1
|
Shah KA, Razzaq A, You B, Dormocara A, Iqbal H, Cui JH. Unveiling the potential of pulmonary surfactant-based nanocarriers for protein inhalation therapy. Eur J Pharm Biopharm 2024; 205:114574. [PMID: 39521354 DOI: 10.1016/j.ejpb.2024.114574] [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: 07/07/2024] [Revised: 08/31/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
The study investigates the effect of pulmonary surfactant (PS) coating on the performance of lysozyme-loaded poly(lactic-co-glycolic) acid (PLGA) nanoparticles (NPs). The NPs were fabricated using a double emulsification technique and optimized using the Box-Behnken experimental design (BBED). The NPs were assessed for size, polydispersity index (PDI), zeta potential, drug loading (DL%), and encapsulation efficiency (EE%). In addition, the optimized PLGA NPs were modified with either a neutral dipalmitoylphosphatidylcholine DPPC or an anionic dipalmitoyl phosphatidylglycerol (DPPG) with different molar ratios of PS to PLGA (PS: PLGA = 1:2, 1:1 and 2:1). These NPs were assessed for biological activity, drug release, mucus adhesion, mucus penetration, cellular uptake, toxicity, and in vivo destiny after intratracheal (IT) instillation to mice. Results showed a bi-phasic drug release, with no significant effect of PS on the release and biological activities of PLGA NPs. The PS@PLGA NPs improved mucus adhesion, decreased mucus penetration, and increased cellular internalization of PLGA NPs. In addition, ex vivo experiments demonstrated that DPPC@PLGA NPs and DPPG@PLGA NPs could adhere to mucus. These NPs created a thicker layer at the interface of the airway compared to unmodified PLGA NPs. Moreover, interaction of PS@PLGA NPs with BALF suggested improved mucoadhesive characteristics. Finally, the in vivo studies confirmed the precise distribution of all NPs in the lungs after IT administration. The study presents empirical evidence and scientific guidance for developing a lung surfactant-modified nanocarrier system for lung drug delivery.
Collapse
Affiliation(s)
- Kiramat Ali Shah
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Anam Razzaq
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Bengang You
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Amos Dormocara
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Haroon Iqbal
- Eye Hospital, School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou 325027, Zhejiang, China.
| | - Jing-Hao Cui
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
2
|
Yang S, Zhang Y, Zheng C. β-Sitosterol Mitigates Apoptosis, Oxidative Stress and Inflammatory Response by Inactivating TLR4/NF-кB Pathway in Cell Models of Diabetic Nephropathy. Cell Biochem Biophys 2024:10.1007/s12013-024-01559-4. [PMID: 39424766 DOI: 10.1007/s12013-024-01559-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Podocyte injury plays a pivotal role in the pathogenesis of diabetic nephropathy (DN), leading to proteinuria formation. β-Sitosterol is a natural compound with anti-inflammatory, anti-diabetic, nephroprotective and antioxidant properties. The studyaimed to explore whether and how β-Sitosterol protected podocytes against high glucose (HG)-induced inflammatory andoxidative injury. DN cell models were established by stimulating podocytes or renal tubular epithelial cells (HK-2) cells with 25 mM glucose. Cell viability and apoptosis were evaluated using cell counting kit-8 assays and flow cytometry analyses. Westernblotting was used to quantify protein levels of genes related to podocyte injury, HK-2 cell damage, inflammation, and TLR4/NF-кB pathway. Contents of oxidative stress biomarkers were evaluated by corresponding commercial kits while proinflammatorycytokine levels were determined by enzyme-linked immunosorbent assay. Immunofluorescence staining was performed todetect intracellular levels of reactive oxygen species (ROS) and Nrf2 nuclear translocation. Experimental results revealed that HG treatment induced podocyte dysfunction by impairing cell viability while accelerating theapoptosis, and the changes were reversed by β-sitosterol treatment. Moreover, β-sitosterol repressed HG-evoked oxidative stressby reducing ROS and malondialdehyde (MDA) levels while increasing activities of antioxidant enzymes. The reduction ofproinflammatory cytokines mediated by β-sitosterol in HG-stimulated podocytes suggested the anti-inflammatory role of β-sitosterol. Additionally, the activation of the TLR4/NF-кB signaling induced by HG was inhibited by β-sitosterol in podocytes.Inactivation of the TLR4 using TAK-242 enhanced the protective effects of β-sitosterol against HG-mediated oxidative stressand inflammation. Similarly, β-sitosterol also protected HK-2 cells from HG-induced oxidative stress, inflammation, andapoptosis. In summary, β-sitosterol exerts anti-inflammatory, anti-oxidative, and anti-apoptotic activities in HG-induced podocytes or HK-2 cells by inhibiting TLR4/NF-кB signaling.
Collapse
Affiliation(s)
| | | | - Chenghong Zheng
- Hubei University of Chinese Medicine, Wuhan Hospital of Traditional Chinese Medicine, Wuhan, China.
| |
Collapse
|
3
|
Costabile G, Baldassi D, Müller C, Groß B, Ungaro F, Schubert S, Firestine SM, Merkel OM. Antibiotic-loaded nanoparticles for the treatment of intracellular methicillin-resistant Staphylococcus Aureus infections: In vitro and in vivo efficacy of a novel antibiotic. J Control Release 2024; 374:454-465. [PMID: 39181163 DOI: 10.1016/j.jconrel.2024.08.029] [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: 03/19/2024] [Revised: 07/29/2024] [Accepted: 08/19/2024] [Indexed: 08/27/2024]
Abstract
Antimicrobial resistance is considered one of the biggest threats to public health worldwide. Methicillin-resistant S. aureus is the causative agent of a number of infections and lung colonization in people suffering from cystic fibrosis. Moreover, a growing body of evidence links the microbiome to the development of cancer, as well as to the success of the treatment. In this view, the development of novel antibiotics is of critical importance, and SV7, a novel antibiotic active against MRSA at low concentrations, represents a promising candidate. However, the low aqueous solubility of SV7 hampers its therapeutic translation. In this study, SV7 was encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) to improve the solubility profile, to ensure sustained release and eventually support deposition in the airways. Furthermore, PLGA NPs were formulated as dry powder to extend their shelf-life and were shown to efficiently target intracellular infections. After identifying a formulation with suitable physico-chemical characteristics, SV7-loaded NPs were investigated in vitro in terms of inhibitory activity against MRSA, and their safety profile in lung epithelial cells. Subsequently, the activity against MRSA intracellular infections was investigated in a co-culture model of MRSA and macrophages. To test the translatability of our findings, SV7-loaded NPs were tested in vivo in a Galleria mellonella infection model. In conclusion, SV7-loaded NPs showed a safe profile and efficient inhibitory activity against MRSA at low concentrations. Furthermore, their activity against intracellular infections was confirmed, and was retained in vivo, rendering them a promising candidate for treatment of MRSA lung infections.
Collapse
Affiliation(s)
- Gabriella Costabile
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany; Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, IT, Italy
| | - Domizia Baldassi
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany
| | - Christoph Müller
- Department of Pharmacy, Center for Drug Research, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany
| | - Birgit Groß
- Max von Pettenkofer-Institut Munich für Hygiene und Medizinische Mikrobiologie, Elisabeth-Winterhalter-Weg 6, 81377 Munich, DE, Germany
| | - Francesca Ungaro
- Department of Pharmacy, University of Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, IT, Italy
| | - Sören Schubert
- Max von Pettenkofer-Institut Munich für Hygiene und Medizinische Mikrobiologie, Elisabeth-Winterhalter-Weg 6, 81377 Munich, DE, Germany
| | - Steven M Firestine
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Eugene Applebaum College of Pharmacy and Health Sciences, 259 Mack Ave, Detroit, MI 48201, USA
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology & Biopharmaceutics, Ludwig-Maximilians-Universität München, Butenandtstr. 5-13, 81377 Munich, DE, Germany.
| |
Collapse
|
4
|
Prasad A, Bakr MM, ElMeshad AN. Surface-functionalised polymeric nanoparticles for breast cancer treatment: processes and advances. J Drug Target 2024; 32:770-784. [PMID: 38717907 DOI: 10.1080/1061186x.2024.2353359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/09/2024] [Accepted: 05/03/2024] [Indexed: 05/18/2024]
Abstract
The World Health Organization (WHO) reported that of all the non-communicable diseases, cancer is considered the second cause of death worldwide. This has driven the big pharma companies to prioritise anticancer products in their pipeline. In addition, research has focused on exploration of new anticancer molecules and design of suitable dosage forms to achieve effective drug delivery to the tumour site. Nanotechnology is a valuable tool to build nano delivery systems with controlled and targeted drug release properties. Nanoparticles can be fabricated by robust, scalable and economic techniques using various polymers. Moreover, specific functional groups can be introduced to the surface of nanoparticles enabling targeting to a specific tissue; besides, they exhibit versatile drug release patterns according to the rate of polymer degradation. This review outlines the processes and advances in surface functionalisation of nanoparticles employed for treatment of breast cancer. The therapeutic molecules, the polymers used to fabricate nanoparticles, the techniques used to prepare the nanoparticles have been reviewed with a focus on the processes employed to functionalise these nanoparticles with suitable ligands to target different types of breast cancer.
Collapse
Affiliation(s)
- Aprameya Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Mohamed Mofreh Bakr
- Department of Pharmaceutics, Egyptian Drug Authority, Formerly Known as National Organization for Drug Control and Research, Giza, Egypt
| | - Aliaa N ElMeshad
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Department of Pharmaceutics, Faculty of Pharmacy and Drug Technology, The Egyptian Chinese University, Cairo, Egypt
| |
Collapse
|
5
|
Yang L, Wang L, Bao E, Wang J, Zhu P. Causal association of dietary factors with five common cancers: univariate and multivariate Mendelian randomization studies. Front Nutr 2024; 11:1428844. [PMID: 39135550 PMCID: PMC11317396 DOI: 10.3389/fnut.2024.1428844] [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: 05/07/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Background Daily dietary habits are closely related to human health, and long-term unhealthy dietary intake, such as excessive consumption of alcohol and pickled foods, may promote the development of cancers. However, comprehensive research on the causal relationship between dietary habits and cancer is lacking. Therefore, this study aimed to reveal the potential causal link between dietary risk factors and the prognosis of cancer-related to genetic susceptibility. Methods GWAS (Genome-Wide Association Studies) summary data on dietary habits and five common types of cancer and their pathological subtypes were obtained from the UK Biobank and various cancer association consortia. A univariable two-sample Mendelian randomization (UVMR) and FDR correction analysis was conducted to explore the causal relationships between 45 dietary habits and five common types of cancer and their histopathological subtypes. In addition, multivariable Mendelian randomization analysis (MVMR) was performed to adjust for traditional risk factors for dietary habits, and the direct or indirect effects of diet on cancer were evaluated. Finally, the prognostic impact of selected instrumental variables on cancer was analyzed using an online data platform. Results In the UVMR analysis, four dietary habits were identified as risk factors for cancer, while five dietary habits were identified as protective factors. Among the latter, one dietary habit showed a significant association with cancer even after FDR correction, indicating a potential causal relationship. The MVMR analysis revealed that weekly beer and cider intake, may act as an independent risk factor for cancer development. Other causal associations between dietary habits and cancer risk may be mediated by intermediate factors. In the prognostic analysis, the SNPs (Single Nucleotide Polymorphisms) of average weekly beer and cider intake were set as independent risk factors and were found to significantly impact overall survival (OS) and cancer-specific survival (CSS) in lung cancer. Conclusion This causal relationship study supports the notion that adjusting daily dietary habits and specific dietary interventions may decrease the risk of cancer.
Collapse
Affiliation(s)
- Lin Yang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Li Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
| | - Erhao Bao
- Department of Urology, The First People's Hospital of Dazhou, Dazhou, Sichuan, China
| | - Jiahao Wang
- Department of Urology, People's Hospital of Xichong County, Nanchong, Sichuan, China
| | - Pingyu Zhu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
6
|
Kanchan S, Marwaha D, Tomar B, Agrawal S, Mishra S, Kapoor R, Sushma, Jha G, Sharma D, Bhatta RS, Mishra PR, Rath SK. Nanocarrier - Mediated Salinomycin Delivery Induces Apoptosis and Alters EMT Phenomenon in Prostate Adenocarcinoma. AAPS PharmSciTech 2024; 25:104. [PMID: 38724836 DOI: 10.1208/s12249-024-02817-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/22/2024] [Indexed: 09/05/2024] Open
Abstract
Salinomycin (Sal) has been recently discovered as a novel chemotherapeutic agent against various cancers including prostate cancer which is one of the most commonly diagnosed cancers affecting male populations worldwide. Herein we designed salinomycin nanocarrier (Sal-NPs) to extend its systemic circulation and to increase its anticancer potential. Prepared nanoform showed high encapsulation and sustained release profile for salinomycin. The present study elucidated the cytotoxicity and mechanism of apoptotic cell death of Sal-NPs against prostate cancer both in vitro and in vivo. At all measured concentrations, Sal-NPs showed more significant cytotoxicity to DU145 and PC3 cells than Sal alone. This effect was mediated by apoptosis, as confirmed by ROS generation, loss of MMP and cell cycle arrest at the G1 phase in both cells. Sal-NPs efficiently inhibited migration of PC3 and DU145 cells via effectively downregulating the epithelial mesenchymal transition. Also, the results confirmed that Sal-NPs can effectively inhibit the induction of Prostate adenocarcinoma in male Wistar rats. Sal-NPs treatment exhibited a decrease in tumour sizes, a reduction in prostate weight, and an increase in body weight, which suggests that Sal-NPs is more effective than salinomycin alone. Our results suggest that the molecular mechanism underlying the Sal-NPs anticancer effect may lead to the development of a potential therapeutic strategy for treating prostate adenocarcinoma.
Collapse
Affiliation(s)
- Sonam Kanchan
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Bhawna Tomar
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sristi Agrawal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sakshi Mishra
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Radhika Kapoor
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Sushma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Gaurav Jha
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Divyansh Sharma
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India
| | - Rabi Sankar Bhatta
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Srikanta Kumar Rath
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, B.S. 10/1, Sector 10, Jankipuram Extension, Sitapur Road, Lucknow, 226031, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
7
|
Wang J, Ni BY, Wang J, Han L, Ni X, Wang XM, Cao LC, Sun QH, Han XP, Cui HJ. Research progress of Paris polyphylla in the treatment of digestive tract cancers. Discov Oncol 2024; 15:31. [PMID: 38324023 PMCID: PMC10850040 DOI: 10.1007/s12672-024-00882-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/01/2024] [Indexed: 02/08/2024] Open
Abstract
Cancer has become one of the most important causes of human death. In particular, the 5 year survival rate of patients with digestive tract cancer is low. Although chemotherapy drugs have a certain efficacy, they are highly toxic and prone to chemotherapy resistance. With the advancement of antitumor research, many natural drugs have gradually entered basic clinical research. They have low toxicity, few adverse reactions, and play an important synergistic role in the combined targeted therapy of radiotherapy and chemotherapy. A large number of studies have shown that the active components of Paris polyphylla (PPA), a common natural medicinal plant, can play an antitumor role in a variety of digestive tract cancers. In this paper, the main components of PPA such as polyphyllin, C21 steroids, sterols, and flavonoids, amongst others, are introduced, and the mechanisms of action and research progress of PPA and its active components in the treatment of various digestive tract cancers are reviewed and summarized. The main components of PPA have been thoroughly explored to provide more detailed references and innovative ideas for the further development and utilization of similar natural antitumor drugs.
Collapse
Affiliation(s)
- Jia Wang
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Bao-Yi Ni
- Heilongjiang University of Chinese Medicine, Harbin, China
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jing Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate College, Beijing University of Chinese Medicine, Chaoyang, China
| | - Lei Han
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Xin Ni
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Xin-Miao Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Lu-Chang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian-Hui Sun
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xin-Pu Han
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hu-Jun Cui
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China.
| |
Collapse
|
8
|
Nandi S, Nag A, Khatua S, Sen S, Chakraborty N, Naskar A, Acharya K, Calina D, Sharifi-Rad J. Anticancer activity and other biomedical properties of β-sitosterol: Bridging phytochemistry and current pharmacological evidence for future translational approaches. Phytother Res 2024; 38:592-619. [PMID: 37929761 DOI: 10.1002/ptr.8061] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/13/2023] [Accepted: 10/15/2023] [Indexed: 11/07/2023]
Abstract
Sterols, including β-sitosterol, are essential components of cellular membranes in both plant and animal cells. Despite being a major phytosterol in various plant materials, comprehensive scientific knowledge regarding the properties of β-sitosterol and its potential applications is essential for scholarly pursuits and utilization purposes. β-sitosterol shares similar chemical characteristics with cholesterol and exhibits several pharmacological activities without major toxicity. This study aims to bridge the gap between phytochemistry and current pharmacological evidence of β-sitosterol, focusing on its anticancer activity and other biomedical properties. The goal is to provide a comprehensive understanding of β-sitosterol's potential for future translational approaches. A thorough examination of the literature was conducted to gather relevant information on the biological properties of β-sitosterol, particularly its anticancer therapeutic potential. Various databases were searched, including PubMed/MedLine, Scopus, Google Scholar, and Web of Science using appropriate keywords. Studies investigating the effects of β-sitosterol on different types of cancer were analyzed, focusing on mechanisms of action, pharmacological screening, and chemosensitizing properties. Modern pharmacological screening studies have revealed the potential anticancer therapeutic properties of β-sitosterol against various types of cancer, including leukemia, lung, stomach, breast, colon, ovarian, and prostate cancer. β-sitosterol has demonstrated chemosensitizing effects on cancer cells, interfering with multiple cell signaling pathways involved in proliferation, cell cycle arrest, apoptosis, survival, metastasis invasion, angiogenesis, and inflammation. Structural derivatives of β-sitosterol have also shown anti-cancer effects. However, research in the field of drug delivery and the detailed mode of action of β-sitosterol-mediated anticancer activities remains limited. β-sitosterol, as a non-toxic compound with significant pharmacological potential, exhibits promising anticancer effects against various cancer types. Despite being relatively less potent than conventional cancer chemotherapeutics, β-sitosterol holds potential as a safe and effective nutraceutical against cancer. Further comprehensive studies are recommended to explore the biological properties of β-sitosterol, including its mode of action, and develop novel formulations for its potential use in cancer treatment. This review provides a foundation for future investigations and highlights the need for further research on β-sitosterol as a potent superfood in combating cancer.
Collapse
Affiliation(s)
- Sudeshna Nandi
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Anish Nag
- Department of Life Sciences, CHRIST (Deemed to be University), Bangalore, India
| | - Somanjana Khatua
- Department of Botany, Faculty of Science, University of Allahabad, Prayagraj, India
| | - Surjit Sen
- Department of Botany, Fakir Chand College, Kolkata, India
| | | | - Arghya Naskar
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Krishnendu Acharya
- Molecular and Applied Mycology and Plant Pathology Laboratory, Department of Botany, University of Calcutta, Kolkata, India
| | - Daniela Calina
- Department of Clinical Pharmacy, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | |
Collapse
|
9
|
Kumar M, Gupta S, Kalia K, Kumar D. Role of Phytoconstituents in Cancer Treatment: A Review. RECENT ADVANCES IN FOOD, NUTRITION & AGRICULTURE 2024; 15:115-137. [PMID: 38369892 DOI: 10.2174/012772574x274566231220051254] [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: 09/04/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 02/20/2024]
Abstract
Over the years, natural compounds have become a significant advancement in cancer treatment, primarily due to their effectiveness, safety, bio-functionality, and wide range of molecular structures. They are now increasingly preferred in drug discovery due to these attributes. These compounds, whether occurring naturally or with synthetic modifications, find applications in various fields like biology, medicine, and engineering. While chemotherapy has been a successful method for treating cancer, it comes with systemic toxicity. To address this issue, researchers and medical practitioners are exploring the concept of combinational chemotherapy. This approach aims to reduce toxicity by using a mix of natural substances and their derivatives in clinical trials and prescription medications. Among the most extensively studied natural anticancer compounds are quercetin, curcumin, vincristine, and vinblastine. These compounds play crucial roles as immunotherapeutics and chemosensitizers, both as standalone treatments and in combination therapies with specific mechanisms. This review article provides a concise overview of the functions, potentials, and combinations of natural anticancer compounds in cancer treatment, along with their mechanisms of action and clinical applications.
Collapse
Affiliation(s)
- Manish Kumar
- Department of Pharmacy, IEC College of Eng & Tech. Gautam Buddha Nagar, India
| | | | | | - Dharmendra Kumar
- Department of Pharmacy, IEC College of Eng & Tech. Gautam Buddha Nagar, India
| |
Collapse
|
10
|
Wang H, Wang Z, Zhang Z, Liu J, Hong L. β-Sitosterol as a Promising Anticancer Agent for Chemoprevention and Chemotherapy: Mechanisms of Action and Future Prospects. Adv Nutr 2023; 14:1085-1110. [PMID: 37247842 PMCID: PMC10509430 DOI: 10.1016/j.advnut.2023.05.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/15/2023] [Accepted: 05/23/2023] [Indexed: 05/31/2023] Open
Abstract
Cancer is one of the primary causes of death worldwide, and its incidence continues to increase yearly. Despite significant advances in research, the search for effective and nontoxic preventive and therapeutic agents remains greatly important. Cancer is a multimodal disease, where various mechanisms play significant roles in its occurrence and progression. This highlights the need for multitargeted approaches that are not only safe and inexpensive but also provide effective alternatives for current therapeutic regimens. β-Sitosterol (SIT), the most abundant phytosterol found in various plant foods, represents such an option. Preclinical evidence over the past few decades has overwhelmingly shown that SIT exhibits multiple anticancer activities against varied cancers, such as liver, cervical, colon, stomach, breast, lung, pancreatic, and prostate cancers, in addition to leukemia, multiple myeloma, melanoma, and fibrosarcoma. In this article, we present the latest advances and perspectives on SIT-systematically summarizing its antitumor mechanisms of action into 7 main sections and combining current challenges and prospects-for its use as a promising agent for cancer prevention and treatment. In particular, SIT plays a role in cancer prevention and treatment mainly by enhancing apoptosis, inducing cell cycle arrest, bidirectionally regulating oxidative stress, improving metabolic reprogramming, inhibiting invasion and metastasis, modulating immunity and inflammation, and combating drug resistance. Although SIT holds such great promise, the poor aqueous solubility and bioavailability coupled with low targeting efficacy limit its therapeutic efficacy and clinical application. Further research on novel drug delivery systems may improve these deficiencies. Overall, through complex and pleiotropic mechanisms, SIT has good potential for tumor chemoprevention and chemotherapy. However, no clinical trials have yet proven this potential. This review provides theoretical basis and rationality for the further design and conduct of clinical trials to confirm the anticancer activity of SIT.
Collapse
Affiliation(s)
- Haoyu Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhi Wang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zihui Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingchun Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Li Hong
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
11
|
Fitzgerald DM, Zhang H, Bordeianu C, Colson YL, Grinstaff MW. Synthesis of Polyethylene Glycol-Poly(glycerol carbonate) Block Copolymeric Micelles as Surfactant-Free Drug Delivery Systems. ACS Macro Lett 2023; 12:974-979. [PMID: 37390500 PMCID: PMC11331582 DOI: 10.1021/acsmacrolett.3c00275] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/02/2023]
Abstract
We report the synthesis of block copolymers of monomethoxylated polyethylene glycol and poly(glycerol carbonate) (mPEG-b-PGC) via the ring-opening polymerization of benzyl glycidyl ether, monomethoxylated polyethylene glycol, and carbon dioxide using a cobalt salen catalyst. The resulting block copolymers display high polymer/cyclic carbonate selectivity (>99%) and, if two oxirane monomers are used, random incorporation into the polymer feed. The resulting diblock mPEG-b-PGC polymer shows promise as a nanocarrier for surfactant-free, sustained chemotherapeutic delivery. mPEG-b-PGC, with paclitaxel conjugated to the pendant primary alcohol of the glycerol polymer backbone, readily forms 175 nm diameter particles in solution and contains 4.6 wt % paclitaxel (PTX), which is released over 42 days. The mPEG-b-PGC polymer itself is noncytotoxic, whereas the PTX-loaded nanoparticles are cytotoxic to lung, breast, and ovarian cancer cell lines.
Collapse
Affiliation(s)
- Danielle M. Fitzgerald
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| | - Heng Zhang
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| | - Catalina Bordeianu
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| | - Yolonda L. Colson
- Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, Boston, MA 02214
| | - Mark W. Grinstaff
- Departments of Chemistry and Biomedical Engineering Boston University, 590 Commonwealth Ave, Boston, MA, 02115
| |
Collapse
|
12
|
Koniuch N, Ilett M, Collins SM, Hondow N, Brown A, Hughes L, Blade H. Structure of polymeric nanoparticles encapsulating a drug - pamoic acid ion pair by scanning transmission electron microscopy. Heliyon 2023; 9:e16959. [PMID: 37360079 PMCID: PMC10285183 DOI: 10.1016/j.heliyon.2023.e16959] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 05/31/2023] [Accepted: 06/02/2023] [Indexed: 06/28/2023] Open
Abstract
Drug-delivery systems based on polymeric nanoparticles are useful for improving drug bioavailability and/or delivery of the active ingredient for example directly to the cancerous tumour. The physical and chemical characterization of a functionalized nanoparticle system is required to measure drug loading and dispersion but also to understand and model the rate and extent of drug release to help predict performance. Many techniques can be used, however, difficulties related to structure determination and identifying the precise location of the drug fraction make mathematical prediction complex and in many published examples the final conclusions are based on assumptions regarding an expected structure. Cryogenic scanning transmission electron microscopy imaging in combination with electron energy loss spectroscopy techniques are used here to address this issue and provide a multi-modal approach to the characterisation of a self-assembled polymeric nanoparticle system based upon a polylactic acid - polyethylene glycol (PLA-PEG) block copolymer containing a hydrophobic ion-pair between pamoic acid and an active pharmaceutical ingredient (API). Results indicate a regular dispersion of spherical nanoparticles of 88 ± 9 nm diameter. The particles are shown to have a multi-layer structure consisting of a 25 nm radius hydrophobic core of PLA and pamoic acid-API material with additional enrichment of the pamoic acid-API material within the inner core (that can be off-centre), surrounded by a 9 nm dense PLA-PEG layer all with a low-density PEG surface coating of around 10 nm thickness. This structure suggests that release of the API can only occur by diffusion through or degradation of the dense, 9 nm thick PLA-PEG layer either of which is a process consistent with the previously reported steady release kinetics of the API and counter ion from these nanoparticle formulations. Establishing accurate measures of product structure enables a link to performance by providing appropriate physical parameters for future mathematical modelling of barriers controlling API release in these nanoparticle formulations.
Collapse
Affiliation(s)
- Natalia Koniuch
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Martha Ilett
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Sean M. Collins
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
- School of Chemistry, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Nicole Hondow
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Andy Brown
- School of Chemical and Process Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Les Hughes
- AstraZeneca, Oral Product Development, Pharmaceutical Technology & Development, Operations, Macclesfield, SK10 2NA, United Kingdom
| | - Helen Blade
- AstraZeneca, Oral Product Development, Pharmaceutical Technology & Development, Operations, Macclesfield, SK10 2NA, United Kingdom
| |
Collapse
|
13
|
Van Gheluwe L, David S, Buchy E, Chourpa I, Munnier E. Smart PEG-Block-PLA/PLA Nanosystems: Impact of the Characteristics of the Polymer Blend on the Redox Responsiveness. MATERIALS (BASEL, SWITZERLAND) 2023; 16:539. [PMID: 36676276 PMCID: PMC9864163 DOI: 10.3390/ma16020539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 06/17/2023]
Abstract
Nanocarriers (NCs) were designed from three polymer blends (B1, B2 and B3) and investigated as smart drug delivery systems (SDDS). The blends are composed of a "smart" copolymer, where methoxy poly(ethylene glycol) and poly(lactic acid) are connected via a redox-responsive disulfide bond (mPEG-SS-PLA), and of a "conventional" polymer, poly(lactic acid) (PLA). They differ by mPEG-SS-PLA/PLA ratio and PLA molecular weight. Nanoprecipitation was used to prepare NCs. Three concentrations were tested, and fluorescent dye Nile red (NR) was used as a model payload. The results show that the characteristics of the NCs, such as size and drug release kinetics, are influenced by the type of blend and the concentration used during the nanoprecipitation process. The more redox-responsive blend was B2 (ratio 1:3, PLA 5 kDa) at 16 mg/mL: the quantity of NR released was tripled upon 24 h of incubation in a reducing medium. This study reveals that the amount of disulfide bonds present in a NC is not the only parameter to be considered to design an SDDS. The stability of the SDDS in a presumably non-stimulating environment is also important to limit uncontrolled release during storage or in the body before the biological target is reached.
Collapse
Affiliation(s)
- Louise Van Gheluwe
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| | - Stephanie David
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| | - Eric Buchy
- Laboratoires Eriger, 33 rue Augustin Fresnel, 37170 Chambray les Tours, France
| | - Igor Chourpa
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| | - Emilie Munnier
- EA 6295 Nanomédicaments et Nanosondes, Université de Tours, Faculté de Pharmacie, 31 Avenue Monge, 37200 Tours, France
| |
Collapse
|
14
|
Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural Products as Anticancer Agents: Current Status and Future Perspectives. Molecules 2022; 27:molecules27238367. [PMID: 36500466 PMCID: PMC9737905 DOI: 10.3390/molecules27238367] [Citation(s) in RCA: 138] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Natural products have been an invaluable and useful source of anticancer agents over the years. Several compounds have been synthesized from natural products by modifying their structures or by using naturally occurring compounds as building blocks in the synthesis of these compounds for various purposes in different fields, such as biology, medicine, and engineering. Multiple modern and costly treatments have been applied to combat cancer and limit its lethality, but the results are not significantly refreshing. Natural products, which are a significant source of new therapeutic drugs, are currently being investigated as potential cytotoxic agents and have shown a positive trend in preclinical research and have prompted numerous innovative strategies in order to combat cancer and expedite the clinical research. Natural products are becoming increasingly important for drug discovery due to their high molecular diversity and novel biofunctionality. Furthermore, natural products can provide superior efficacy and safety due to their unique molecular properties. The objective of the current review is to provide an overview of the emergence of natural products for the treatment and prevention of cancer, such as chemosensitizers, immunotherapeutics, combinatorial therapies with other anticancer drugs, novel formulations of natural products, and the molecular mechanisms underlying their anticancer properties.
Collapse
Affiliation(s)
- Abid Naeem
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Zhang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yali Liu
- Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang Medical College, Nanchang 330006, China
- Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang Medical College, Nanchang 330006, China
| | - Weifeng Zhu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Correspondence:
| |
Collapse
|
15
|
Ni B, Song X, Shi B, Wang J, Sun Q, Wang X, Xu M, Cao L, Zhu G, Li J. Research progress of ginseng in the treatment of gastrointestinal cancers. Front Pharmacol 2022; 13:1036498. [PMID: 36313365 PMCID: PMC9603756 DOI: 10.3389/fphar.2022.1036498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 10/03/2022] [Indexed: 11/24/2022] Open
Abstract
Cancer has become one of the major causes of human death. Several anticancer drugs are available; howeve their use and efficacy are limited by the toxic side effects and drug resistance caused by their continuous application. Many natural products have antitumor effects with low toxicity and fewer adverse effects. Moreover, they play an important role in enhancing the cytotoxicity of chemotherapeutic agents, reducing toxic side effects, and reversing chemoresistance. Consequently, natural drugs are being applied as potential therapeutic options in the field of antitumor treatment. As natural medicinal plants, some components of ginseng have been shown to have excellent efficacy and a good safety profile for cancer treatment. The pharmacological activities and possible mechanisms of action of ginseng have been identified. Its broad range of pharmacological activities includes antitumor, antibacterial, anti-inflammatory, antioxidant, anti-stress, anti-fibrotic, central nervous system modulating, cardioprotective, and immune-enhancing effects. Numerous studies have also shown that throuth multiple pathways, ginseng and its active ingredients exert antitumor effects on gastrointestinal (GI) tract tumors, such as esophageal, gastric, colorectal, liver, and pancreatic cancers. Herein, we introduced the main components of ginseng, including ginsenosides, polysaccharides, and sterols, etc., and reviewed the mechanism of action and research progress of ginseng in the treatment of various GI tumors. Futhermore, the pathways of action of the main components of ginseng are discussed in depth to promote the clinical development and application of ginseng in the field of anti-GI tumors.
Collapse
Affiliation(s)
- Baoyi Ni
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaotong Song
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bolun Shi
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jia Wang
- Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Qianhui Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmiao Wang
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Manman Xu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Luchang Cao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | | | - Jie Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- *Correspondence: Jie Li,
| |
Collapse
|
16
|
Bao X, Zhang Y, Zhang H, Xia L. Molecular Mechanism of β-Sitosterol and its Derivatives in Tumor Progression. Front Oncol 2022; 12:926975. [PMID: 35756648 PMCID: PMC9213880 DOI: 10.3389/fonc.2022.926975] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
β-Sitosterol (SIT), a white powdery organic substance with a molecular formula of C29H50O, is one of the most abundant naturally occurring phytosterols in plants. With a chemical composition similar to that of cholesterol, SIT is applied in various fields such as medicine, agriculture, and chemical industries, owing to its unique biological and physicochemical properties. Modern pharmacological studies have elucidated good anti-tumor therapeutic effect activity of SIT, which mainly manifests as pro-apoptotic, anti-proliferative, anti-metastatic, anti-invasive, and chemosensitizing on tumor cells. In addition, SIT exerts an anti-tumor effect on multiple malignant tumors such as breast, gastric, lung, kidney, pancreatic, prostate, and other cancers. Further, SIT derivatives with structural modifications are promising anti-tumor drugs with significant anti-tumor effects. This review article focuses on recent studies relevant to the anti-tumor effects of SIT and summarizes its anti-tumor mechanism to provide a reference for the clinical treatment of malignant tumors and the development of novel anti-tumor drugs.
Collapse
Affiliation(s)
- Xingxun Bao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan, China
| | - Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
17
|
Li F, Shi Y, Zhang Y, Yang X, Wang Y, Jiang K, Hua C, Wu C, Sun C, Qin Y, Liu S. Investigating the mechanism of Xian-ling-lian-xia-fang for inhibiting vasculogenic mimicry in triple negative breast cancer via blocking VEGF/MMPs pathway. Chin Med 2022; 17:44. [PMID: 35379271 PMCID: PMC8981688 DOI: 10.1186/s13020-022-00597-5] [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: 01/10/2022] [Accepted: 03/20/2022] [Indexed: 11/12/2022] Open
Abstract
Background Xian-ling-lian-xia-fang (XLLXF), a Chinese medicine decoction, is widely used in the treatment of triple negative breast cancer (TNBC). However, the underlying mechanism of XLLXF in TNBC treatment has not been totally elucidated. Methods Here, network pharmacology and molecular docking were used to explore the mechanism of Traditional Chinese medicine in the treatment of TNBC. Then, biological experiments were integrated to verify the results of network pharmacology. Results Network pharmacology showed that the candidate active ingredients mainly included quercetin, kaempferol, stigmasterol, and β-sitosterol through the “XLLXF–active ingredients–targets” network. Vascular endothelial growth factor A (VEGFA) and matrix metalloproteinase (MMP) 2 were the potential therapeutic targets obtained through the protein–protein interaction (PPI) network. Molecular docking confirmed that quercetin, kaempferol, stigmasterol, and β-sitosterol could stably combine with VEGFA and MMP2. Experimental verification showed that XLLXF could inhibit proliferation, colony ability, and vasculogenic mimicry (VM) formation and promote cell apoptosis in TNBC. Laser confocal microscopy found that XLLXF impaired F-actin cytoskeleton organization and inhibited epithelial mesenchymal transition. Animal experiments also found that XLLXF could inhibit tumor growth and VM formation in TNBC xenograft model. Western blot analysis and immunohistochemical staining showed that XLLXF inhibited the protein expression of VEGFA, MMP2, MMP9, Vimentin, VE-cadherin, and Twist1 and increased that of E-cadherin, tissue inhibitors of metalloproteinase (TIMP)-1, and TIMP-3 in vitro and in vivo. Conclusions Integrating the analysis of network pharmacology and experimental validation revealed that XLLXF could inhibit VM formation via downregulating the VEGF/MMPs signaling pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00597-5.
Collapse
Affiliation(s)
- Feifei Li
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Youyang Shi
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Yang Zhang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Xiaojuan Yang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Yi Wang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Kexin Jiang
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Ciyi Hua
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Chunyu Wu
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Chenping Sun
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Yuenong Qin
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| | - Sheng Liu
- Integrated Traditional Chinese and Western Medicine Breast Department, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China.
| |
Collapse
|
18
|
Mingrou L, Guo S, Ho C, Bai N. Review on chemical compositions and biological activities of peanut (
Arachis hypogeae
L.). J Food Biochem 2022; 46:e14119. [DOI: 10.1111/jfbc.14119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/04/2022] [Accepted: 01/29/2022] [Indexed: 12/27/2022]
Affiliation(s)
- Li Mingrou
- College of Food Science and Technology Northwest University Xi’an China
| | - Sen Guo
- College of Food Science and Technology Northwest University Xi’an China
| | - Chi‐Tang Ho
- Department of Food Science Rutgers University New Brunswick New Jersey USA
| | - Naisheng Bai
- College of Food Science and Technology Northwest University Xi’an China
| |
Collapse
|
19
|
Wadhawan A, Singh J, Sharma H, Handa S, Singh G, Kumar R, Barnwal RP, Pal Kaur I, Chatterjee M. Anticancer Biosurfactant-Loaded PLA-PEG Nanoparticles Induce Apoptosis in Human MDA-MB-231 Breast Cancer Cells. ACS OMEGA 2022; 7:5231-5241. [PMID: 35187338 PMCID: PMC8851644 DOI: 10.1021/acsomega.1c06338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/26/2022] [Indexed: 06/14/2023]
Abstract
Despite various advancements in cancer therapies, treating cancer efficiently without side effects is still a major concern for researchers. Anticancer drugs from natural sources need to be explored as a replacement for chemo drugs to overcome their limitations. In our previous studies, isolation, characterization, and anticancer properties of a novel biosurfactant from Candida parapsilosis were reported. In this study, we report the cytotoxicity of the polymeric nanoparticles of this novel biosurfactant toward breast cancer cells. Biosurfactant-encapsulated polymeric nanoparticles of polylactic acid-poly(ethylene glycol) (PLA-PEG) copolymers were synthesized by the double emulsion solvent evaporation method. Folic acid (FA) was used as a targeting ligand to actively deliver the anticancer cargo to the cancer site. The encapsulation efficiency of nanoparticles was observed as 84.9%, and Fickian diffusion was observed as a kinetic model for the release of biosurfactant from nanoparticles. The controlled delivery of the biosurfactant was noticed when encapsulated in PLA-PEG copolymer nanoparticles. Additionally, it was observed that FA enhanced the uptake and cytotoxicity of biosurfactant-loaded nanoparticles in MDA-MB-231 cancer cells compared to biosurfactant-loaded plain nanoparticles. Induction of apoptosis was observed in cancer cells by these nanoparticles. We explore a potential anticancer agent that can be further analyzed for its efficiency and can be used as an alternative tool.
Collapse
Affiliation(s)
- Aishani Wadhawan
- Biotechnology
Branch, University Institute of Engineering and Technology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Joga Singh
- University
Institute of Pharmaceutical Sciences, Panjab
University, Sector 14, Chandigarh 160014, India
| | - Himani Sharma
- Department
of Zoology, Panjab University, Sector 14, Chandigarh 160014, India
| | - Shristi Handa
- Biotechnology
Branch, University Institute of Engineering and Technology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Gurpal Singh
- University
Institute of Pharmaceutical Sciences, Panjab
University, Sector 14, Chandigarh 160014, India
| | - Ravinder Kumar
- Department
of Zoology, Panjab University, Sector 14, Chandigarh 160014, India
| | - Ravi Pratap Barnwal
- Department
of Biophysics, Panjab University, Sector 25, Chandigarh 160014, India
| | - Indu Pal Kaur
- University
Institute of Pharmaceutical Sciences, Panjab
University, Sector 14, Chandigarh 160014, India
| | - Mary Chatterjee
- Biotechnology
Branch, University Institute of Engineering and Technology, Panjab University, Sector 25, Chandigarh 160014, India
| |
Collapse
|
20
|
Albukhaty S, Al-Karagoly H, Allafchian AR, Jalali SAH, Al-Kelabi T, Muhannad M. Production and characterization of biocompatible nanofibrous scaffolds made of β- sitosterolloaded polyvinyl alcohol/tragacanth gum composites. NANOTECHNOLOGY 2021; 33:085102. [PMID: 34749350 DOI: 10.1088/1361-6528/ac3789] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/08/2021] [Indexed: 06/13/2023]
Abstract
Electrospun polyvinyl alcohol (PVA) and tragacanth gum (TG) were used to develop nanofibrous scaffolds containing poorly water-solubleβ-Sitosterol (β-S). Different concentrations and ratios of the polymeric composite includingβ-S (10% w v-1) in PVA (8% w v-1) combined with TG (0.5 and 1% w v-1) were prepared and electrospun. The synthesis method includes four electrospinning parameters of solution concentration, feeding rate, voltage, and distance of the collector to the tip of the needle, which are independently optimized to achieve uniform nanofibers with a desirable mean diameter for cell culture. The collected nanofibers were characterized by SEM, FTIR, and XRD measurements. A contact angle measurement described the hydrophilicity of the scaffold. MTT test was carried out on the obtained nanofibers containing L929 normal fibroblast cells. The mechanical strength, porosity, and deterioration of the scaffolds were well discussed. The mean nanofiber diameters ranged from 63 ± 20 nm to 97 ± 46 nm. The nanofibers loaded withβ-S were freely soluble in water and displayed a remarkable biocompatible nature. The cultured cells illustrated sheet-like stretched growth morphology and penetrated the nanofibrous pores of the PVA/β-S/TG scaffolds. The dissolution was related to submicron-level recrystallization ofβ-S with sufficient conditions for culturing L929 cells. It was concluded that electrospinning is a promising technique for poorly water-solubleβ-S formulations that could be used in biomedical applications.
Collapse
Affiliation(s)
- Salim Albukhaty
- Department of Basic Sciences, University of Misan, Maysan 62001, Iraq
| | - Hassan Al-Karagoly
- Department of Internal and Preventive Medicine, Veterinary Medicine College, University of Al-Qadisiyah, Diwaniyah 58002, Iraq
| | - Ali Reza Allafchian
- Research Institute for Nanotechnology and Advanced Materials, Isfahan University of Technology, Isfahan 84156-83111, Iran
- Research Institute for Biotechnology and Bioengineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Seyed Amir Hossein Jalali
- Research Institute for Biotechnology and Bioengineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
- Department of Natural Resources, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Thair Al-Kelabi
- Directorate of Military Medical Affairs, Ministry of Defense, Baghdad, Iraq
| | - Mustafa Muhannad
- Department of Natural Resources, Isfahan University of Technology, Isfahan 84156-83111, Iran
| |
Collapse
|
21
|
Ahmed MM, Fatima F, Anwer MK, Aldawsari MF, Bhatia S, Al-Harrasi A. Brigatinib loaded poly(d,l-lactide-co-glycolide) nanoparticles for improved anti-tumoral activity against non-small cell lung cancer cell lines. Drug Dev Ind Pharm 2021; 47:1112-1120. [PMID: 34551665 DOI: 10.1080/03639045.2021.1983585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVE The aim of the current investigation was to develop poly(d,l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) to sustain the brigatinib (BTB) release for prolong time period and to examine the antitumor effect of the optimized NPs. SIGNIFICANCE Optimized PLGA-based NPs of BTB could be potentially used as a promising nanocarrier for the treatment of non-small cell lung cancer. METHODS BTB-loaded NPs were fabricated with core-shell of PLGA by solvent evaporation technique using different proportions of PLGA polymer and poly-vinyl alcohol (PVA) stabilizer. The prepared NPs were evaluated for particle characterizations; size, polydispersity index (PDI), zeta-potential, entrapment efficiency (EE), and drug loading (DL), Fourier-transform infrared (FTIR) spectroscopy, differential scanning calorimetry (DSC), and X-ray diffraction studies. The optimized NPs (BN5) were further evaluated for morphology, stability, and cytotoxicity studies against A549 cell-lines. RESULTS Among the nine different NPs formulae (BN1-BN9), BN5 was optimized with composition of BTB (30 mg), PLGA (75 mg), PVA (0.55% w/v), represents an average particle size of (267.1 ± 1.01 nm), PDI (0.101 ± 0.007), and zeta potential (-42.1 ± 0.75 mV), high EE (66.83 ± 0.06%), and DL (6.17 ± 0.69%). SEM image of selected NPs was spherical with smooth surface. In vitro drug release profile in phosphate buffers (pH 5 and pH 7.4) showed a biphasic release with initial burst phase followed by sustained release for prolong time. Furthermore, optimized NPs (BN5) exhibited excellent cytotoxic activity against A549 cell-lines with IC50 value of 5.25 ± 0.23 µg/mL. CONCLUSION The overall results suggest that BTB-loaded PLGA NPs could be a potential nanocarrier for lung cancer treatment.
Collapse
Affiliation(s)
- Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Farhat Fatima
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Mohammed F Aldawsari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Saurabh Bhatia
- Amity Institute of Pharmacy, Amity University Haryana, Gurgaon, India.,Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mauz, Sultanate of Oman
| | - Ahmed Al-Harrasi
- Natural and Medical Sciences Research Center, University of Nizwa, Birkat Al-Mauz, Sultanate of Oman
| |
Collapse
|
22
|
Zhang F, Liu Z, He X, Li Z, Shi B, Cai F. β-Sitosterol-loaded solid lipid nanoparticles ameliorate complete Freund's adjuvant-induced arthritis in rats: involvement of NF-кB and HO-1/Nrf-2 pathway. Drug Deliv 2021; 27:1329-1341. [PMID: 32945205 PMCID: PMC7534215 DOI: 10.1080/10717544.2020.1818883] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Rheumatoid arthritis (RA), autoimmune disease that is categorized via chronic
inflammation manifestation, obesity, cardiovascular risk and even enhanced the mortality
and affect the 0.3 and 1% of population worldwide. The current experimental study was
scrutinize the anti-arthritic effect of β-sitosterol loaded solid lipid nanoparticles
(SLN) against complete Fruend adjuvant (CFA)-induced arthritis via dual pathway. Double
emulsion solvent displacement method was used for the preparation of β-sitosterol solid
lipid nanoparticles (SLN). CFA was used to induce arthritis and rats were divided into
different groups for 28 days. Biochemical, anti-inflammatory, pro-inflammatory cytokines
and inflammatory mediator were estimated, respectively. Receptor activator of nuclear
factor kappa-B ligand (RANKL), signal transducer and activator of transcription-3 (STAT3)
nuclear factor erythroid 2–related factor 2 (Nrf2), Heme Oxygenase-1(HO-1) and
Nuclear factor-κB (NF-κB) expression were estimated. β-sitosterol-SLN significantly
(p < .001) reduced the paw edema, arthritic index and
increased the body weight. β-sitosterol-SLN increased the redox status of synovium {reduce
the malonaldehyde (MDA) and increase superoxide dismutase (SOD), glutathione (GSH) and
catalase (CAT)} level and reduced the cytokines such as tumor necrosis factor-α (TNF-α),
interleukin-1β (IL-1β), interleukin-2, interleukin-6, interleukin-16, interleukin-17 and
increased level of interleukin-10, Transforming growth factor beta (TGF-β).
β-sitosterol-SLN significantly (p < .001) reduced the
level of cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), vascular
Endothelial Growth Factor (VEGF) and NF-κB. β-sitosterol-SLN significantly increased the
expression of HO-1,Nrf2 and decreased the expression of NF-κB, RANKL, STAT3. In
conclusion, β-sitosterol SLN showed the antiarthritic effect via suppression of NF-kB and
activation of HO-1/Nrf-2 pathway.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Orthopedics, Xi'an Fourth Hospital, Xi'an, China
| | - Zhiyu Liu
- Department of Orthopedics, Xi'an Fourth Hospital, Xi'an, China
| | - Xijing He
- Department of Orthopedics, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhanqi Li
- Department of Orthopedics, Xi'an Fourth Hospital, Xi'an, China
| | - Bin Shi
- Department of Orthopedics, Xi'an Fourth Hospital, Xi'an, China
| | - Fengmei Cai
- Department of Pathology, Xi'an Fourth Hospital, Xi'an, China
| |
Collapse
|
23
|
Russo E, Spallarossa A, Tasso B, Villa C, Brullo C. Nanotechnology of Tyrosine Kinase Inhibitors in Cancer Therapy: A Perspective. Int J Mol Sci 2021; 22:6538. [PMID: 34207175 PMCID: PMC8235113 DOI: 10.3390/ijms22126538] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/23/2022] Open
Abstract
Nanotechnology is an important application in modern cancer therapy. In comparison with conventional drug formulations, nanoparticles ensure better penetration into the tumor mass by exploiting the enhanced permeability and retention effect, longer blood circulation times by a reduced renal excretion and a decrease in side effects and drug accumulation in healthy tissues. The most significant classes of nanoparticles (i.e., liposomes, inorganic and organic nanoparticles) are here discussed with a particular focus on their use as delivery systems for small molecule tyrosine kinase inhibitors (TKIs). A number of these new compounds (e.g., Imatinib, Dasatinib, Ponatinib) have been approved as first-line therapy in different cancer types but their clinical use is limited by poor solubility and oral bioavailability. Consequently, new nanoparticle systems are necessary to ameliorate formulations and reduce toxicity. In this review, some of the most important TKIs are reported, focusing on ongoing clinical studies, and the recent drug delivery systems for these molecules are investigated.
Collapse
Affiliation(s)
- Eleonora Russo
- Section of Medicinal and Cosmetic Chemistry, Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-16132 Genova, Italy; (A.S.); (B.T.); (C.V.)
| | | | | | | | - Chiara Brullo
- Section of Medicinal and Cosmetic Chemistry, Department of Pharmacy, University of Genova, Viale Benedetto XV, 3-16132 Genova, Italy; (A.S.); (B.T.); (C.V.)
| |
Collapse
|
24
|
Kavithaa K, Paulpandi M, Ramya S, Ramesh M, Balachandar V, Ramasamy K, Narayanasamy A. Sitosterol-fabricated chitosan nanocomplex induces apoptotic cell death through mitochondrial dysfunction in lung cancer animal model: an enhanced synergetic drug delivery system for lung cancer therapy. NEW J CHEM 2021. [DOI: 10.1039/d1nj00913c] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lung carcinoma is an aggressive form of cancer, with an increasing rate of morbidity, dismal outlook, poor prognosis and limited therapeutic approaches.
Collapse
Affiliation(s)
- Krishnamoorthy Kavithaa
- Disease Proteomics Laboratory
- Department of Zoology
- Bharathiar University
- Coimbatore-641046
- India
| | - Manickam Paulpandi
- Disease Proteomics Laboratory
- Department of Zoology
- Bharathiar University
- Coimbatore-641046
- India
| | - Sennimalai Ramya
- Disease Proteomics Laboratory
- Department of Zoology
- Bharathiar University
- Coimbatore-641046
- India
| | - Mathan Ramesh
- Unit of Toxicology
- Department of Zoology
- School of Life Sciences
- Bharathiar University
- Coimbatore
| | - Vellingiri Balachandar
- Human Molecular Cytogenetics and Stem Cell Laboratory
- Department of Human Genetics and Molecular Biology
- Bharathiar University
- Coimbatore-641046
- India
| | - Karthikeyan Ramasamy
- Department of Biochemistry and Biotechnology
- Annamalai University
- Tamil Nadu – 608002
- India
| | - Arul Narayanasamy
- Disease Proteomics Laboratory
- Department of Zoology
- Bharathiar University
- Coimbatore-641046
- India
| |
Collapse
|
25
|
Acharya S, Praveena J, Guru BR. In Vitro Studies of Prednisolone Loaded PLGA Nanoparticles-Surface Functionalized With Folic Acid on Glioma and Macrophage Cell Lines. PHARMACEUTICAL SCIENCES 2020; 27:407-417. [DOI: 10.34172/ps.2020.94] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/13/2020] [Indexed: 01/03/2025] Open
Abstract
Background: Glucocorticoids are employed for their anti-inflammatory effects in treatingglioma, whose cells are known to overexpress the folate receptors. Some glucocorticoids haveshown inhibitory effects, but the efficacy of prednisolone when delivered via folate receptormediateduptake, has not been attempted. The study aimed to assess the efficacy of targeteddelivery of prednisolone on glioma cell lines like C6 and U87 via the folate receptors. Methods: Targeted delivery of prednisolone was achieved by initially conjugating folic acid (FA)to the di-block copolymer of polylactic acid (PLA) – polyethylene glycol (PEG). This moietycarrying di-block copolymer was incorporated on the surface of the drug-loaded poly lactic-coglycolicacid (PLGA) nanoparticle (NP) by employing the Interfacial Activity Assisted SurfaceFunctionalization (IAASF) technique. The NPs were evaluated for size, zeta potential, and drugloading. It was characterized using particle size analyser, SEM, 1H-NMR, and XRD. cell uptake,cytotoxicity, and anti-inflammatory activities were studied for various formulations. Results: The cytotoxicity assay indicated a high cell growth inhibitory effect of drug encapsulatedNPs with FA moiety as compared to free drug and NPs without the moiety for an incubationperiod of three, five, and six days. The growth-inhibitory effect of the free drug was short-lived,whereas FA functionalized NPs showed higher uptake and sustained inhibitory effect, and werealso able to significantly control the release of pro-inflammatory cytokines like tumour necrosisfactor-alpha (TNF-α) and nitric oxide (NO). Conclusion: Uptake, attenuation of pro-inflammatory signals, and the inhibitory effect ofprednisolone on the cells were more effective when targeted with the FA moiety on the surfaceof NPs as compared to free drug and NPs without the moiety.
Collapse
Affiliation(s)
- Sriprasad Acharya
- Department of Chemical Engineering, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Joyceline Praveena
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Bharath Raja Guru
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| |
Collapse
|
26
|
Raj R K, D E, S R. β‐Sitosterol‐assisted silver nanoparticles activates Nrf2 and triggers mitochondrial apoptosis via oxidative stress in human hepatocellular cancer cell line. J Biomed Mater Res A 2020; 108:1899-1908. [DOI: 10.1002/jbm.a.36953] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/18/2020] [Accepted: 03/28/2020] [Indexed: 12/24/2022]
Affiliation(s)
- Kathiswar Raj R
- Department of Pharmacology, Saveetha Dental CollegeSaveetha Institute of Medical and Technical Sciences (SIMATS) Chennai Tamil Nadu India
| | - Ezhilarasan D
- Department of Pharmacology, Saveetha Dental CollegeSaveetha Institute of Medical and Technical Sciences (SIMATS) Chennai Tamil Nadu India
| | - Rajeshkumar S
- Department of Pharmacology, Saveetha Dental CollegeSaveetha Institute of Medical and Technical Sciences (SIMATS) Chennai Tamil Nadu India
| |
Collapse
|
27
|
Wadhawan A, Chatterjee M, Singh G. Present Scenario of Bioconjugates in Cancer Therapy: A Review. Int J Mol Sci 2019; 20:ijms20215243. [PMID: 31652668 PMCID: PMC6862033 DOI: 10.3390/ijms20215243] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 08/24/2019] [Accepted: 08/30/2019] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the deadliest diseases and poses a risk to people all over the world. Surgery, chemo, and radiation therapy have been the only options available until today to combat this major problem. Chemotherapeutic drugs have been used for treatment for more than 50 years. Unfortunately, these drugs have inherent cytotoxicities and tumor cells have started inducing resistance against these drugs. Other common techniques such as surgery and radiotherapy have their own drawbacks. Therefore, such techniques are incompetent tools to alleviate the disease efficiently without any adverse effects. This scenario has inspired researchers to develop alternative techniques with enhanced therapeutic effects and minimal side effects. Such techniques include targeted therapy, liposomal therapy, hormonal therapy, and immunotherapy, etc. However, these therapies are expensive and not effective enough. Furthermore, researchers have conjugated therapeutic agents or drugs with different molecules, delivery vectors, and/or imaging modalities to combat such problems and enhance the therapeutic effect. This conjugation technique has led to the development of bioconjugation therapy, in which at least one molecule is of biological origin. These bioconjugates are the new therapeutic strategies, having prospective synergistic antitumor effects and have potency to overcome the complications being produced by chemo drugs. Herein, we provide an overview of various bioconjugates developed so far, as well as their classification, characteristics, and targeting approach for cancer. Additionally, the most popular nanostructures based on their organic or inorganic origin (metallic, magnetic, polymeric nanoparticles, dendrimers, and silica nanoparticles) characterized as nanocarriers are also discussed. Moreover, we hope that this review will provide inspiration for researchers to develop better bioconjugates as therapeutic agents.
Collapse
Affiliation(s)
- Aishani Wadhawan
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh Pin code-160014, India.
| | - Mary Chatterjee
- Biotechnology Branch, University Institute of Engineering and Technology, Sector-25, South Campus, Panjab University, Chandigarh Pin code-160014, India.
| | - Gurpal Singh
- Department of Pharmaceutical Sciences, University Institute of Pharmaceutical Sciences, Sector-14, Panjab University, Chandigarh Pin code-160014, India.
| |
Collapse
|
28
|
Recent developments in functionalized polymer nanoparticles for efficient drug delivery system. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.nanoso.2019.100397] [Citation(s) in RCA: 172] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
29
|
Vijayalakshmi S, Mariadoss AVA, Ramachandran V, Shalini V, Agilan B, Sangeetha CC, Balu P, Kotakadi VS, Karthikkumar V, Ernest D. Polydatin Encapsulated Poly [Lactic-co-glycolic acid] Nanoformulation Counteract the 7,12-Dimethylbenz[a] Anthracene Mediated Experimental Carcinogenesis through the Inhibition of Cell Proliferation. Antioxidants (Basel) 2019; 8:E375. [PMID: 31491872 PMCID: PMC6770361 DOI: 10.3390/antiox8090375] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 12/24/2022] Open
Abstract
In the present study, the authors have attempted to fabricate Polydatin encapsulated Poly [lactic-co-glycolic acid] (POL-PLGA-NPs) to counteract 7,12-dimethyl benzyl anthracene (DMBA) promoted buccal pouch carcinogenesis in experimental animals. The bio-formulated POL-PLGA-NPs were characterized by dynamic light scattering (DLS), Fourier transform infrared (FTIR) spectroscopy, X-ray powder diffraction (XRD) pattern analysis, and transmission electron microscope (TEM). In addition, the nano-chemopreventive potential of POL-PLGA-NPs was assessed by scrutinizing the neoplastic incidence and analyzing the status of lipid peroxidation, antioxidants, phase I, phase II detoxification status, and histopathological changes and in DMBA-treated animals. In golden Syrian hamsters, oral squamous cell carcinoma (OSCC) was generated by painting with 0.5% DMBA in liquid paraffin three times a week for 14 weeks. After 100% tumor formation was observed, high tumor volume, tumor burden, and altered levels of biochemical status were observed in the DMBA-painted hamsters. Intra-gastric administration of varying concentration of POL-PLGA-NPs (7.5, 15, and 30 mg/kg b.wt) to DMBA-treated hamsters assumedly prevents oncological incidences and restores the status of the biochemical markers. It also significantly enhances the apoptotic associated and inhibits the cancer cell proliferative markers expression (p53, Bax, Bcl-2, cleaved caspase 3, cyclin-D1). The present study reveals that POL-PLGA-NPs is a penitential candidate for nano-chemopreventive, anti-lipid peroxidative, and antioxidant potential, and also has a modulating effect on the phase I and Phase II detoxification system, which is associated with reduced cell proliferation and induced apoptosis in experimental oral carcinogenesis.
Collapse
Affiliation(s)
- Sankaran Vijayalakshmi
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | | | - Vinayagam Ramachandran
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | - Vijayakumar Shalini
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | - Balupillai Agilan
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | - Casimeer C Sangeetha
- Department of Physics, Sri Padmavati Mahila Visvavidyalayam, Tirupati 517502, Andra Pradesh, India
| | - Periyasamy Balu
- Department of Chemistry, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India
| | | | - Venkatachalam Karthikkumar
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, UAE University, Al Ain 17666, UAE.
| | - David Ernest
- Department of Biotechnology, Thiruvalluvar University, Serkadu, Vellore 632 115, Tamilnadu, India.
| |
Collapse
|
30
|
Mehta A, Dalle Vedove E, Isert L, Merkel OM. Targeting KRAS Mutant Lung Cancer Cells with siRNA-Loaded Bovine Serum Albumin Nanoparticles. Pharm Res 2019; 36:133. [PMID: 31289919 DOI: 10.1007/s11095-019-2665-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 06/25/2019] [Indexed: 12/19/2022]
Abstract
PURPOSE KRAS is the most frequently mutated gene in human cancers. Despite its direct involvement in malignancy and intensive effort, direct inhibition of KRAS via pharmacological inhibitors has been challenging. RNAi induced knockdown using siRNAs against mutant KRAS alleles offers a promising tool for selective therapeutic silencing in KRAS-mutant lung cancers. However, the major bottleneck for clinical translation is the lack of efficient biocompatible siRNA carrier systems. METHODS Bovine serum albumin (BSA) nanoparticles were prepared by desolvation method to deliver siRNA targeting the KRAS G12S mutation. The BSA nanoparticles were characterized with respect to their size, zeta potential, encapsulation efficiency and nucleic acid release. Nanoparticle uptake, cellular distribution of nucleic acids, cytotoxicity and gene knock down to interfere with cancer hallmarks, uncontrolled proliferation and migration, were evaluated in KRAS G12S mutant A459 cells, a lung adenocarcinoma cell line. RESULTS BSA nanoparticles loaded with siRNA resulted in nanoparticles smaller than 200 nm in diameter and negative zeta potentials, displaying optimal characteristics for in vivo application. Encapsulating and protecting the siRNA payload well, the nanoparticles enabled transport to A549 cells in vitro, could evade endosomal entrapment and mediated significant sequence-specific KRAS knockdown, resulting in reduced cell growth of siRNA transfected lung cancer cells. CONCLUSIONS BSA nanoparticles loaded with mutant specific siRNA are a promising therapeutic approach for KRAS-mutant cancers.
Collapse
Affiliation(s)
- Aditi Mehta
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilian University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| | - Elena Dalle Vedove
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilian University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Lorenz Isert
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilian University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany
| | - Olivia M Merkel
- Department of Pharmacy, Pharmaceutical Technology and Biopharmaceutics, Ludwig-Maximilian University of Munich, Butenandtstr. 5-13, 81377, Munich, Germany.
| |
Collapse
|
31
|
PEGylated polylactide (PLA) and poly (lactic-co-glycolic acid) (PLGA) copolymers for the design of drug delivery systems. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2019. [DOI: 10.1007/s40005-019-00442-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|