Zawawi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Soad S Ali
- Anatomy Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Zakaria Y Abd Elmageed
- Department of Pharmacology, Edward Via College of Osteopathic Medicine, University of LA at Monroe, Monroe, LA, USA
| | - Shaker Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, USA
| |
Collapse
|
7
|
Sarma S, Deka DJ, Rajak P, Laloo D, Das T, Chetia P, Saha D, Bharali A, Deka B. Potential injectable hydrogels as biomaterials for central nervous system injury: A narrative review. IBRAIN 2023; 9:402-420. [PMID: 38680508 PMCID: PMC11045191 DOI: 10.1002/ibra.12137] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/20/2023] [Accepted: 10/27/2023] [Indexed: 05/01/2024]
Abstract
Numerous modalities exist through which the central nervous system (CNS) may sustain injury or impairment, encompassing traumatic incidents, stroke occurrences, and neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease. Presently available pharmacological and therapeutic interventions are incapable of restoring or regenerating damaged CNS tissue, leading to substantial unmet clinical needs among patients with CNS ailments or injuries. To address and facilitate the recovery of the impaired CNS, cell-based repair strategies encompass multiple mechanisms, such as neuronal replacement, therapeutic factor secretion, and the promotion of host brain plasticity. Despite the progression of cell-based CNS reparation as a therapeutic strategy throughout the years, substantial barriers have impeded its widespread implementation in clinical settings. The integration of cell technologies with advancements in regenerative medicine utilizing biomaterials and tissue engineering has recently facilitated the surmounting of several of these impediments. This comprehensive review presents an overview of distinct CNS conditions necessitating cell reparation, in addition to exploring potential biomaterial methodologies that enhance the efficacy of treating brain injuries.
Collapse
Affiliation(s)
- Santa Sarma
- Girijananda Chowdhury Institute of Pharmaceutical ScienceAssam Science and Technology UniversityGuwahatiAssamIndia
| | - Dhruva J. Deka
- Girijananda Chowdhury Institute of Pharmaceutical ScienceAssam Science and Technology UniversityGuwahatiAssamIndia
| | - Prakash Rajak
- Department of Pharmaceutical SciencesDibrugarh UniversityDibrugarhAssamIndia
| | - Damiki Laloo
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Trishna Das
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Purbajit Chetia
- Department of PharmacologyNETES Institute of Pharmaceutical Science, Nemcare Group of Institutes, MirzaGuwahatiAssamIndia
| | - Dipankar Saha
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Alakesh Bharali
- Department of Pharmaceutical SciencesDibrugarh UniversityDibrugarhAssamIndia
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| | - Bhargab Deka
- School of Pharmaceutical SciencesGirijananda Chowdhury UniversityGuwahatiAssamIndia
| |
Collapse
|
8
|
Afshari AR, Sanati M, Kesharwani P, Sahebkar A. Recent Advances in Curcumin-Based Combination Nanomedicines for Cancer Therapy. J Funct Biomater 2023; 14:408. [PMID: 37623653 PMCID: PMC10455605 DOI: 10.3390/jfb14080408] [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: 06/11/2023] [Revised: 07/17/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Standard cancer chemotherapeutics often produce significant adverse effects and eventually lose their effectiveness due to the emergence of resistance mechanisms. As a result, patients with malignant tumors experience a poor quality of life and a short lifespan. Thus, combination medication regimens provide various advantages, including increased success rate, fewer side effects, and fewer occurrences of resistance. Curcumin (Cur), a potential phytochemical from turmeric, when coupled with traditional chemotherapeutics, has been established to improve the effectiveness of cancer treatment in clinical and preclinical investigations. Cur not only exerts multiple mechanisms resulting in apoptotic cancer cell death but also reduces the resistance to standard chemotherapy drugs, mainly through downregulating the multi-drug resistance (MDR) cargoes. Recent reports showed the beneficial outcomes of Cur combination with many chemotherapeutics in various malignancies. Nevertheless, owing to the limited bioavailability, devising co-delivery strategies for Cur and conventional pharmaceuticals appears to be required for clinical settings. This review summarized various Cur combinations with standard treatments as cancer therapeutics.
Collapse
Affiliation(s)
- Amir R. Afshari
- Department of Physiology and Pharmacology, Faculty of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
- Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 602105, India
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
9
|
Serini S, Trombino S, Curcio F, Sole R, Cassano R, Calviello G. Hyaluronic Acid-Mediated Phenolic Compound Nanodelivery for Cancer Therapy. Pharmaceutics 2023; 15:1751. [PMID: 37376199 DOI: 10.3390/pharmaceutics15061751] [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: 05/18/2023] [Revised: 06/13/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Phenolic compounds are bioactive phytochemicals showing a wide range of pharmacological activities, including anti-inflammatory, antioxidant, immunomodulatory, and anticancer effects. Moreover, they are associated with fewer side effects compared to most currently used antitumor drugs. Combinations of phenolic compounds with commonly used drugs have been largely studied as an approach aimed at enhancing the efficacy of anticancer drugs and reducing their deleterious systemic effects. In addition, some of these compounds are reported to reduce tumor cell drug resistance by modulating different signaling pathways. However, often, their application is limited due to their chemical instability, low water solubility, or scarce bioavailability. Nanoformulations, including polyphenols in combination or not with anticancer drugs, represent a suitable strategy to enhance their stability and bioavailability and, thus, improve their therapeutic activity. In recent years, the development of hyaluronic acid-based systems for specific drug delivery to cancer cells has represented a pursued therapeutic strategy. This is related to the fact that this natural polysaccharide binds to the CD44 receptor that is overexpressed in most solid cancers, thus allowing its efficient internalization in tumor cells. Moreover, it is characterized by high biodegradability, biocompatibility, and low toxicity. Here, we will focus on and critically analyze the results obtained in recent studies regarding the use of hyaluronic acid for the targeted delivery of bioactive phenolic compounds to cancer cells of different origins, alone or in combination with drugs.
Collapse
Affiliation(s)
- Simona Serini
- Department of Translational Medicine and Surgery, Section of General Pathology, School of Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 00168 Rome, Italy
| | - Sonia Trombino
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Federica Curcio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Roberta Sole
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Roberta Cassano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Arcavacata di Rende, 87036 Cosenza, Italy
| | - Gabriella Calviello
- Department of Translational Medicine and Surgery, Section of General Pathology, School of Medicine and Surgery, Università Cattolica del Sacro Cuore, Largo F. Vito, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo F. Vito, 00168 Rome, Italy
| |
Collapse
|
10
|
Yuxue J, Ran S, Minghui F, Minjia S. Applications of nanomaterials in endometriosis treatment. Front Bioeng Biotechnol 2023; 11:1184155. [PMID: 37229500 PMCID: PMC10203239 DOI: 10.3389/fbioe.2023.1184155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 04/26/2023] [Indexed: 05/27/2023] Open
Abstract
Endometriosis is a common disease of the reproductive system in women of childbearing age with an unclear pathogenesis. Endometriosis mainly manifests as dysmenorrhea, abdominal pain, and infertility. Currently, medical therapy and surgical treatment are usually used for endometriosis treatment. However, due to the high recurrence rate and many complications, it has greatly affected patients' quality of life. Nanotechnology is a new technology that mainly investigates the characteristics and applications of nanomaterials. To date, nanotechnology has received widespread attention in the field of biomedicine. Nanomaterials can not only be used as drugs to treat endometriosis directly, but also enhance the therapeutic effect of endometriosis by delivering drugs, siRNA, antibodies, vesicles, etc. This review comprehensively discusses nanomaterial-based therapies for endometriosis treatment, such as nanomaterial-based gene therapy, photothermal therapy, immunotherapy, and magnetic hyperthermia, which provides a theoretical reference for the clinical application of nanotechnology in the treatment of endometriosis.
Collapse
|
11
|
Khan MS, Altwaijry N, Jabir NR, Alamri AM, Tarique M, Khan AU. Potential of green-synthesized ZnO NPs against human ovarian teratocarcinoma: an in vitro study. Mol Biol Rep 2023; 50:4447-4457. [PMID: 37014566 DOI: 10.1007/s11033-023-08367-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/01/2023] [Indexed: 04/05/2023]
Abstract
BACKGROUND Ovarian cancer leads to devastating outcomes, and its treatment is highly challenging. At present, there is a lack of clinical symptoms, well-known sensitivity biomarkers, and patients are diagnosed at an advanced stage. Currently, available therapeutics against ovarian cancer are inefficient, costly, and associated with severe side effects. The present study evaluated the anticancer potential of zinc oxide nanoparticles (ZnO NPs) that were successfully biosynthesized in an ecofriendly mode using pumpkin seed extracts. METHODS AND RESULTS The anticancer potential of the biosynthesized ZnO NPs was assessed using an in vitro human ovarian teratocarcinoma cell line (PA-1) by well-known assays such as MTT assay, morphological alterations, induction of apoptosis, measurement of reactive oxygen species (ROS) production, and inhibition of cell adhesion/migration. The biogenic ZnO NPs exerted a high level of cytotoxicity against PA-1 cells. Furthermore, the ZnO NPs inhibited cellular adhesion and migration but induced ROS production and cell death through programmed cell death. CONCLUSION The aforementioned anticancer properties highlight the therapeutic utility of ZnO NPs in ovarian cancer treatment. However, further research is recommended to envisage their mechanism of action in different cancer models and validation in a suitable in vivo system.
Collapse
Affiliation(s)
- Mohd Shahnawaz Khan
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, 11451, Saudi Arabia.
| | - Nojood Altwaijry
- Department of Biochemistry, College of Sciences, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Nasimudeen R Jabir
- Department of Biochemistry and Biotechnology, Centre for Research and Development, PRIST University, Vallam, Thanjavur, TN, 613403, India
| | | | - Mohammad Tarique
- Department of Child Health, University of Missouri, Columbia, MO, USA
| | - Azhar U Khan
- Department of Chemistry, School of Life and Basic Sciences, SIILAS CAMPUS, Jaipur National University, Jaipur, India
| |
Collapse
|
12
|
Peng P, Chen Z, Wang M, Wen B, Deng X. Polysaccharide-modified liposomes and their application in cancer research. Chem Biol Drug Des 2023; 101:998-1011. [PMID: 36597375 DOI: 10.1111/cbdd.14201] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 11/25/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023]
Abstract
Nanodrug delivery systems have been widely used in cancer treatment. Among these, liposomal drug carriers have gained considerable attention due to their biocompatibility, biodegradability, and low toxicity. However, conventional liposomes have several shortcomings, such as poor stability, rapid clearance, aggregation, fusion, degradation, hydrolysis, and oxidation of phospholipids. Polysaccharides are natural polymers of biological origin that exhibit structural stability, excellent biocompatibility and biodegradability, flexibility, non-immunogenicity, low toxicity, and targetability. Therefore, they represent a promising class of polymers for the modification of the surface properties of liposomes to overcome their shortcomings. In addition, polysaccharides can be readily combined with other materials to develop new composite materials. Hence, they represent the optimal choice for liposomal modification to improve pharmacokinetics and clinical utility. Polysaccharide-coated liposomes exhibit better stability, drug release kinetics, and cellular uptake than conventional liposomes. The oncologic application of polysaccharide-coated liposomes has become a research hotspot. We summarize the preparation, physicochemical properties, and antineoplastic effects of polysaccharide-coated liposomes to facilitate antitumor drug development.
Collapse
Affiliation(s)
- Peichun Peng
- International Zhuang Medical Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Zeshan Chen
- Department of Traditional Chinese Medicine, Guangxi Academy of Medical Sciences, Nanning, China
| | - Miaodong Wang
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Bin Wen
- Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, China
| | - Xin Deng
- Department of Basic Medical Science College, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
13
|
Gong J, Shi T, Liu J, Pei Z, Liu J, Ren X, Li F, Qiu F. Dual-drug codelivery nanosystems: An emerging approach for overcoming cancer multidrug resistance. Biomed Pharmacother 2023; 161:114505. [PMID: 36921532 DOI: 10.1016/j.biopha.2023.114505] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Multidrug resistance (MDR) promotes tumor recurrence and metastasis and heavily reduces anticancer efficiency, which has become a primary reason for the failure of clinical chemotherapy. The mechanisms of MDR are so complex that conventional chemotherapy usually fails to achieve an ideal therapeutic effect and even accelerates the occurrence of MDR. In contrast, the combination of chemotherapy with dual-drug has significant advantages in tumor therapy. A novel dual-drug codelivery nanosystem, which combines dual-drug administration with nanotechnology, can overcome the application limitation of free drugs. Both the characteristics of nanoparticles and the synergistic effect of dual drugs contribute to circumventing various drug-resistant mechanisms in tumor cells. Therefore, developing dual-drug codelivery nanosystems with different multidrug-resistant mechanisms has an important reference value for reversing MDR and enhancing the clinical antitumor effect. In this review, the advantages, principles, and common codelivery nanocarriers in the application of dual-drug codelivery systems are summarized. The molecular mechanisms of MDR and the dual-drug codelivery nanosystems designed based on different mechanisms are mainly introduced. Meanwhile, the development prospects and challenges of codelivery nanosystems are also discussed, which provide guidelines to exploit optimized combined chemotherapy strategies in the future.
Collapse
Affiliation(s)
- Jianing Gong
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Taoran Shi
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jinfeng Liu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Zerong Pei
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jingbo Liu
- College of Horticulture and Landscape Architecture, Tianjin Agricultural University, Tianjin 300384, China
| | - Xiaoliang Ren
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Fengyun Li
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
14
|
Pulmonary delivery of curcumin and quercetin nanoparticles for lung cancer – Part 2: Toxicity and endocytosis. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
|
15
|
Liu X, Qi M, Li X, Wang J, Wang M. Curcumin: a natural organic component that plays a multi-faceted role in ovarian cancer. J Ovarian Res 2023; 16:47. [PMID: 36859398 PMCID: PMC9976389 DOI: 10.1186/s13048-023-01120-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 02/15/2023] [Indexed: 03/03/2023] Open
Abstract
Curcumin, a natural organic component obtained from Curcuma longa's rhizomes, shows abundant anti-tumor, antioxidant and anti-inflammatory pharmacological activities, among others. Notably the anti-tumor activity has aroused widespread attention from scholars worldwide. Numerous studies have reported that curcumin can delay ovarian cancer (OC), increase its sensitivity to chemotherapy, and reduce chemotherapy drugs' side effects. It has been shown considerable anticancer potential by promoting cell apoptosis, suppressing cell cycle progression, inducing autophagy, inhibiting tumor metastasis, and regulating enzyme activity. With an in-depth study of curcumin's anti-OC mechanism, its clinical application will have broader prospects. This review summarizes the latest studies on curcumin's anti-OC activities, and discusses the specific mechanism, hoping to provide references for further research and applications.
Collapse
Affiliation(s)
- Xiaoping Liu
- grid.216417.70000 0001 0379 7164Department of gynaecology and obstetrics, the Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, 412000 Zhuzhou, Hunan China
| | - Mingming Qi
- grid.216417.70000 0001 0379 7164Department of gynaecology and obstetrics, the Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, 412000 Zhuzhou, Hunan China
| | - Xidie Li
- grid.216417.70000 0001 0379 7164Department of gynaecology and obstetrics, the Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, 412000 Zhuzhou, Hunan China
| | - Jingjin Wang
- Department of gynaecology and obstetrics, the Affiliated Zhuzhou Hospital Xiangya Medical College, Central South University, 412000, Zhuzhou, Hunan, China.
| | - Mingyuan Wang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China. .,Department of Geriatric Surgery, Xiangya Hospital, Central South University, 410008, Changsha, Hunan, China.
| |
Collapse
|
16
|
Idlas P, Lepeltier E, Bastiat G, Pigeon P, McGlinchey MJ, Lautram N, Vessières A, Jaouen G, Passirani C. Physicochemical Characterization of Ferrocifen Lipid Nanocapsules: Customized Drug Delivery Systems Guided by the Molecular Structure. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:1885-1896. [PMID: 36693216 DOI: 10.1021/acs.langmuir.2c02910] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Ferrocifens, lipophilic organometallic complexes, comprise a biologically active redox motif [ferrocenyl-ene-p-phenol] which confers very interesting cytotoxic properties to this family. However, because of their highly lipophilic nature, a formulation stage is required before being administered in vivo. In recent decades, ferrocifen lipid nanocapsules (LNCs) have been successfully formulated and have demonstrated anticancer activity on multidrug-resistant cancers in several mice and rat models (glioblastoma, breast cancer, and metastatic melanoma). A recent family of ferrocifens (succinimidoalkyl-ferrociphenols, including P722) appears to be most efficacious on several resistant cancer cell lines, with IC50 values in the nanomolar range together with promising in vivo results on murine ovarian cancer models. As LNCs are composed of an oily core (caprylic/capric triglycerides), modulation of the succinimido-ferrociphenol lipophilicity could be a valuable approach toward improving the drug loading in LNCs. As the drug loading of the diphenol P722 in LNCs was low, it was structurally modified to increase its lipophilicity and thereby the payload in LNCs. Chemical modification led to a series of five succinimido-ferrocifens. Results confirmed that these slight structural modifications led to increased drug loading in LNCs for all ferrocifens, with no reduction of their cytotoxicity on the SKOV3 ovarian cancer cell line. Interestingly, encapsulation of two of the ferrocifens, diester P769 and monophenolic ester (E)-P998, led to the formation of a gel. This was unprecedented behavior, a phenomenon that could be rationalized in terms of the positioning of ferrocifens in LNCs as shown by the decrease of interfacial tension measurements at the water/oil interface. Moreover, these results highlighted the importance of obtaining a gel of this particular motif, in which the acetylated phenolic ring and the succinimidoalkyl moieties are mutually cis relative to the central double bond. Promising perspectives to use these ferrocifen-loaded LNCs to treat glioblastoma could be readily envisaged by local application of the gel in the cavity after tumor resection.
Collapse
Affiliation(s)
- Pierre Idlas
- Micro et Nanomédecines Translationnelles, MINT, Université Angers, INSERM 1066, CNRS 6021, Angers49100, France
| | - Elise Lepeltier
- Micro et Nanomédecines Translationnelles, MINT, Université Angers, INSERM 1066, CNRS 6021, Angers49100, France
| | - Guillaume Bastiat
- Micro et Nanomédecines Translationnelles, MINT, Université Angers, INSERM 1066, CNRS 6021, Angers49100, France
| | - Pascal Pigeon
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), Paris75005, France
- Chimie Paris Tech, PSL University, Paris75005, France
| | | | - Nolwenn Lautram
- Micro et Nanomédecines Translationnelles, MINT, Université Angers, INSERM 1066, CNRS 6021, Angers49100, France
| | - Anne Vessières
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), Paris75005, France
| | - Gerard Jaouen
- Sorbonne Université, CNRS, Institut Parisien de Chimie Moléculaire (IPCM), Paris75005, France
- Chimie Paris Tech, PSL University, Paris75005, France
| | - Catherine Passirani
- Micro et Nanomédecines Translationnelles, MINT, Université Angers, INSERM 1066, CNRS 6021, Angers49100, France
| |
Collapse
|
17
|
El-Saadony MT, Yang T, Korma SA, Sitohy M, Abd El-Mageed TA, Selim S, Al Jaouni SK, Salem HM, Mahmmod Y, Soliman SM, Mo’men SAA, Mosa WFA, El-Wafai NA, Abou-Aly HE, Sitohy B, Abd El-Hack ME, El-Tarabily KA, Saad AM. Impacts of turmeric and its principal bioactive curcumin on human health: Pharmaceutical, medicinal, and food applications: A comprehensive review. Front Nutr 2023; 9:1040259. [PMID: 36712505 PMCID: PMC9881416 DOI: 10.3389/fnut.2022.1040259] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/09/2022] [Indexed: 01/11/2023] Open
Abstract
The yellow polyphenolic pigment known as curcumin, originating from the rhizome of the turmeric plant Curcuma longa L., has been utilized for ages in ancient medicine, as well as in cooking and food coloring. Recently, the biological activities of turmeric and curcumin have been thoroughly investigated. The studies mainly focused on their antioxidant, antitumor, anti-inflammatory, neuroprotective, hepatoprotective, and cardioprotective impacts. This review seeks to provide an in-depth, detailed discussion of curcumin usage within the food processing industries and its effect on health support and disease prevention. Curcumin's bioavailability, bio-efficacy, and bio-safety characteristics, as well as its side effects and quality standards, are also discussed. Finally, curcumin's multifaceted uses, food appeal enhancement, agro-industrial techniques counteracting its instability and low bioavailability, nanotechnology and focused drug delivery systems to increase its bioavailability, and prospective clinical use tactics are all discussed.
Collapse
Affiliation(s)
- Mohamed T. El-Saadony
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Tao Yang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Sameh A. Korma
- Department of Food Science, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Mahmoud Sitohy
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Taia A. Abd El-Mageed
- Department of Soils and Water, Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka, Saudi Arabia
| | - Soad K. Al Jaouni
- Department of Hematology/Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Heba M. Salem
- Department of Poultry Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Yasser Mahmmod
- Department of Veterinary Sciences, Faculty of Health Sciences, Higher Colleges of Technology, Al Ain, United Arab Emirates
| | - Soliman M. Soliman
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Shaimaa A. A. Mo’men
- Department of Entomology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Walid F. A. Mosa
- Plant Production Department (Horticulture-Pomology), Faculty of Agriculture Saba Basha, Alexandria University, Alexandria, Egypt
| | - Nahed A. El-Wafai
- Department of Agricultural Microbiology, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Hamed E. Abou-Aly
- Department of Agricultural Microbiology, Faculty of Agriculture, Benha University, Benha, Egypt
| | - Basel Sitohy
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
- Department of Radiation Sciences, Oncology, Umeå University, Umeå, Sweden
| | - Mohamed E. Abd El-Hack
- Department of Poultry Diseases, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Khaled A. El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, United Arab Emirates
- Khalifa Center for Genetic Engineering and Biotechnology, United Arab Emirates University, Al Ain, United Arab Emirates
- Harry Butler Institute, Murdoch University, Murdoch, WA, Australia
| | - Ahmed M. Saad
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| |
Collapse
|
18
|
Jia W, Zhou L, Li L, Zhou P, Shen Z. Nano-Based Drug Delivery of Polyphenolic Compounds for Cancer Treatment: Progress, Opportunities, and Challenges. Pharmaceuticals (Basel) 2023; 16:ph16010101. [PMID: 36678599 PMCID: PMC9865384 DOI: 10.3390/ph16010101] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/12/2023] Open
Abstract
Polyphenols and their derivates, a kind of natural product distributed in herb plants, vegetables, and fruits, are the most abundant antioxidants in the human diet and have been found to display cancer-preventative effects in several epidemiological studies. The scientific community has also validated the anti-cancer bioactivities and low toxicities of polyphenolic compounds, including flavones, tannins, phenolic acids, and anthocyanins, through in vitro and in vivo studies. However, the low stability, weak targeting ability, poor solubility, and low bioavailability of pure polyphenolic agents have significantly impaired their treatment efficacy. Nowadays, nano-based technology has been applied to surmount these restrictions and maximize the treatment efficacy of polyphenols. In this review, we summarize the advantages and related mechanisms of polyphenols in cancer treatment. Moreover, aiming at the poor solubility and low bioavailability of pure polyphenols in vivo, the advantages of nano-based delivery systems and recent research developments are highlighted. Herein, particular emphasis is mainly placed on the most widely used nanomaterials in the delivery of natural products, including liposomes, micelles, and nanogels. Finally, we present an overview and the challenges of future implementations of nano-based delivery systems of polyphenolic compounds in the cancer therapeutic field.
Collapse
Affiliation(s)
- Wenhui Jia
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China School of Basic Medical Sciences & Forensic Medicine, Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, China
| | - Li Zhou
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400016, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ping Zhou
- Department of Radiotherapy, The First Affiliated Hospital of Hainan Medical University, Haikou 571199, China
- Correspondence: (P.Z.); (Z.S.)
| | - Zhisen Shen
- Department of Otorhinolaryngology and Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo 315211, China
- Correspondence: (P.Z.); (Z.S.)
| |
Collapse
|
19
|
Lee P, Zhou C, Hu X. VX-765 has a Protective Effect on Mice with Ovarian Injury Caused by Chemotherapy. Curr Cancer Drug Targets 2023; 23:307-318. [PMID: 36200259 DOI: 10.2174/1568009622666220930110024] [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: 03/21/2022] [Revised: 06/29/2022] [Accepted: 08/15/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Malignant tumors continue to remain a main global public health issue. In the past 40 years, due to strides made in multi-disciplinary comprehensive treatment schemes for patients suffering from malignant tumors, especially chemotherapy schemes, the survival rate has been greatly improved in such patients. This group can be expected to maintain their fertility or have restored endocrine function following successful malignant tumor treatment. Therefore, focusing on the ovarian damage caused by chemotherapy in women of childbearing age is vital in order to protect their fertility and improve their quality of life. OBJECTIVE This study attempted to evaluate whether VX-765 possesses an ovarian protective effect in ovarian injury induced by chemotherapy in the mice model. METHODS Female C57BL/6J mice were administered with VX-765 gavage once a day for 21 consecutive days. Use of cyclophosphamide (Cy) began one week after the last gavage administration of VX- 765. Detailed classification of follicles at various levels was then quantified in each group. Immunohistochemistry and Western blot analysis were then used in order to analyze the expression of key proteins (FOXO3a, mTOR, RPS6 and AKT) as well as their phosphorylation of the PI3K / PTEN / AKT pathways in the ovary. The concentrations of AMH were measured by ELISA. RESULTS The follicles at all levels of Cy treated mice were less than those of the normal group (P < 0.05). Meanwhile, mice treated with VX-765 prior to receiving Cy treatment had more primordial follicles (PMF) than mice treated with Cy alone (P < 0.05). In early growing follicles (EGF) and antral follicles (AF), no difference was observed among the experimental groups (P > 0.05), however, they were lower than those in the normal group (P < 0.05). In mice treated with continuous Cy, the total follicle number (TF) of mice combined with VX-765 (C-Cy-Vx765) was higher than that of mice without VX-765, and the TF of the two groups was lower than that of the normal group (P < 0.05). The value of PMF/TF in C-Cy-Vx765 group was significantly higher than that in the other three groups, while that of EGF/TF was significantly lower (P < 0.05). Immunohistochemical results showed that the phosphorylated forms of the main proteins of the PI3K / PTEN / AKT pathway were found to be more positive in Cy treated mice. The Western blot analysis showed that when Cy and VX-765 were cotreated, the increased levels of these phosphorylated proteins decreased compared with those treated with Cy alone. The AMH level of infancy Cy and VX-765 co-treated mice was higher than that of infancy normal mice (P < 0.05). After the mice grew to sexual maturity, the AMH level of Cy and VX- 765 co-treated mice was still higher than that of Cy treated mice (P < 0.05), and there was no significant difference with normal mice (P > 0.05). CONCLUSION VX-765 can maintain the level of AMH and inhibit the recruitment of PMF, thus protecting mice from Cy induced gonadotropic toxicity. Accordingly, VX-765 may play a protective role in mice with ovarian injury caused by chemotherapy.
Collapse
Affiliation(s)
- Pingyin Lee
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Canquan Zhou
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaokun Hu
- Reproductive Medicine Center, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Reproductive Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
20
|
Wu Y, Yang Y, Lv X, Gao M, Gong X, Yao Q, Liu Y. Nanoparticle-Based Combination Therapy for Ovarian Cancer. Int J Nanomedicine 2023; 18:1965-1987. [PMID: 37077941 PMCID: PMC10106804 DOI: 10.2147/ijn.s394383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Accepted: 03/19/2023] [Indexed: 04/21/2023] Open
Abstract
Ovarian cancer is one of the most common malignant tumors in gynecology with a high incidence. Combination therapy, eg, administration of paclitaxel followed by a platinum anticancer drug is recommended to treat ovarian cancer due to its advantages in, eg, reducing side effects and reversing (multi)drug-resistance compared to single treatment. However, the benefits of combination therapy are often compromised. In chemo and chemo/gene combinations, co-deposition of the combined therapeutics in the tumor cells is required, which is difficult to achieve due to dramatic pharmacokinetic differences between combinational agents in free forms. Moreover, some undesired properties such as the low-water solubility of chemodrugs and the difficulty of cellular internalization of gene therapeutics also hinder the therapeutic potential. Delivery of dual or multiple agents by nanoparticles provides opportunities to tackle these limits. Nanoparticles encapsulate hydrophobic drug(s) to yield aqueous dispersions facilitating its administration and/or to accommodate hydrophilic genes facilitating its access to cells. Moreover, nanoparticle-based therapeutics can not only improve drug properties (eg, in vivo stability) and ensure the same drug disposition behavior with controlled drug ratios but also can minimize drug exposure of the normal tissues and increase drug co-accumulation at targeted tissues via passive and/or active targeting strategies. Herein, this work summarizes nanoparticle-based combination therapies, mainly including anticancer drug-based combinations and chemo/gene combinations, and emphasizes the advantageous outcomes of nanocarriers in the combination treatment of ovarian cancer. In addition, we also review mechanisms of synergetic effects resulting from different combinations.
Collapse
Affiliation(s)
- Yingli Wu
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Jinan, Shandong, 250117, People’s Republic of China
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, People’s Republic of China
| | - Yu Yang
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Jinan, Shandong, 250117, People’s Republic of China
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, People’s Republic of China
| | - Xiaolin Lv
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Jinan, Shandong, 250117, People’s Republic of China
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, People’s Republic of China
| | - Menghan Gao
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
| | - Xujin Gong
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Jinan, Shandong, 250117, People’s Republic of China
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, People’s Republic of China
| | - Qingqiang Yao
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Jinan, Shandong, 250117, People’s Republic of China
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, People’s Republic of China
- Jining Medical University, Jining, Shandong, 272067, People’s Republic of China
- Correspondence: Qingqiang Yao, Jining Medical University, No. 133 HeHua Road, Jinan, Shandong, 272067, People’s Republic of China, Email
| | - Yanna Liu
- School of Pharmacy and Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, People’s Republic of China
- NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Jinan, Shandong, 250117, People’s Republic of China
- Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, Shandong, 250117, People’s Republic of China
- Yanna Liu, Shandong First Medical University, No. 6699 Qingdao Road, HuaiYin District, Jinan, Shandong, 250117, People’s Republic of China, Email
| |
Collapse
|
21
|
Wang YJ, Tang L, Lu XH, Liu JT, Wang YY, Geng HX, Li XT, An Q. Efficacy of epi-1 modified epirubicin and curcumin encapsulated liposomes targeting-EpCAM in the inhibition of epithelial ovarian cancer cells. J Liposome Res 2022:1-17. [PMID: 36440599 DOI: 10.1080/08982104.2022.2153138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Treatment of epithelial ovarian cancer (EOC) is a challenge because it still leads to unsatisfactory clinical prognosis. This is due to the toxicity and poor targeting of chemotherapeutic agents, as well as metastasis of the tumor. In this study, we designed a targeted liposome with nanostructures to overcome these problems. In the liposomes, epirubicin and curcumin were encapsulated to achieve their synergistic antitumor efficacy, while Epi-1 was modified on the liposomal surface to target epithelial cell adhesion molecule (EpCAM). Epi-1, a macrocyclic peptide, exhibits active targeting for enhanced cellular uptake and potent cytotoxicity against tumor cells. The encapsulation of epirubicin and curcumin synergistically inhibited the formation of neovascularization and vasculogenic mimicry (VM) channels, thereby suppressing tumor metastasis on SKOV3 cells. The dual drug loaded Epi-1-liposomes also induced apoptosis and downregulated metastasis-related proteins for effective antitumor in vitro. In vivo studies showed that dual drug loaded Epi-1-liposomes prolonged circulation time in the blood and increased the selective accumulation of drug at the tumor site. H&E staining and immunohistochemistry with Ki-67 also showed that targeted liposomes elevated antitumor activity. Also, targeted liposomes downregulated angiogenesis-related proteins to inhibit angiogenesis and thus tumor metastasis. In conclusion, the production of dual drug loaded Epi-1-liposomes is an effective strategy for the treatment of EOC.
Collapse
Affiliation(s)
- Yu-Jia Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Ling Tang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Xu-Hong Lu
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Ji-Tao Liu
- Technology Research and Development Centre, Yunnan Baiyao Group Health Products Co., Ltd, Kunming, China
| | - Yuan-Yuan Wang
- Department of Obstetrics and Gynecology, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Hong-Xia Geng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Xue-Tao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, China
| | - Quan An
- Technology Research and Development Centre, Yunnan Baiyao Group Health Products Co., Ltd, Kunming, China
| |
Collapse
|
22
|
Li B, Shao H, Gao L, Li H, Sheng H, Zhu L. Nano-drug co-delivery system of natural active ingredients and chemotherapy drugs for cancer treatment: a review. Drug Deliv 2022; 29:2130-2161. [PMID: 35815678 PMCID: PMC9275501 DOI: 10.1080/10717544.2022.2094498] [Citation(s) in RCA: 80] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy drugs have been used for a long time in the treatment of cancer, but serious side effects are caused by the inability of the drug to be solely delivered to the tumor when treating cancer with chemotherapy. Natural products have attracted more and more attention due to the antitumor effect in multiple ways, abundant resources and less side effects. Therefore, the combination of natural active ingredients and chemotherapy drugs may be an effective antitumor strategy, which can inhibit the growth of tumor and multidrug resistance, reduce side effects of chemotherapy drugs. Nano-drug co-delivery system (NDCDS) can play an important role in the combination of natural active ingredients and chemotherapy drugs. This review provides a comprehensive summary of the research status and application prospect of nano-delivery strategies for the combination of natural active ingredients and chemotherapy drugs, aiming to provide a basis for the development of anti-tumor drugs.
Collapse
Affiliation(s)
- Bingqian Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huili Shao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Lei Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huan Li
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
23
|
Iqubal MK, Kaur H, Md S, Alhakamy NA, Iqubal A, Ali J, Baboota S. A technical note on emerging combination approach involved in the onconanotherapeutics. Drug Deliv 2022; 29:3197-3212. [PMID: 36226570 PMCID: PMC9578464 DOI: 10.1080/10717544.2022.2132018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Cancer is the second cause of mortality worldwide, and the currently available conventional treatment approach is associated with serious side effects and poor clinical outcomes. Based on the outcome of the exploratory preclinical and clinical studies, it was found that therapeutic response increases multiple folds when anticancer drugs are used in combination. However, the conventional combination of anticancer drugs was associated with various limitations such as increased cost of treatment, systemic toxicity, drug resistance, and reduced pharmacokinetic attributes. Hence, attempts were made to formulate nanocarrier fabricated combinatorial drugs (NFCDs) to effectively manage and treat cancer. This approach offers several advantages, such as improved stability, lower drug exposure, targeted drug delivery, low side effects, and improved clinical outcome. Hence, in this review, first time, we have discussed the recent advancement and various types of nano carrier-based combinatorial drug delivery systems in a different type of cancer and highlighted the personalized combinatorial theranostic medicine as a futuristic anticancer treatment approach.
Collapse
Affiliation(s)
- Mohammad Kashif Iqubal
- Product Development Department, Sentiss Research Centre, Sentiss Pharma Pvt Ltd, Gurugram, India.,Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Harsimran Kaur
- Department of Pharmaceutics, Delhi Pharmaceutical Science and Research University, New Delhi, India
| | - Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nabil A Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ashif Iqubal
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
24
|
Da J, Li Y, Zhang K, Ren J, Wang J, Liu X, Liu X, Zhang J, Liu L, Zhang W, Zhang S, Guo Y, Zhang B, Jin H. Functionalized Prussian Blue Nanozyme as Dual-Responsive Drug Therapeutic Nanoplatform Against Maxillofacial Infection via Macrophage Polarization. Int J Nanomedicine 2022; 17:5851-5868. [PMCID: PMC9719692 DOI: 10.2147/ijn.s385899] [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: 08/12/2022] [Accepted: 11/18/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose Maxillofacial infection is a common disease in stomatology and is difficult to treat owing to its high potential to spread to vital anatomical structures. Excessive levels of reactive oxygen species (ROS) in infected tissues lead to cellular damage and impede tissue regeneration. However, uncontrollable strategies to remove ROS have limited therapeutic efficacy. Nanoparticle systems for scavenging ROS and remodeling the inflammatory microenvironment offer much promise in the treatment of maxillofacial inflammation. Methods Here, a novel microenvironment-stimuli-responsive drug delivery nanoplatform (HMPB@Cur@PDA) based on a polydopamine (PDA)-functionalized hollow mesoporous Prussian blue (HMPB) nanozyme was developed for the delivery of curcumin (Cur) in the treatment of maxillofacial infection. Low pH and excess ROS in the inflammatory microenvironment cause degradation of the outer PDA layer of the nanocomplex, exposing the HMPB nanozyme and loaded Cur, which synergistically act as a ROS scavenger and anti-inflammatory agent, respectively, and induce macrophage polarization from the pro-inflammatory M1 to the anti-inflammatory M2 phenotype. Results Experiments in vitro provided strong evidence for the application of novel nanocomplexes in scavenging multiple ROS and inhibiting lipopolysaccharide-induced inflammation. In addition, in vivo results obtained using a mouse maxillofacial infection model demonstrated that HMPB@Cur@PDA had excellent biocompatibility, significantly attenuated the inflammatory response in periodontal tissue, and improved the repair of damaged tissue. Conclusion Our results indicate that HMPB@Cur@PDA nanocomposites have great potential for ROS regulation as well as having anti-inflammatory effects, providing new insights for the development of dual-response maxillofacial infection treatments.
Collapse
Affiliation(s)
- Junlong Da
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Ying Li
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Kai Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Junyu Ren
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jianqun Wang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xinpeng Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Xiaoyao Liu
- Department of Orthodontics, the First Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Jiahui Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Lixue Liu
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Wenxuan Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Shujian Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Yuyao Guo
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China
| | - Bin Zhang
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Heilongjiang Academy of Medical Sciences, Harbin, People’s Republic of China
| | - Han Jin
- Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, People’s Republic of China,Correspondence: Han Jin; Bin Zhang, Heilongjiang Provincial Key Laboratory of Hard Tissue Development and Regeneration, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People’s Republic of China, Tel/Fax +86 0451-86297231, Email ;
| |
Collapse
|
25
|
Ye X, Chen X, He R, Meng W, Chen W, Wang F, Meng X. Enhanced anti-breast cancer efficacy of co-delivery liposomes of docetaxel and curcumin. Front Pharmacol 2022; 13:969611. [PMID: 36324685 PMCID: PMC9618653 DOI: 10.3389/fphar.2022.969611] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/27/2022] [Indexed: 08/28/2023] Open
Abstract
The successful treatment of breast cancer is hampered by toxicity to normal cells, impaired drug accumulation at the tumor site, and multidrug resistance. We designed a novel multifunctional liposome, CUR-DTX-L, to co-deliver curcumin (CUR) and the chemotherapeutic drug docetaxel (DTX) for the treatment of breast cancer in order to address multidrug resistance (MDR) and the low efficacy of chemotherapy. The mean particle size, polydispersity index, zeta potential, and encapsulation efficiency of CUR-DTX-L were 208.53 ± 6.82 nm, 0.055 ± 0.001, -23.1 ± 2.1 mV, and 98.32 ± 2.37%, respectively. An in vitro release study and CCK-8 assays showed that CUR-DTX-L has better sustained release effects and antitumor efficacy than free drugs, the antitumor efficacy was verified by MCF-7 tumor-bearing mice, the CUR-DTX-L showed better antitumor efficacy than other groups, and the in vivo pharmacokinetic study indicated that the plasma concentration-time curve, mean residence time, and biological half-life time of CUR-DTX-L were significantly increased compared with free drugs, suggesting that it is a promising drug delivery system for the synergistic treatment of breast cancer.
Collapse
Affiliation(s)
- Xi Ye
- Department of Pharmacy, The Second People’s Hospital of Hefei, Hefei, China
- Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
- Hefei Hospital Affiliated to Bengbu Medical College, Hefei, China
| | - Xin Chen
- Department of Pharmacy, Anhui Provincial Crops Hospital, Hefei, China
| | - Ruixi He
- Anhui University of Chinese Medicine, Hefei, China
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, China
| | - Wangyang Meng
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Weidong Chen
- Institute of Drug Metabolism, School of Pharmaceutical Sciences, Anhui University of Chinese Medicine, Hefei, China
| | - Fengling Wang
- Department of Pharmacy, The Second People’s Hospital of Hefei, Hefei, China
- Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
- Hefei Hospital Affiliated to Bengbu Medical College, Hefei, China
| | - Xiangyun Meng
- Department of Pharmacy, The Second People’s Hospital of Hefei, Hefei, China
- Hefei Hospital Affiliated to Anhui Medical University, Hefei, China
- Hefei Hospital Affiliated to Bengbu Medical College, Hefei, China
| |
Collapse
|
26
|
Yadav P, Ambudkar SV, Rajendra Prasad N. Emerging nanotechnology-based therapeutics to combat multidrug-resistant cancer. J Nanobiotechnology 2022; 20:423. [PMID: 36153528 PMCID: PMC9509578 DOI: 10.1186/s12951-022-01626-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/07/2022] [Indexed: 11/17/2022] Open
Abstract
Cancer often develops multidrug resistance (MDR) when cancer cells become resistant to numerous structurally and functionally different chemotherapeutic agents. MDR is considered one of the principal reasons for the failure of many forms of clinical chemotherapy. Several factors are involved in the development of MDR including increased expression of efflux transporters, the tumor microenvironment, changes in molecular targets and the activity of cancer stem cells. Recently, researchers have designed and developed a number of small molecule inhibitors and derivatives of natural compounds to overcome various mechanisms of clinical MDR. Unfortunately, most of the chemosensitizing approaches have failed in clinical trials due to non-specific interactions and adverse side effects at pharmacologically effective concentrations. Nanomedicine approaches provide an efficient drug delivery platform to overcome the limitations of conventional chemotherapy and improve therapeutic effectiveness. Multifunctional nanomaterials have been found to facilitate drug delivery by improving bioavailability and pharmacokinetics, enhancing the therapeutic efficacy of chemotherapeutic drugs to overcome MDR. In this review article, we discuss the major factors contributing to MDR and the limitations of existing chemotherapy- and nanocarrier-based drug delivery systems to overcome clinical MDR mechanisms. We critically review recent nanotechnology-based approaches to combat tumor heterogeneity, drug efflux mechanisms, DNA repair and apoptotic machineries to overcome clinical MDR. Recent successful therapies of this nature include liposomal nanoformulations, cRGDY-PEG-Cy5.5-Carbon dots and Cds/ZnS core–shell quantum dots that have been employed for the effective treatment of various cancer sub-types including small cell lung, head and neck and breast cancers.
Collapse
|
27
|
Recent Trends in Nanomedicine-Based Strategies to Overcome Multidrug Resistance in Tumors. Cancers (Basel) 2022; 14:cancers14174123. [PMID: 36077660 PMCID: PMC9454760 DOI: 10.3390/cancers14174123] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022] Open
Abstract
Cancer is the leading cause of economic and health burden worldwide. The commonly used approaches for the treatment of cancer are chemotherapy, radiotherapy, and surgery. Chemotherapy frequently results in undesirable side effects, and cancer cells may develop resistance. Combating drug resistance is a challenging task in cancer treatment. Drug resistance may be intrinsic or acquired and can be due to genetic factors, growth factors, the increased efflux of drugs, DNA repair, and the metabolism of xenobiotics. The strategies used to combat drug resistance include the nanomedicine-based targeted delivery of drugs and genes using different nanocarriers such as gold nanoparticles, peptide-modified nanoparticles, as well as biomimetic and responsive nanoparticles that help to deliver payload at targeted tumor sites and overcome resistance. Gene therapy in combination with chemotherapy aids in this respect. siRNA and miRNA alone or in combination with chemotherapy improve therapeutic response in tumor cells. Some natural substances, such as curcumin, quercetin, tocotrienol, parthenolide, naringin, and cyclosporin-A are also helpful in combating the drug resistance of cancer cells. This manuscript summarizes the mechanism of drug resistance and nanoparticle-based strategies used to combat it.
Collapse
|
28
|
Younes M, Mardirossian R, Rizk L, Fazlian T, Khairallah JP, Sleiman C, Naim HY, Rizk S. The Synergistic Effects of Curcumin and Chemotherapeutic Drugs in Inhibiting Metastatic, Invasive and Proliferative Pathways. PLANTS 2022; 11:plants11162137. [PMID: 36015440 PMCID: PMC9414747 DOI: 10.3390/plants11162137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 08/14/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022]
Abstract
Curcumin, the main phytochemical identified from the Curcuma longa L. family, is one of the spices used in alternative medicine worldwide. It has exhibited a broad range of pharmacological activities as well as promising effects in the treatment of multiple cancer types. Moreover, it has enhanced the activity of other chemotherapeutic drugs and radiotherapy by promoting synergistic effects in the regulation of various cancerous pathways. Despite all the literature addressing the molecular mechanism of curcumin on various cancers, no review has specifically addressed the molecular mechanism underlying the effect of curcumin in combination with therapeutic drugs on cancer metastasis. The current review assesses the synergistic effects of curcumin with multiple drugs and light radiation, from a molecular perspective, in the inhibition of metastasis, invasion and proliferation. A systemic review of articles published during the past five years was performed using MEDLINE/PubMed and Scopus. The assessment of these articles evidenced that the combination therapy with various drugs, including doxorubicin, 5-fluorouracil, paclitaxel, berberine, docetaxel, metformin, gemcitabine and light radiation therapy on various types of cancer, is capable of ameliorating different metastatic pathways that are presented and evaluated. However, due to the heterogeneity of pathways and proteins in different cell lines, more research is needed to confirm the root causes of these pathways.
Collapse
Affiliation(s)
- Maria Younes
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Rita Mardirossian
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Liza Rizk
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Tia Fazlian
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Jean Paul Khairallah
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Christopher Sleiman
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
| | - Hassan Y. Naim
- Department of Biochemistry, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
- Correspondence: (H.Y.N.); (S.R.)
| | - Sandra Rizk
- Department of Natural Sciences, School of Arts and Sciences, Lebanese American University, Byblos P.O. Box 36, Lebanon
- Correspondence: (H.Y.N.); (S.R.)
| |
Collapse
|
29
|
Gao Q, Feng J, Liu W, Wen C, Wu Y, Liao Q, Zou L, Sui X, Xie T, Zhang J, Hu Y. Opportunities and challenges for co-delivery nanomedicines based on combination of phytochemicals with chemotherapeutic drugs in cancer treatment. Adv Drug Deliv Rev 2022; 188:114445. [PMID: 35820601 DOI: 10.1016/j.addr.2022.114445] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 02/08/2023]
Abstract
The therapeutic limitations such as insufficient efficacy, drug resistance, metastasis, and undesirable side effects are frequently caused by the long duration monotherapy based on chemotherapeutic drugs. multiple combinational anticancer strategies such as nucleic acids combined with chemotherapeutic agents, chemotherapeutic combinations, chemotherapy and tumor immunotherapy combinations have been embraced, holding great promise to counter these limitations, while still taking including some potential risks. Nowadays, an increasing number of research has manifested the anticancer effects of phytochemicals mediated by modulating cancer cellular events directly as well as the tumor microenvironment. Specifically, these natural compounds exhibited suppression of cancer cell proliferation, apoptosis, migration and invasion of cancer cells, P-glycoprotein inhibition, decreasing vascularization and activation of tumor immunosuppression. Due to the low toxicity and multiple modulation pathways of these phytochemicals, the combination of chemotherapeutic agents with natural compounds acts as a novel approach to cancer therapy to increase the efficiency of cancer treatments as well as reduce the adverse consequences. In order to achieve the maximized combination advantages of small-molecule chemotherapeutic drugs and natural compounds, a variety of functional nano-scaled drug delivery systems, such as liposomes, host-guest supramolecules, supramolecules, dendrimers, micelles and inorganic systems have been developed for dual/multiple drug co-delivery. These co-delivery nanomedicines can improve pharmacokinetic behavior, tumor accumulation capacity, and achieve tumor site-targeting delivery. In that way, the improved antitumor effects through multiple-target therapy and reduced side effects by decreasing dose can be implemented. Here, we present the synergistic anticancer outcomes and the related mechanisms of the combination of phytochemicals with small-molecule anticancer drugs. We also focus on illustrating the design concept, and action mechanisms of nanosystems with co-delivery of drugs to synergistically improve anticancer efficacy. In addition, the challenges and prospects of how these insights can be translated into clinical benefits are discussed.
Collapse
Affiliation(s)
- Quan Gao
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiao Feng
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wencheng Liu
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chengyong Wen
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Xinbing Sui
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Tian Xie
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China.
| |
Collapse
|
30
|
Zhao M, Zhang M, Yu Q, Fei W, Li T, Zhu L, Yao Y, Zheng C, Zhang X. Hyaluronic Acid-Modified Nanoplatforms as a Vector for Targeted Delivery of Autophagy-Related Gene to the Endometriotic Lesions in Mice. Front Bioeng Biotechnol 2022; 10:918368. [PMID: 35845410 PMCID: PMC9283728 DOI: 10.3389/fbioe.2022.918368] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/23/2022] [Indexed: 12/15/2022] Open
Abstract
This investigation probed endometriosis treatment using targeted nanoparticles (NPs) to modulate autophagic activity. To that end, a novel form of polymer-based NP gene delivery platform consisting of polyethyleneimine (PEI) conjugated to stearic acid (SA) and nucleotides (DNA/siRNAs) and enclosed by hyaluronic acid (HA) was prepared. CD44 is highly upregulated in cystic lesions, and HA–CD44 binding in this specific nanoplatform was used to achieve targeted drug delivery to CD44-expression endometriotic tissues. The expression of autophagy-related genes was modulated to explore the importance of this process in the development of endometriosis. By inducing autophagic activity, we were able to reduce the size of endometriotic cysts and suppress the development of ectopic endometrium. To further confirm the relationship between autophagic activity and this disease in humans and animals, numbers of autophagic vesicles and autophagic protein expression were assessed in lesion tissue samples from patients, revealing there may be consistency between animal and human data. Overall, these data revealed the ability of this (PEI–SA/DNA) HA gene delivery system to regulate autophagic activity and, thereby, aid in the treatment of endometriosis.
Collapse
|
31
|
Sritharan S, Guha S, Hazarika S, Sivalingam N. Meta analysis of bioactive compounds, miRNA, siRNA and cell death regulators as sensitizers to doxorubicin induced chemoresistance. Apoptosis 2022; 27:622-646. [PMID: 35716277 DOI: 10.1007/s10495-022-01742-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2022] [Indexed: 11/02/2022]
Abstract
Cancer has presented to be the most challenging disease, contributing to one in six mortalities worldwide. The current treatment regimen involves multiple rounds of chemotherapy administration, alone or in combination. The treatment has adverse effects including cardiomyopathy, hepatotoxicity, and nephrotoxicity. In addition, the development of resistance to chemo has been attributed to cancer relapse and low patient overall survivability. Multiple drug resistance development may be through numerous factors such as up-regulation of drug transporters, drug inactivation, alteration of drug targets and drug degradation. Doxorubicin is a widely used first line chemotherapeutic drug for a myriad of cancers. It has multiple intracellular targets, DNA intercalation, adduct formation, topoisomerase inhibition, iron chelation, reactive oxygen species generation and promotes immune mediated clearance of the tumor. Agents that can sensitize the resistant cancer cells to the chemotherapeutic drug are currently the focus to improve the clinical efficiency of cancer therapy. This review summarizes the recent 10-year research on the use of natural phytochemicals, inhibitors of apoptosis and autophagy, miRNAs, siRNAs and nanoformulations being investigated for doxorubicin chemosensitization.
Collapse
Affiliation(s)
- Sruthi Sritharan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Sampurna Guha
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Snoopy Hazarika
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India
| | - Nageswaran Sivalingam
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, Faculty of Engineering and Technology, SRM Institute of Science and Technology, SRM Nagar, Kattankulathur, Chengalpattu District, Chennai, Tamil Nadu, 603203, India.
| |
Collapse
|
32
|
Using GPCRs as Molecular Beacons to Target Ovarian Cancer with Nanomedicines. Cancers (Basel) 2022; 14:cancers14102362. [PMID: 35625966 PMCID: PMC9140059 DOI: 10.3390/cancers14102362] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/03/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
The five-year survival rate for women with ovarian cancer is very poor despite radical cytoreductive surgery and chemotherapy. Although most patients initially respond to platinum-based chemotherapy, the majority experience recurrence and ultimately develop chemoresistance, resulting in fatal outcomes. The current administration of cytotoxic compounds is hampered by dose-limiting severe adverse effects. There is an unmet clinical need for targeted drug delivery systems that transport chemotherapeutics selectively to tumor cells while minimizing off-target toxicity. G protein-coupled receptors (GPCRs) are the largest family of membrane receptors, and many are overexpressed in solid tumors, including ovarian cancer. This review summarizes the progress in engineered nanoparticle research for drug delivery for ovarian cancer and discusses the potential use of GPCRs as molecular entry points to deliver anti-cancer compounds into ovarian cancer cells. A newly emerging treatment paradigm could be the personalized design of nanomedicines on a case-by-case basis.
Collapse
|
33
|
Fahira AI, Amalia R, Barliana MI, Gatera VA, Abdulah R. Polyethyleneimine (PEI) as a Polymer-Based Co-Delivery System for Breast Cancer Therapy. BREAST CANCER (DOVE MEDICAL PRESS) 2022; 14:71-83. [PMID: 35422657 PMCID: PMC9005234 DOI: 10.2147/bctt.s350403] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Accepted: 03/15/2022] [Indexed: 11/24/2022]
Abstract
Cancer has become one of the leading causes of morbidity and mortality worldwide. This disease is classified broadly by tissue, organ, and system; different cancer types and subtypes require different treatments. Drug bioavailability, selectivity, and high dosage, as well as extended treatment, are significantly associated with the development of resistance – a complex problem in cancer therapy. It is expected that the combination of anticancer drugs and drug delivery systems, using polymers to increase the access of such agents to their site of action, will improve the efficacy of therapy. Polyethyleneimine (PEI) is a polymer used as a co-delivery system for anticancer drugs and gene therapy. PEI is also useful for other purposes, such as transfection and bio-adsorbent agents. In co-delivery, PEI can promote drug internalization. However, PEI with a high molecular weight is linked to higher cytotoxicity, thus requiring further evaluation of clinical safety. This review focuses on the utilization of PEI as a co-delivery system for anticancer therapy, as well as its potential to overcome resistance, particularly in the treatment of specific subtypes (eg, breast cancer). In conclusion, PEI has promising applications and is improvable for the development of anticancer drugs.
Collapse
Affiliation(s)
- Alistia Ilmiah Fahira
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Riezki Amalia
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia.,Center of Excellence in Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Melisa Intan Barliana
- Center of Excellence in Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia.,Department of Biology Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Vesara Ardhe Gatera
- Center of Excellence in Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia.,Department of Biology Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Rizky Abdulah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia.,Center of Excellence in Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
34
|
Nguyen NT, Bui QA, Huynh PD, Nguyen QH, Tran NQ, Viet NT, Nguyen DT. Curcumin and Paclitaxel co-Loaded Heparin and Poloxamer P403 Hybrid Nanocarrier for Improved Synergistic Efficacy in Breast Cancer. Curr Drug Deliv 2022; 19:966-979. [DOI: 10.2174/1567201819666220401095923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 11/22/2022]
Abstract
Introduction:
Multi-drug nanosystem has been employed in several therapeutic models due to the synergistic effect of the drugs and/or bioactive compounds, which help in tumor-targeting and limit usual side effects of chemotherapy.
Methods:
In this research, we developed the amphiphilic Heparin-Poloxamer P403 (HSP) nanogel that can load curcumin (CUR) and Paclitaxel (PTX) through the hydrophobic core of Poloxamer P403. The features of HSP nanogel are assessed through Fourier-transform infrared spectroscopy (FT-IR), transmission electron microscopy (TEM), differential light scattering (DLS), and critical micelle concentration (CMC). Nanogel and its duel-loaded platform show high stability and spherical morphology.
Results:
The drug release profile indicates fast release at pH 5.5, suggesting effective drug distribution at the tumor site. In vitro research confirms lower cytotoxicity of HSP@CUR@PTX compared with free PTX and higher inhibition effect with MCF-7 than HSP@PTX. These results support the synergism between PTX and CUR.
Conclusion,:
HSP@CUR@PTX suggests a prominent strategy for achieving the synergistic effect of PTX and CUR to circumvent undesirable effects in breast cancer treatment.
Collapse
Affiliation(s)
- Ngoc The Nguyen
- Faculty of Medicine - Pharmacy, Tra Vinh University, Tra Vinh City, Vietnam
| | - Quynh Anh Bui
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| | - Phuong Duy Huynh
- Faculty of Medicine - Pharmacy, Tra Vinh University, Tra Vinh City, Vietnam
| | | | - Ngoc Quyen Tran
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam;
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi City, Vietnam
| | - Nguyen Thanh Viet
- Institute of Environmental Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City, Vietnam
| | - Dinh Trung Nguyen
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, Ho Chi Minh City, Vietnam
| |
Collapse
|
35
|
Curcumin Inhibits the Proliferation of Renal Cancer 786-O Cells through MTOR Signaling Pathway and Its Mechanism. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:1842389. [PMID: 35399832 PMCID: PMC8986413 DOI: 10.1155/2022/1842389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/10/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Objectives. The mechanism of curcumin inhibiting renal cancer 786-O cells proliferation through MTOR signaling pathway was investigated. Methods. Human renal cancer 786-O cells were cultured with curcumin for 48 h. The OD values were measured by the MTT method, and the growth inhibition rate of 786-O cells was calculated. The cell cycle distribution and apoptosis rate were detected by flow cytometry (FCM). Transwell chamber was introduced to detect cell invasion ability. Cell migration ability was detected by the cell scratch test. The protein expression was assessed by Western blot. Results. With curcumin concentration increasing, the expressions of MMP2, MMP9, MTOR, and p-MTOR proteins and the number of cells in the S phase decreased gradually, while number of cells in G1 and G2/M phases and cells apoptosis rate increased continuously. With the increasing of concentration and time, growth of 786-O cells in each treatment group was inhibited to varying degrees. The higher the inhibition rate was, the cells migration and transmembrane cells proportion decreased significantly. Conclusions. Curcumin inhibits the proliferation, migration, and invasion and induces apoptosis of renal cancer 786-O cells by blocking the MTOR signaling pathway. It may be related to the downregulation of MMP2 and MMP9 proteins.
Collapse
|
36
|
Mohamadian M, Bahrami A, Moradi Binabaj M, Asgharzadeh F, Ferns GA. Molecular Targets of Curcumin and Its Therapeutic Potential for Ovarian Cancer. Nutr Cancer 2022; 74:2713-2730. [PMID: 35266849 DOI: 10.1080/01635581.2022.2049321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Ovarian cancer is the fifth most common gynecological cancer in women globally. Conventional chemotherapy is the first therapeutic approach in the treatment of ovarian cancer, but its success is limited by severe side effects, transient response, and the high prevalence of relapse. Curcumin is a natural product found in the rhizome extract of Curcuma longa and has been extensively used over the last decades for its unique biological and medicinal properties, which include: having antioxidant, analgesic, anti-inflammation, and anti-tumor activities. Curcumin exerts its anticancer properties against ovarian cancer via multiple mechanisms: interfering with cellular interactions necessary for metastasis and recurrence of OC cells, increasing pro-apoptotic proteins as well as inducing or suppressing generation of different molecules such as cytokines, transcription factors, enzymes, protein kinases, and growth factors. Moreover, curcumin down-regulates various signaling pathways such as PI3K/Akt, Wnt/β-catenin, JAK/STAT3, and MEK/ERK1/2 axes, which at least in part have a role in inhibiting further tumor proliferation, growth, and angiogenesis. In this review, we overview the potential of incorporating curcumin into the treatment of ovarian cancer. In particular, we summarize the preclinical evidence supporting its use in combination with current chemotherapeutic regimens as well as new analogues and formulations under investigation.
Collapse
Affiliation(s)
- Malihe Mohamadian
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Afsane Bahrami
- Clinical Research Development Unit, Imam Reza Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Clinical Research Development Unit of Akbar Hospital, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Moradi Binabaj
- Cellular and Molecular Research Center, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Fereshteh Asgharzadeh
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Department of Medical Education, Falmer, Brighton, Sussex, UK
| |
Collapse
|
37
|
Pang L, Zhang L, Zhou H, Cao L, Shao Y, Li T. Reactive Oxygen Species-Responsive Nanococktail With Self-Amplificated Drug Release for Efficient Co-Delivery of Paclitaxel/Cucurbitacin B and Synergistic Treatment of Gastric Cancer. Front Chem 2022; 10:844426. [PMID: 35308794 PMCID: PMC8931329 DOI: 10.3389/fchem.2022.844426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 02/03/2022] [Indexed: 12/21/2022] Open
Abstract
Application of drug combinations is a powerful strategy for the therapy of advanced gastric cancer. However, the clinical use of such combinations is greatly limited by the occurrence of severe systemic toxicity. Although polymeric-prodrug-based nanococktails can significantly reduce toxicity of drugs, they have been shown to have low intracellular drug release. To balance between efficacy and safety during application of polymeric-prodrug-based nanococktails, a reactive oxygen species (ROS)-responsive nanococktail (PCM) with self-amplification drug release was developed in this study. In summary, PCM micelles were co-assembled from ROS-sensitive cucurbitacin B (CuB) and paclitaxel (PTX) polymeric prodrug, which were fabricated by covalently grafting PTX and CuB to dextran via an ROS-sensitive linkage. To minimize the side effects of the PCM micelles, a polymeric-prodrug strategy was employed to prevent premature leakage. Once it entered cancer cells, PCM released CuB and PTX in response to ROS. Moreover, the released CuB further promoted ROS generation, which in turn enhanced drug release for better therapeutic effects. In vivo antitumor experiments showed that the PCM-treated group had lower tumor burden (tumor weight was reduced by 92%), but bodyweight loss was not significant. These results indicate that the developed polymeric prodrug, with a self-amplification drug release nanococktail strategy, can be an effective and safe strategy for cancer management.
Collapse
Affiliation(s)
- Lijun Pang
- Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Lei Zhang
- Department of Pharmacy, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Hong Zhou
- Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Ling Cao
- Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Yueqin Shao
- Department of Oncology, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Tengyun Li
- Department of Pharmacy, The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Tengyun Li,
| |
Collapse
|
38
|
Zhang C, Zhou X, Zhang H, Han X, Li B, Yang R, Zhou X. Recent Progress of Novel Nanotechnology Challenging the Multidrug Resistance of Cancer. Front Pharmacol 2022; 13:776895. [PMID: 35237155 PMCID: PMC8883114 DOI: 10.3389/fphar.2022.776895] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Multidrug resistance (MDR) of tumors is one of the clinical direct reasons for chemotherapy failure. MDR directly leads to tumor recurrence and metastasis, with extremely grievous mortality. Engineering a novel nano-delivery system for the treatment of MDR tumors has become an important part of nanotechnology. Herein, this review will take those different mechanisms of MDR as the classification standards and systematically summarize the advances in nanotechnology targeting different mechanisms of MDR in recent years. However, it still needs to be seriously considered that there are still some thorny problems in the application of the nano-delivery system against MDR tumors, including the excessive utilization of carrier materials, low drug-loading capacity, relatively narrow targeting mechanism, and so on. It is hoped that through the continuous development of nanotechnology, nano-delivery systems with more universal uses and a simpler preparation process can be obtained, for achieving the goal of defeating cancer MDR and accelerating clinical transformation.
Collapse
Affiliation(s)
- Chengyuan Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Xuemei Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Hanyi Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Xuanliang Han
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Baijun Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Ran Yang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, China
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing, China
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
39
|
Maleki Dizaj S, Alipour M, Dalir Abdolahinia E, Ahmadian E, Eftekhari A, Forouhandeh H, Rahbar Saadat Y, Sharifi S, Zununi Vahed S. Curcumin nanoformulations: Beneficial nanomedicine against cancer. Phytother Res 2022; 36:1156-1181. [PMID: 35129230 DOI: 10.1002/ptr.7389] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 01/11/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022]
Abstract
Curcumin is a phytochemical achieved from the plant turmeric. It is extensively utilized for the treatment of several types of diseases such as cancers. Nevertheless, its efficiency has been limited because of rapid metabolism, low bioavailability, poor water solubility, and systemic elimination. Scientists have tried to solve these problems by exploring novel drug delivery systems such as lipid-based nanoparticles (NPs) (e.g., solid lipid NPs, nanostructured lipid carriers, and liposomes), polymeric NPs, micelles, nanogels, cyclodextrin, gold, and mesoporous silica NPs. Among these, liposomes have been the most expansively studied. This review mainly focuses on the different curcumin nanoformulations and their use in cancer therapy in vitro, in vivo, and clinical studies. Despite the development of curcumin-containing NPs for the treatment of cancer, potentially serious side effects, including interactions with other drugs, some toxicity aspects of NPs may occur that require more high-quality investigations to firmly establish the clinical efficacy.
Collapse
Affiliation(s)
- Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Dental Biomaterials, Faculty of Dentistry, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elaheh Dalir Abdolahinia
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Ahmadian
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aziz Eftekhari
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Health innovation and acceleration center, Tabriz University of Medical Sciences, Tabriz, Iran.,Russian Institute for Advanced Study, Moscow State Pedagogical University, Moscow, Russian Federation
| | - Haleh Forouhandeh
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Simin Sharifi
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
40
|
Yang J, He Q, Wang Y, Pan Z, Zhang G, Liang J, Su L, Wang A, Zeng C, Luo H, Liu L, Li J, Rao Q, Wang B, Wang H, Chen P. Gegen Qinlian Decoction ameliorates type 2 diabetes osteoporosis via IGFBP3/MAPK/NFATc1 signaling pathway based on cytokine antibody array. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 94:153810. [PMID: 34798519 DOI: 10.1016/j.phymed.2021.153810] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Osteoporosis affects more than half the patients with type 2 diabetes mellitus (T2DM). Up to data, there is no effective clinical practice in managing type 2 diabetes osteoporosis (T2DOP) because of its complex pathogenesis. Gegen Qinlian Decoction (GQD) has been used for the long-term management of T2DM. However, the underlying mechanism of GQD in the treatment of T2DOP remains unknown. PURPOSE To reveal the role of GQD in T2DOP and its potential therapeutic targets in the management of T2DOP. STUDY DESIGN The effect of GQD on T2DOP was observed in db/db mice in four groups: model group, GQD low-dose group (GQD-L), GQD high-dose group (GQD-H), and metformin (positive control) group. C57BL/6J mice were used as the negative control group. METHODS Quantitative phytochemical analysis of GQD was performed using high-performance liquid chromatography (HPLC). Micro-CT and hematoxylin-eosin (H&E) staining were used to evaluate bone histomorphometry. To screen for candidate targets of GQD, a cytokine antibody array was used, followed by bioinformatics analysis. Quantitative real-time PCR (qRT-PCR) and western blotting (WB) were used to determine expression levels. RESULTS The major active components of GQD were confirmed by HPLC. Micro-CT and H&E staining showed that bone mass was significantly increased in the GQD-H group compared with the model group. Antibody arrays revealed that the expression of insulin-like growth factor binding protein 3 (IGFBP3) was elevated in the GQD-H group. The MAPK pathway was identified using bioinformatics analysis. Additionally, the levels of osteoclastogenesis-related genes, including cathepsin K (Ctsk), acid phosphatase 5 (Acp5), matrix metallopeptidase 9 (Mmp9), and ATPase H+ transporting V0 subunit D2 (Atp6v0d2) were significantly decreased in the GQD-H group. Compared with the model group, high-dosage GQD inhibited phosphorylation of extracellular signal-regulated kinases (ERKs) and P38 mitogen-activated protein kinase (MAPK) and the expression of c-Fos and nuclear factor of activated T cells 1 (NFATc1). CONCLUSION GQD plays a protective role in T2DOP by upregulating IGFBP3 expression and downregulating the IGFBP3/MAPK/NFATc1 signaling pathway. IGFBP3 in serum may also be a novel biomarker in the treatment of T2DOP. Our current findings not only expand the application of GQD, but also provide a theoretical basis and guidance for T2DOP.
Collapse
Affiliation(s)
- Junzheng Yang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Qi He
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Yunhan Wang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Zhaofeng Pan
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Gangyu Zhang
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Jianming Liang
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Lijun Su
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Ailin Wang
- Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Chuning Zeng
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Haoran Luo
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Lingyun Liu
- College of Basic Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Jianliang Li
- First School of Clinical Medicine, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China; The Laboratory of Orthopaedics and Traumatology of Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Qiuhong Rao
- Department of Pharmacy, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China
| | - Baohua Wang
- Department of Endocrinology, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, PR China.
| | - Haibin Wang
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou, Guangdon 510405, PR China.
| | - Peng Chen
- Department of Orthopaedic Surgery, The First Affiliated Hospital, Guangzhou University of Chinese Medicine, 16 Jichang Road, Baiyun District, Guangzhou, Guangdon 510405, PR China.
| |
Collapse
|
41
|
Rezaei-Tazangi F, Roghani-Shahraki H, Khorsand Ghaffari M, Abolhasani Zadeh F, Boostan A, ArefNezhad R, Motedayyen H. The Therapeutic Potential of Common Herbal and Nano-Based Herbal Formulations against Ovarian Cancer: New Insight into the Current Evidence. Pharmaceuticals (Basel) 2021; 14:1315. [PMID: 34959716 PMCID: PMC8705681 DOI: 10.3390/ph14121315] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
Ovarian cancer (OCa) is characterized as one of the common reasons for cancer-associated death in women globally. This gynecological disorder is chiefly named the "silent killer" due to lacking an association between disease manifestations in the early stages and OCa. Because of the disease recurrence and resistance to common therapies, discovering an effective therapeutic way against the disease is a challenge. According to documents, some popular herbal formulations, such as curcumin, quercetin, and resveratrol, can serve as an anti-cancer agent through different mechanisms. However, these herbal products may be accompanied by some pharmacological limitations, such as poor bioavailability, instability, and weak water solubility. On the contrary, using nano-based material, e.g., nanoparticles (NPs), micelles, liposomes, can significantly solve these limitations. Therefore, in the present study, we will summarize the anti-cancer aspects of these herbal and-nano-based herbal formulations with a focus on their mechanisms against OCa.
Collapse
Affiliation(s)
- Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa 7345149573, Iran;
| | | | - Mahdi Khorsand Ghaffari
- Department of Physiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz 1433671348, Iran;
| | - Firoozeh Abolhasani Zadeh
- Department of Surgery, Faculty of Medicine, Kerman University of Medical Sciences, Kerman 7616913555, Iran;
| | - Aynaz Boostan
- Department of Obstetrics & Gynecology, Saveh Chamran Hospital, Saveh 3919676651, Iran;
| | - Reza ArefNezhad
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz 1433671348, Iran
| | - Hossein Motedayyen
- Autoimmune Diseases Research Center, Kashan University of Medical Sciences, Kashan 8715973474, Iran
| |
Collapse
|
42
|
Gavas S, Quazi S, Karpiński TM. Nanoparticles for Cancer Therapy: Current Progress and Challenges. NANOSCALE RESEARCH LETTERS 2021; 16:173. [PMID: 34866166 PMCID: PMC8645667 DOI: 10.1186/s11671-021-03628-6] [Citation(s) in RCA: 277] [Impact Index Per Article: 92.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 11/19/2021] [Indexed: 05/04/2023]
Abstract
Cancer is one of the leading causes of death and morbidity with a complex pathophysiology. Traditional cancer therapies include chemotherapy, radiation therapy, targeted therapy, and immunotherapy. However, limitations such as lack of specificity, cytotoxicity, and multi-drug resistance pose a substantial challenge for favorable cancer treatment. The advent of nanotechnology has revolutionized the arena of cancer diagnosis and treatment. Nanoparticles (1-100 nm) can be used to treat cancer due to their specific advantages such as biocompatibility, reduced toxicity, more excellent stability, enhanced permeability and retention effect, and precise targeting. Nanoparticles are classified into several main categories. The nanoparticle drug delivery system is particular and utilizes tumor and tumor environment characteristics. Nanoparticles not only solve the limitations of conventional cancer treatment but also overcome multidrug resistance. Additionally, as new multidrug resistance mechanisms are unraveled and studied, nanoparticles are being investigated more vigorously. Various therapeutic implications of nanoformulations have created brand new perspectives for cancer treatment. However, most of the research is limited to in vivo and in vitro studies, and the number of approved nanodrugs has not much amplified over the years. This review discusses numerous types of nanoparticles, targeting mechanisms, and approved nanotherapeutics for oncological implications in cancer treatment. Further, we also summarize the current perspective, advantages, and challenges in clinical translation.
Collapse
Affiliation(s)
- Shreelaxmi Gavas
- Department of Life Sciences, GenLab Biosolutions Private Limited, Bangalore, Karnataka 560043 India
| | - Sameer Quazi
- GenLab Biosolutions Private Limited, Bangalore, Karnataka 560043 India
| | - Tomasz M. Karpiński
- Chair and Department of Medical Microbiology, Poznań University of Medical Sciences, Wieniawskiego 3, 61-712 Poznań, Poland
| |
Collapse
|
43
|
Fraser B, Peters AE, Sutherland JM, Liang M, Rebourcet D, Nixon B, Aitken RJ. Biocompatible Nanomaterials as an Emerging Technology in Reproductive Health; a Focus on the Male. Front Physiol 2021; 12:753686. [PMID: 34858208 PMCID: PMC8632065 DOI: 10.3389/fphys.2021.753686] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 10/06/2021] [Indexed: 12/24/2022] Open
Abstract
A growing body of research has confirmed that nanoparticle (NP) systems can enhance delivery of therapeutic and imaging agents as well as prevent potentially damaging systemic exposure to these agents by modifying the kinetics of their release. With a wide choice of NP materials possessing different properties and surface modification options with unique targeting agents, bespoke nanosystems have been developed for applications varying from cancer therapeutics and genetic modification to cell imaging. Although there remain many challenges for the clinical application of nanoparticles, including toxicity within the reproductive system, some of these may be overcome with the recent development of biodegradable nanoparticles that offer increased biocompatibility. In recognition of this potential, this review seeks to present recent NP research with a focus on the exciting possibilities posed by the application of biocompatible nanomaterials within the fields of male reproductive medicine, health, and research.
Collapse
Affiliation(s)
- Barbara Fraser
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Alexandra E Peters
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Jessie M Sutherland
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Mingtao Liang
- Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia.,Priority Research Centre for Reproductive Science, School of Biomedical Science and Pharmacy, University of Newcastle, Callaghan, NSW, Australia
| | - Diane Rebourcet
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Brett Nixon
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Robert J Aitken
- Priority Research Centre for Reproductive Science, University of Newcastle, Callaghan, NSW, Australia.,Pregnancy and Reproduction Program, Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
44
|
Curcumin Administered in Combination with Glu-GNPs Induces Radiosensitivity in Transplanted Tumor MDA-MB-231-luc Cells in Nude Mice. BIOMED RESEARCH INTERNATIONAL 2021; 2021:9262453. [PMID: 34825004 PMCID: PMC8610687 DOI: 10.1155/2021/9262453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 10/11/2021] [Indexed: 12/24/2022]
Abstract
Curcumin is a type of plant polyphenol extracted from Curcuma longa L. rhizome, which demonstrates antitumor activity in breast cancer cells in vitro. To investigate the combined effect and possible mechanism of curcumin and glucose-gold nanoparticles (Glu-GNPs), the radiosensitivity of breast carcinoma xenografts was assessed in nude mice. MDA-MB-231 cells labeled with firefly luciferase were inoculated into the mammary fatty pads of nude mice to establish a transplantation tumor model of human breast cancer. The tumor-bearing mice were treated with different drugs (curcumin, Glu-GNPs, and cisplatin) for 3 weeks prior to radiotherapy. The body weights and tumor volumes of the mice were measured in regular intervals. Tumor bioluminescence intensity was determined in real-time using an in vivo bioluminescence imaging system to monitor tumor growth. Transplanted tumor tissue samples were taken for hematoxylin and eosin (HE) staining, and the expression of VEGF, HSP90, HIF-1α, and MMP9 was evaluated via reverse transcription-quantitative PCR or immunohistochemistry. The results revealed that the breast tumor-bearing nude mouse model was successfully established, as evidenced by a stable expression of luciferase. Curcumin inhibited the growth of tumors without causing significant weight loss in mice. Furthermore, additive inhibition was demonstrated when curcumin was administered in combination with Glu-GNPs and irradiation. Tumor bioluminescence intensity was decreased in the model group following curcumin, Glu-GNPs, and irradiation treatment. HE staining demonstrated that transplanted tumors were malignant, with necrotic tissue exhibited centrally. It was concluded that curcumin administered in combination with Glu-GNPs and X-ray irradiation could reduce the protein expression of VEGF, HSP90, HIF-1α, and MMP9 in tumor tissue when compared with the model group. Curcumin and Glu-GNPs administered with X-ray irradiation significantly inhibited tumor growth and induced radiosensitivity, which may be associated with the inhibition of angiogenesis in tumor tissue.
Collapse
|
45
|
Zhang J, Hu K, Di L, Wang P, Liu Z, Zhang J, Yue P, Song W, Zhang J, Chen T, Wang Z, Zhang Y, Wang X, Zhan C, Cheng YC, Li X, Li Q, Fan JY, Shen Y, Han JY, Qiao H. Traditional herbal medicine and nanomedicine: Converging disciplines to improve therapeutic efficacy and human health. Adv Drug Deliv Rev 2021; 178:113964. [PMID: 34499982 DOI: 10.1016/j.addr.2021.113964] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 02/08/2023]
Abstract
Traditional herbal medicine (THM), an ancient science, is a gift from nature. For thousands of years, it has helped humans fight diseases and protect life, health, and reproduction. Nanomedicine, a newer discipline has evolved from exploitation of the unique nanoscale morphology and is widely used in diagnosis, imaging, drug delivery, and other biomedical fields. Although THM and nanomedicine differ greatly in time span and discipline dimensions, they are closely related and are even evolving toward integration and convergence. This review begins with the history and latest research progress of THM and nanomedicine, expounding their respective developmental trajectory. It then discusses the overlapping connectivity and relevance of the two fields, including nanoaggregates generated in herbal medicine decoctions, the application of nanotechnology in the delivery and treatment of natural active ingredients, and the influence of physiological regulatory capability of THM on the in vivo fate of nanoparticles. Finally, future development trends, challenges, and research directions are discussed.
Collapse
|
46
|
Jiang Y, Meng W, Wu L, Shao K, Wang L, Ding M, Shi J, Kong X. Image-Guided TME-Improving Nano-Platform for Ca 2+ Signal Disturbance and Enhanced Tumor PDT. Adv Healthc Mater 2021; 10:e2100789. [PMID: 34165254 DOI: 10.1002/adhm.202100789] [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: 04/23/2021] [Revised: 05/26/2021] [Indexed: 01/13/2023]
Abstract
Dysfunction of the calcium balancing system and disruption of calcium distribution can induce abnormal intracellular calcium overload, further causing serious damage and even cell death, which provides a potential therapeutic approach for tumor treatment. Herein, a nano-platform, which includes UCNPs-Ce6@RuR@mSiO2 @PL-HA NPs (UCRSPH) and SA-CaO2 nanoparticles, is prepared for improving the tumor micro-environment (TME), Ca2+ signal disturbance as well as enhanced photodynamic tumor therapy (PDT). UCRSPH combined with SA-CaO2 can alter TME and relieve hypoxia of the tumor to realize self-reinforcing PDT under near-IR irradiation (980 nm). The ruthenium red (RuR) in the UCRSPH NPs can be released to the cytoplasm after endocytosis of the nanoparticles, target Ca2+ channel proteins on the endoplasmic reticulum and mitochondria, sarcoplasmic reticulum Ca2+ -ATPase (SERCA), and mitochondrial calcium uniporter (MCU). The combined participation of nanoparticles and RuR promotes Ca2+ imbalance and cytoplasmic calcium overload with the assistance of CaO2 , and provides tumor cells higher sensitivity to PDT. Furthermore, the nano-platform also provides fluorescence imaging and calcification computed tomography imaging for in vivo treatment guidance. In conclusion, this image-guided nano-platform show potential for highly specific, efficient combined therapy against tumor cells with minimal side-effects to normal cells by integrating TME improvement, self-reinforcing PDT, and Ca2+ signal disturbance.
Collapse
Affiliation(s)
- Yuping Jiang
- College of Chemistry and Pharmaceutical Sciences Qingdao Agricultural University 700 Changcheng Road Qingdao 266109 China
| | - Wei Meng
- Second Internal Medicine Department Zaozhuang Yicheng People's Hospital 121 Chengshui Road Zaozhuang 277300 China
| | - Lijuan Wu
- College of Medicine and Pharmacy Ocean University of China 5 Yushan Road Qingdao 266071 China
| | - Kai Shao
- Department of Central Laboratory Qilu Hospital (Qingdao) Cheeloo College of Medicine Shandong University 758 Hefei Road Qingdao 266035 China
| | - Lili Wang
- College of Science and Information Qingdao Agricultural University 700 Changcheng Road Qingdao 266109 China
| | - Mengchao Ding
- College of Chemistry and Pharmaceutical Sciences Qingdao Agricultural University 700 Changcheng Road Qingdao 266109 China
| | - Jinsheng Shi
- College of Chemistry and Pharmaceutical Sciences Qingdao Agricultural University 700 Changcheng Road Qingdao 266109 China
| | - Xiaoying Kong
- College of Chemistry and Pharmaceutical Sciences Qingdao Agricultural University 700 Changcheng Road Qingdao 266109 China
| |
Collapse
|
47
|
Chaudhry GES, Akim A, Naveed Zafar M, Safdar N, Sung YY, Muhammad TST. Understanding Hyaluronan Receptor (CD44) Interaction, HA-CD44 Activated Potential Targets in Cancer Therapeutics. Adv Pharm Bull 2021; 11:426-438. [PMID: 34513617 PMCID: PMC8421618 DOI: 10.34172/apb.2021.050] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 07/14/2020] [Accepted: 07/15/2020] [Indexed: 12/11/2022] Open
Abstract
Cancer is a complex mechanism involving a series of cellular events. The glycoproteins such as hyaluronan (HA) are a significant element of extracellular matrix (ECM), involve in the onset of cancer developmental process. The pivotal roles of HA in cancer progression depend on dysregulated expression in various cancer. HA, also gain attention due to consideration as a primary ligand of CD44 receptor. The CD44, complex transmembrane receptor protein, due to alternative splicing in the transcription process, various CD44 isoforms predominantly exist. The overexpression of distinct CD44 isoforms (CD44v) standard (CD44s) depends on the tumour type and stage. The receptor proteins, CD44 engage in a variety of biological processes, including cell growth, apoptosis, migration, and angiogenesis. HA-CD44 interaction trigger survival pathways that result in cell proliferation, invasion ultimately complex metastasis. The interaction and binding of ligand-receptor HA-CD44 regulate the downstream cytoskeleton pathways involve in cell survival or cell death. Thus, targeting HA, CD44 (variant and standard) isoform, and HA-CD44 binding consider as an attractive and useful approach towards cancer therapeutics. The use of various inhibitors of HA, hyaluronidases (HYALs), and utilizing targeted Nano-delivery of anticancer agents and antibodies against CD44, peptides gives promising results in vitro and in vivo. However, they are in clinical trials with favourable and unfavourable outcomes, which reflects the need for various modifications in targeting agents and a better understanding of potential targets in tumour progression pathways.
Collapse
Affiliation(s)
- Gul-E-Saba Chaudhry
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Terengganu, Malaysia
| | - Abdah Akim
- Department of Biomedical Sciences, Universiti Putra Malaysia, Seri Kembangan, Selangor, Malaysia
| | | | - Naila Safdar
- Department of Environmental Sciences, Fatima Jinnah University, Rawalpindi, Pakistan
| | - Yeong Yik Sung
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, 21030 Kuala Terengganu, Malaysia
| | | |
Collapse
|
48
|
Zuo S, Wang Z, An X, Wang J, Zheng X, Shao D, Zhang Y. Self-Assembly Engineering Nanodrugs Composed of Paclitaxel and Curcumin for the Combined Treatment of Triple Negative Breast Cancer. Front Bioeng Biotechnol 2021; 9:747637. [PMID: 34504835 PMCID: PMC8421550 DOI: 10.3389/fbioe.2021.747637] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Accepted: 08/11/2021] [Indexed: 12/16/2022] Open
Abstract
The clinical outcomes of triple-negative breast cancer (TNBC) chemotherapy are unsatisfactory. Water solubility and biosafety of chemo drugs are also major barriers for achieving satisfactory treatment effect. In this study, we have reported a combinational strategy by self-assembly engineering nanodrugs PC NDs, which were composed of paclitaxel (PTX) and curcumin (Cur), for effective and safe TNBC chemotherapy. PC NDs were prepared through reprecipitation method without using any additional carriers. The PC NDs were preferentially taken up by TNBC cells and we also observed pH-related drug release. Compared with free PTX and simple PTX/Cur mixture, PC NDs have shown higher therapeutic efficiency and better prognosis while the metastasis rate was significantly lower than that of either PTX or PTX/Cur mix group. Therefore, the self-assembly engineered PC NDs might be a promising nanodrugs for efficient and safe TNBC chemotherapy.
Collapse
Affiliation(s)
- Shuting Zuo
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Zhenyu Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xianquan An
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, China
| | - Jing Wang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Xiao Zheng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, China
| | - Yan Zhang
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
49
|
Zhao ZQ, Song W, Yan XQ, Tang JH, Hou JC, Wang DD, Yang SJ, Zhang Q, Zhang J. Autophagy Modulation and Synergistic Therapy to Combat Multidrug Resistance Breast Cancer Using Hybrid Cell Membrane Nanoparticles. J Biomed Nanotechnol 2021; 17:1404-1416. [PMID: 34446143 DOI: 10.1166/jbn.2021.3116] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The development of multidrug resistance (MDR) is a commonly observed phenomenon in many cancer types. It contributed significantly to the poor outcome of many currently available chemotherapies. Considering autophagy as one of the most important physiological process in cancer progression, we thereby proposed an anti-autophagy siRNA and doxorubicin (Dox) co-delivery system (MC/D-siR) to combat MDR breast cancer using sequential construction. Our results demonstrated the potential of MC/D-siR to effectively transfect the loaded siRNA to result in significant downregulation of intracellular autophagy level in MCF-7/Adr (Dox resistance MCF-7 cell line) cells, which in turn cut off the ATP supply and to reverse the MDR and potentiated accumulated drug retention in cells. As a result, MC/D-siR showed much elevated anticancer benefits than single loaded platforms (MC/Dox or MC/siRNA), indicating the ability for effective MDR cancer treatment through the combination of autophagy regulation and chemotherapy.
Collapse
Affiliation(s)
- Zhi-Qiang Zhao
- The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an, 223002, P. R. China
| | - Wei Song
- Department of General Surgery, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu 223300, China
| | - Xue-Qin Yan
- Department of General Surgery, Huai'an People's Hospital of Hongze District, Huai'an, 223002, P. R. China
| | - Jin-Hai Tang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Jun-Chen Hou
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Dan-Dan Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Su-Jin Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Jian Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| |
Collapse
|
50
|
Nanoplatform-based natural products co-delivery system to surmount cancer multidrug-resistant. J Control Release 2021; 336:396-409. [PMID: 34175367 DOI: 10.1016/j.jconrel.2021.06.034] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 12/15/2022]
Abstract
The emergence of multidrug resistance (MDR) in malignant tumors is the primary reason for invalid chemotherapy. Antitumor drugs are often adversely affected by the MDR of tumor cells. Treatments using conventional drugs, which have specific drug targets, hardly regulate the complex signaling pathway of MDR cells because of the complex formation mechanism of MDR. However, natural products have positive advantages, such as high efficiency, low toxicity, and ability to target multiple mechanism pathways associated with MDR. Natural products, as MDR reversal agents, synergize with chemotherapeutics and enhance the sensitivity of tumor cells to chemotherapeutics, and the co-delivery of natural products and antitumor drugs with nanocarriers maximizes the synergistic effects against MDR in tumor cells. This review summarizes the molecular mechanisms of MDR, the advantages of natural products combined with chemotherapeutics in offsetting complicated MDR mechanisms, and the types and mechanisms of natural products that are potential MDR reversal modulators. Meanwhile, aiming at the low bioavailability of cocktail combined natural products and chemotherapeutic in vivo, the advantages of nanoplatform-based co-delivery system and recent research developments are illustrated on the basis of our previous research. Finally, prospective horizons are analyzed, which are expected to considerably improve the nano-co-delivery of natural products and chemotherapeutic systems for MDR reversal in cancer.
Collapse
|