1
|
Meng K, Tu X, Sun F, Hou L, Shao Z, Wang J. Carbohydrate polymer-based nanoparticles in curcumin delivery for cancer therapy. Int J Biol Macromol 2025; 304:140441. [PMID: 39884595 DOI: 10.1016/j.ijbiomac.2025.140441] [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: 12/01/2024] [Revised: 12/31/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The application of natural products for cancer treatment has a long history. The safety and multifunctionality of naturally occurring substances have made them appropriate for cancer treatment and management. Curcumin affects multiple molecular pathways and is advantageous for treating both hematological and solid tumors. Nonetheless, the effectiveness of curcumin in vivo and in clinical studies has faced challenges due to its poor pharmacokinetic profile. Consequently, nanoparticles have been developed for the administration of curcumin in cancer treatment. The nanoparticles can enhance the distribution of curcumin in tissues and increase its therapeutic effectiveness. Furthermore, nanoparticles expand the uptake of curcumin in cancer cells, leading to increased cytotoxicity. Carbohydrate polymer-based nanoparticles provide a promising solution for the delivery of curcumin in cancer treatment by addressing its low solubility, limited bioavailability, and quick degradation. These biodegradable and biocompatible carriers, originating from polymers such as chitosan, hyaluronic acid, and alginate, protect curcumin, improving its stability and allowing for controlled release. Targeting ligands for functionalization provides selective and specific distribution to the tumor cells, enhancing therapeutic effectiveness and reducing off-target impacts. Their capacity to encapsulate curcumin with other agents allows for synergistic therapies, enhancing anticancer results even more. The adjustable characteristics of carbohydrate nanoparticles, along with their minimal toxicity, develop a revolutionary, functional and safe platform.
Collapse
Affiliation(s)
- Kexin Meng
- Otolaryngology & Head and Neck Center, Cancer Center, Department of Head and Neck Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China; Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, Zhejiang 310014, China; Zhejiang Provincial Clinical Research Center for Malignant Tumor, Hangzhou, Zhejiang 310014, China
| | - Xinzhuo Tu
- Department of Pathology, Air Force Medical Center, PLA, Beijing, China
| | - Feixia Sun
- Nursing Department, Shandong First Medical University Affiliated Occupational Disease Hospital (Shandong Provincial Occupational Disease Hospital), Jinan, China
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer Hospital & Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu 610041, Sichuan, China.
| | - Zhouxiang Shao
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China.
| | - Jinxiang Wang
- Department of Urology, Kidney and Urology Center, Pelvic Floor Disorders Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, Guangdong, China.
| |
Collapse
|
2
|
Pradhan B, Ki JS. Seaweed-derived laminarin and alginate as potential chemotherapeutical agents: An updated comprehensive review considering cancer treatment. Int J Biol Macromol 2025; 293:136593. [PMID: 39426775 DOI: 10.1016/j.ijbiomac.2024.136593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 09/28/2024] [Accepted: 10/12/2024] [Indexed: 10/21/2024]
Abstract
Seaweed-derived bioactive substances such as polysaccharides have proven to be effective chemotherapeutic and chemopreventive agents. Laminarin and alginate antioxidant properties aid in the prevention of cancer through dynamic modulation of critical intracellular signaling pathways via apoptosis which produce low cytotoxicity and potential chemotherapeutic effects. Understanding the effects of laminarin and alginate on human cancer cells and their molecular roles in cell death pathways can help to develop a novel chemoprevention strategy. This review emphasizes the importance of apoptosis-modulating laminarin and alginate in a range of malignancies as well as their extraction, molecular structure, and weight. In addition, future nano-formulation enhancements for greater clinical efficacy are discussed. Laminarin and alginate are perfect ingredients because of their distinct physicochemical and biological characteristics and their use-based delivery systems in cancer. The effectiveness of laminarin and alginate against cancer and more preclinical and clinical trials will open up as new chemotherapeutic natural drugs which lead to established as potential cancer drugs.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea; Department of Botany, Model Degree College, Rayagada 765017, Odisha, India
| | - Jang-Seu Ki
- Department of Life Science, Sangmyung University, Seoul 03016, South Korea.
| |
Collapse
|
3
|
Wang M, Wang Y, Zhang H. Dietary polyphenols for tumor therapy: bioactivities, nano-therapeutic systems and delivery strategies. Food Funct 2025; 16:853-866. [PMID: 39831400 DOI: 10.1039/d4fo04715j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Various dietary polyphenols have demonstrated potent anti-tumor properties and are being evaluated as potential adjuncts in cancer treatment. Although several reviews have offered extensive insights into the anti-tumor activities of dietary polyphenols, they frequently lack a detailed discussion on the design of therapeutic protocols and targeted delivery strategies of these compounds, which impedes the translation of their biological activity into clinical practice. This article aims to deliver a comprehensive review of the anti-tumor properties of dietary polyphenols, while also examining the design and implementation of nanotherapy systems based on these compounds. Additionally, given the challenges of low water solubility and stability of dietary polyphenols, this article outlines the current methodologies for the formulation and delivery of nano-preparations to enhance tumor targeting and therapeutic efficacy. This comprehensive review aspires to deepen our understanding of the operational mechanisms of dietary polyphenols and expand their clinical applications, thereby facilitating the development of polyphenol-based dietary supplements and food additives, and promoting the progress of dietary polyphenol-related nanomedicine.
Collapse
Affiliation(s)
- Minglu Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China.
| | - Ying Wang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China.
| | - Hongyan Zhang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, Key Laboratory of Food Nutrition and Safety of Shandong Normal University, College of Life Sciences, Shandong Normal University, Jinan, 250014, PR China.
| |
Collapse
|
4
|
Bhutta ZA, Choi KC. Phytochemicals as Novel Therapeutics for Triple-Negative Breast Cancer: A Comprehensive Review of Current Knowledge. Phytother Res 2025; 39:364-396. [PMID: 39533509 DOI: 10.1002/ptr.8376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 05/10/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Triple-negative breast cancer is a characteristic subtype of breast cancer that lacks the estrogen receptor, human epidermal growth factor receptor 2, and progesterone receptor. Because of its highly diverse subtypes, increased metastasis capability, and poor prognosis, the risk of mortality for people with triple-negative breast cancers is high as compared with other cancers. Chemotherapy is currently playing a major role in treating triple-negative breast cancer patients; however, poor prognosis due to drug resistance is causing serious concern. Recent studies on several phytochemicals derived from various plants being used in Traditional Chinese Medicine, Traditional Korean Medicine, Ayurveda (Traditional Indian Medicine), and so on, have demonstrated to be a promising agent as a viable therapy against triple-negative breast cancer. Phytochemicals categorized as alkaloids, polyphenols, terpenoids, phytosterols, and organosulfur compounds have been demonstrated to reduce cancer cell proliferation and metastasis by activating various molecular pathways, thereby reducing the spread of triple-negative breast cancer. This review analyzes the molecular mechanisms by which various phytochemicals fight triple-negative breast cancer and offers a perspective on the difficulties and potential prospects for treating triple-negative breast cancer with various phytochemicals.
Collapse
Affiliation(s)
- Zeeshan Ahmad Bhutta
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Republic of Korea
| |
Collapse
|
5
|
Wdowiak K, Tajber L, Miklaszewski A, Cielecka-Piontek J. Application of the Box-Behnken Design in the Development of Amorphous PVP K30-Phosphatidylcholine Dispersions for the Co-Delivery of Curcumin and Hesperetin Prepared by Hot-Melt Extrusion. Pharmaceutics 2024; 17:26. [PMID: 39861675 PMCID: PMC11768460 DOI: 10.3390/pharmaceutics17010026] [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] [Received: 11/28/2024] [Revised: 12/20/2024] [Accepted: 12/25/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Curcumin and hesperetin are plant polyphenols known for their poor solubility. To address this limitation, we prepared amorphous PVP K30-phosphatidylcholine dispersions via hot-melt extrusion. Methods: This study aimed to evaluate the effects of the amounts of active ingredients and phosphatidylcholine, as well as the process temperature, on the performance of the dispersions. A Box-Behnken design was employed to assess these factors. Solid-state characterization and biopharmaceutical studies were then conducted. X-ray powder diffraction (XRPD) was used to confirm the amorphous nature of the dispersions, while differential scanning calorimetry (DSC) provided insight into the miscibility of the systems. Fourier-transform infrared spectroscopy (FTIR) was employed to assess the intermolecular interactions. The apparent solubility and dissolution profiles of the systems were studied in phosphate buffer at pH 6.8. In vitro permeability across the gastrointestinal tract and blood-brain barrier was evaluated using the parallel artificial membrane permeability assay. Results: The quantities of polyphenols and phospholipids were identified as significant factors influencing the biopharmaceutical performance of the systems. Solid-state analysis confirmed the formation of amorphous dispersions and the development of interactions among components. Notably, a significant improvement in solubility was observed, with formulations exhibiting distinct release patterns for the active compounds. Furthermore, the in vitro permeability through the gastrointestinal tract and blood-brain barrier was enhanced. Conclusions: The findings suggest that amorphous PVP K30-phosphatidylcholine dispersions have the potential to improve the biopharmaceutical properties of curcumin and hesperetin.
Collapse
Affiliation(s)
- Kamil Wdowiak
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland;
| | - Lidia Tajber
- School of Pharmacy and Pharmaceutical Sciences, Trinity College Dublin, University of Dublin, D02 PN40 Dublin, Ireland;
| | - Andrzej Miklaszewski
- Faculty of Materials Engineering and Technical Physics, Institute of Materials Science and Engineering, Poznan University of Technology, 5 M. Skłodowska-Curie Square, 60-965 Poznan, Poland;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, 3 Rokietnicka St., 60-806 Poznan, Poland;
| |
Collapse
|
6
|
Manoharan S, Perumal E. A strategic review of STAT3 signaling inhibition by phytochemicals for cancer prevention and treatment: Advances and insights. Fitoterapia 2024; 179:106265. [PMID: 39437855 DOI: 10.1016/j.fitote.2024.106265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 10/15/2024] [Accepted: 10/17/2024] [Indexed: 10/25/2024]
Abstract
Cancer remains a significant global health concern. The dysregulation of signaling networks in tumor cells greatly affects their functions. This review intends to explore phytochemicals possessing potent anticancer properties that specifically target the STAT3 signaling pathway, elucidating strategies and emphasizing their potential as promising candidates for cancer therapy. The review comprehensively examines various STAT3 inhibitors designed to disrupt the signaling cascade, including those targeting upstream activation, SH2 domain phosphorylation, DNA binding domain (DBD), N-terminal domain (NTD), nuclear translocation, and enhancing endogenous STAT3 negative regulators. A literature review was conducted to identify phytochemicals with anticancer activity targeting the STAT3 signaling pathway. Popular research databases such as Google Scholar, PubMed, Science Direct, Scopus, Web of Science, and ResearchGate were searched from the years 1989 - 2023 based on the keywords "Cancer", "STAT3", "Phytochemicals", "Phytochemicals targeting STAT3 signaling", "upstream activation of STAT3", "SH2 domain of STAT3", "DBD of STAT3", "NTD of STAT3, "endogenous negative regulators of STAT3", or "nuclear translocation of STAT3", and their combinations. A total of 264 relevant studies were selected and analyzed based on the mechanisms of action and the efficacy of the phytocompounds. The majority of the discussed phytochemicals primarily focus on inhibiting upstream activation of STAT3. Additionally, flavonoid and terpenoid compounds exhibit multifaceted effects by targeting one or more checkpoints within the STAT3 pathway. Analysis reveals that phytochemicals targeting upstream activation predominantly belong to the classes of flavonoids and terpenoids, which hold significant promise as effective anticancer therapeutics. Future research in this field can be directed towards exploring and developing these scrutinized classes of phytochemicals to achieve desired therapeutic outcomes in cancer treatment.
Collapse
Affiliation(s)
- Suryaa Manoharan
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore 641 046, India.
| |
Collapse
|
7
|
Shahzad A, Liu W, Sun Y, Liu X, Xia J, Cui K, Sai B, Zhu Y, Yang Z, Zhang Q. Flavonoids as modulators of metabolic reprogramming in renal cell carcinoma (Review). Oncol Rep 2024; 52:167. [PMID: 39422066 PMCID: PMC11526433 DOI: 10.3892/or.2024.8826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 08/30/2024] [Indexed: 10/19/2024] Open
Abstract
Renal cell carcinoma (RCC) is distinguished by its varied metabolic reprogramming driven by tumor suppressor gene dysregulation and oncogene activation. Tumors can adapt nutrient uptake and metabolism pathways to meet the altered biosynthetic, bioenergetic and redox demands of cancer cells, whereas conventional chemotherapeutics and molecular inhibitors predominantly target individual metabolic pathways without addressing this adaptability. Flavonoids, which are well‑known for their antioxidant and anti‑inflammatory properties, offer a unique approach by influencing multiple metabolic targets. The present comprehensive review reveals the intricate processes of RCC metabolic reprogramming, encompassing glycolysis, mitochondrial oxidative phosphorylation and fatty acid biosynthesis. The insights derived from the present review may contribute to the understanding of the specific anticancer mechanisms of flavonoids, potentially paving the way for the development of natural antitumor drugs focused on the metabolic reprogramming of RCC.
Collapse
Affiliation(s)
- Asif Shahzad
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Wenjing Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yijian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Xiangjie Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Jiaojiao Xia
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Kun Cui
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Buqing Sai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Yuechun Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| | - Zhe Yang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Qiao Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
8
|
Lu X, Zhang W, Yang X, Yan X, Hussain Z, Wu Q, Zhao J, Yuan B, Yao K, Dong Z, Liu K, Jiang Y. Dronedarone hydrochloride inhibits gastric cancer proliferation in vitro and in vivo by targeting SRC. Transl Oncol 2024; 50:102136. [PMID: 39369581 PMCID: PMC11491965 DOI: 10.1016/j.tranon.2024.102136] [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: 05/03/2024] [Revised: 09/11/2024] [Accepted: 09/19/2024] [Indexed: 10/08/2024] Open
Abstract
BACKGROUND Gastric cancer (GC) is a significant global concern, ranking as the fifth most prevalent cancer. Unfortunately, the five-year survival rate is less than 30 %. Additionally, approximately 50 % of patients experience a recurrence or metastasis. As a result, finding new drugs to prevent relapse is of utmost importance. METHODS The inhibitory effect of Dronedarone hydrochloride (DH) on gastric cancer cells was examined using proliferation assays and anchorage-dependent assays. The binding of DH with SRC was detected by molecular docking, pull-down assays, and cellular thermal shift assays (CETSA). DH's inhibition of Src kinase activity was confirmed through in vitro kinase assays. The SRC knockout cells, established using the CRISPR-Cas9 system, were used to verify Src's role in GC cell proliferation. Patient-derived xenograft (PDX) models were employed to elucidate that DH suppressed tumor growth in vivo. RESULTS Our research discovered DH inhibited GC cell proliferation in vitro and in vivo. DH bound to the SRC protein to inhibit the SRC/AKT1 signaling pathway in gastric cancer. Additionally, we observed a decrease in the sensitivity of gastric cancer cells to DH upon down-regulation of SRC. Notably, we demonstrated DH's anti-tumor effects were similar to those of Dasatinib, a well-known SRC inhibitor, in GC patient-derived xenograft models. CONCLUSION Our research has revealed that Dronedarone hydrochloride, an FDA-approved drug, is an SRC inhibitor that can suppress the growth of GC cells by blocking the SRC/AKT1 signaling pathway. It provides a scientific basis for use in the clinical treatment of GC.
Collapse
Affiliation(s)
- Xuebo Lu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Weizhe Zhang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Xiaoxiao Yang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Xiao Yan
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Zubair Hussain
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Qiong Wu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Jinmin Zhao
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Baoyin Yuan
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China
| | - Ke Yao
- Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China
| | - Zigang Dong
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China
| | - Kangdong Liu
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, Henan, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University 450000, Zhengzhou, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China
| | - Yanan Jiang
- The Pathophysiology Department, The School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450000, China; Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou 450000 Henan, China; China-US (Henan) Hormel Cancer Institute, Zhengzhou 450000, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450000, Henan, China; Provincial Cooperative Innovation Center for Cancer Chemoprevention, Zhengzhou University 450000, Zhengzhou, Henan, China; Cancer Chemoprevention International Collaboration Laboratory, Zhengzhou 450000, Henan, China.
| |
Collapse
|
9
|
Zhao XY, Zhong QH, Tan HW, Yan R, Wang XY, Cai NL, Ji YC, Lau ATY, Xu YM. Non-cytotoxic levels of resveratrol enhance the anticancer effects of cisplatin by increasing the methyltransferase activity of CARM1 in human cancer cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156127. [PMID: 39476485 DOI: 10.1016/j.phymed.2024.156127] [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: 06/12/2024] [Revised: 09/14/2024] [Accepted: 10/02/2024] [Indexed: 12/01/2024]
Abstract
BACKGROUND Resveratrol (RSVL) is a plant-derived polyhydroxyphenolic compound with excellent anticancer properties, alone or in combination with other chemotherapeutic drugs. However, the anticancer mechanism of RSVL is diverse and high concentrations are often required for RSVL to exert its anticancer effect, which would also adversely affect normal cells. PURPOSE The main objective of this study is to investigate the molecular mechanism of how non-cytotoxic concentrations of RSVL enhance the anticancer effect of cisplatin involving a newly identified RSVL-binding protein. METHODS Cell viability of cell lines from three cancer types exposed to RSVL and/or cisplatin was measured by NBB staining assay. RSVL-binding proteins were identified using RSVL-bound CNBr-activated Sepharose 4B beads coupled with LC-MS/MS, and the binding between RSVL and novel RSVL-binding protein was further confirmed with an in vitro pull-down assay. The expression of proteins was examined by immunoblot analysis, and the activity of methyltransferase was evaluated by in vitro methylation assay. The methylation level of H3R17 in the gene promoter was investigated using ChIP-qPCR. Bioinformatics analysis was conducted to identify pathway enrichment of genes, predict drug sensitivity, and analyze the survival of cancer patients. RESULTS Low doses of RSVL might promote cancer cell growth whereas high doses of RSVL showed cytotoxic effects on normal cells. When co-treated with a lower cisplatin dose, non-cytotoxic RSVL levels showed synergistic anticancer effects. Here, coactivator-associated arginine methyltransferase 1 (CARM1) was identified as a novel RSVL-binding protein, and we showed that the upregulation of CARM1 increased the sensitivity of cancer cells to RSVL. Interestingly, we found that CARM1 was essential in the RSVL-induced sensitivity of cisplatin. Further molecular mechanistic studies revealed that RSVL could stabilize CARM1 protein, resulting in the upregulation and increased methyltransferase activity of CARM1. Additionally, we showed that the methylation levels of H3R17 in the promoter of p21, a downstream gene of CARM1 involving cell cycle arrest, were significantly increased after RSVL treatment. Finally, data from our bioinformatics analysis suggested that CARM1 could be utilized as a potential biomarker for chemotherapeutic drug sensitivity and prognosis in cancers. CONCLUSIONS This study identified CARM1 as a RSVL-binding protein for the first time and elucidated the potential roles of CARM1 in enhancing the efficacy of cisplatin by low doses of RSVL, which could have important clinical implications.
Collapse
Affiliation(s)
- Xiao-Yun Zhao
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Qiu-Hua Zhong
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Heng Wee Tan
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Rui Yan
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Xiu-Yun Wang
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Na-Li Cai
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Yan-Chen Ji
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China
| | - Andy T Y Lau
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China.
| | - Yan-Ming Xu
- Clinical Research Center, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, PR China; Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and Genetics, Shantou University Medical College, Shantou, Guangdong 515041, PR China.
| |
Collapse
|
10
|
Chen Y, Tang Y, Li Y, Rui Y, Zhang P. Enhancing the Efficacy of Active Pharmaceutical Ingredients in Medicinal Plants through Nanoformulations: A Promising Field. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1598. [PMID: 39404324 PMCID: PMC11478102 DOI: 10.3390/nano14191598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
This article explores the emerging field of nanomedicine as a drug delivery system, aimed at enhancing the therapeutic efficacy of active pharmaceutical ingredients in medicinal plants. The traditional methods of applying medicinal plants present several limitations, such as low bioavailability, poor solubility, challenges in accurately controlling drug dosage, and inadequate targeting. Nanoformulations represent an innovative approach in drug preparation that employs nanotechnology to produce nanoscale particles or carriers, which are designed to overcome these limitations. Nanoformulations offer distinct advantages, significantly enhancing the solubility and bioavailability of drugs, particularly for the poorly soluble components of medicinal plants. These formulations effectively enhance solubility, thereby facilitating better absorption and utilization by the human body, which in turn improves drug efficacy. Furthermore, nanomedicine enables targeted drug delivery, ensuring precise administration to the lesion site and minimizing side effects on healthy tissues. Additionally, nanoformulations can regulate drug release rates, extend the duration of therapeutic action, and enhance the stability of treatment effects. However, nanoformulations present certain limitations and potential risks; their stability and safety require further investigation, particularly regarding the potential toxicity with long-term use. Nevertheless, nanomaterials demonstrate substantial potential in augmenting the efficacy of active pharmaceutical ingredients in medicinal plants, offering novel approaches and methodologies for their development and application.
Collapse
Affiliation(s)
- Yuhao Chen
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuying Tang
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yuanbo Li
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
| | - Yukui Rui
- Beijing Key Laboratory of Farmland Soil Pollution Prevention and Remediation, College of Resources and Environmental Sciences, China Agricultural University, Beijing 100093, China; (Y.C.); (Y.T.); (Y.L.)
- Tangshan Jinhai New Material Co., Ltd., Tangshan 063000, China
- Faculty of Resources and Environment, China Agricultural University, Shanghe County Baiqiao Town Science and Technology Courtyard, Jinan 250100, China
| | - Peng Zhang
- Department of Environmental Science and Engineering, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
11
|
Wang K, He Z, Jin G, Jin S, Du Y, Yuan S, Zhang J. Targeting DNA methyltransferases for cancer therapy. Bioorg Chem 2024; 151:107652. [PMID: 39024804 DOI: 10.1016/j.bioorg.2024.107652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/29/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
DNA methyltransferases (DNMTs) play a crucial role in genomic DNA methylation. In mammals, DNMTs regulate the dynamic patterns of DNA methylation in embryonic and adult cells. Abnormal functions of DNMTs are often indicative of cancers, including overall hypomethylation and partial hypermethylation of tumor suppressor genes (TSG), which accelerate the malignancy of tumors, worsen the condition of patients, and significantly exacerbate the difficulty of cancer treatment. Currently, nucleoside DNMT inhibitors such as Azacytidine and Decitabine have been approved by the FDA and EMA for the treatment of acute myeloid leukemia (AML), chronic myelomonocytic leukemia (CMML), and myelodysplastic syndrome (MDS). Therefore, targeting DNMTs is a very promising anti-tumor strategy. This review mainly summarizes the therapeutic effects of DNMT inhibitors on cancers. It aims to provide more possibilities for the treatment of cancers by discovering more DNMT inhibitors with high activity, high selectivity, and good drug-like properties in the future.
Collapse
Affiliation(s)
- Kaiyue Wang
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China
| | - Zhangxu He
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China.
| | - Gang Jin
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China
| | - Sasa Jin
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China
| | - Yuanbing Du
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China
| | - Shuo Yuan
- Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, PR China.
| | - Jingyu Zhang
- Pharmacy College, Henan University of Chinese Medicine, 450046 Zhengzhou, PR China.
| |
Collapse
|
12
|
容 逍, 向 茜, 赵 羿, 邱 逦, 杜 方. [Experimental Study on Biomimetic Curcumin-Mediated Sonodynamic Therapy of Melanoma]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2024; 55:1159-1165. [PMID: 39507956 PMCID: PMC11536226 DOI: 10.12182/20240960108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Indexed: 11/08/2024]
Abstract
Objective To study the role of curcumin-mediated sonodynamic therapy in the treatment of malignant melanoma, and to provide a new strategy for the treatment of malignant melanoma. Methods The ultrasonic sound and vibration method was applied to coat curcumin with mouse melanoma cell membrane, thereby forming biomimetic curcumin. The morphology of biomimetic curcumin was observed by transmission electron microscope. Flow cytometry was used to analyze the effect of biomimetic curcumin in terms of in vitro targeting, apoptosis, and intracellular reactive oxygen species (ROS) production. The in vivo experiment was divided into control group, US group, turmeric group, imitation turmeric group, and imitation turmeric+US group, with 3 mice in each group. The in vivo safety of biomimetic curcumin was evaluated by HE staining. In addition, HE, CD31, Ki67, and TUNEL stainings were performed to evaluate the in vivo anti-melanoma therapeutic effect of ultrasound combined with biomimetic curcumin. Results The biomimetic curcumin had a generally uniform morphology and possessed a core-shell structure. Flow cytometry analysis performed with FlowJo showed that the biomimetic curcumin could be effectively taken up by melanoma cells. The apoptosis rate was (10.30±0.61)% in the control group, (10.41±3.13)% in the ultrasound group, (24.97±1.38)% in the curcumin group, (31.39±3.84)% in the biomimetic curcumin group, and (40.89±0.79)% in the biomimetic curcumin and ultrasound combination group. The apoptosis rate in the biomimetic curcumin and ultrasound combination group was higher than those in the other groups (P<0.05). The results of ROS flow cytometry showed that, compared with the control group, the ultrasound group demonstrated almost no increase in the fluorescence intensity, while the other groups showed an increase in the fluorescence intensity to varying degrees. There was no significant difference in the fluorescence intensity between the biomimetic curcumin group ([1.10±0.38]%) and the curcumin group ([0.73±0.26]%) (P>0.05). The fluorescence intensity of the biomimetic curcumin and ultrasound combination group ([3.35±0.04]%) was higher than those of the other groups (P<0.05). HE staining showed no obvious abnormalities in the morphology of heart, liver, spleen, lung, and kidney tissues in any of the treatment groups. HE staining showed the most significant changes in cell morphology in the biomimetic curcumin and ultrasound combination group, followed by the biomimetic curcumin group and the curcumin group. No obvious abnormalities in tumor cell morphology were observed in the ultrasound group. According to the respective results of CD31 staining, Ki67 staining, and TUNEL staining, the biomimetic curcumin and ultrasound combination group had the largest brown area, the highest number of red fluorescence, and the highest number of green fluorescence, followed by the biomimetic curcumin group and the curcumin group. Conclusion The biomimetic curcumin displays uniform morphology, a core-shell structure, and good targeting properties. When it is used in combination with ultrasound, biomimetic curcumin demonstrates a good anti-tumor therapeutic effect both in vivo and in vitro.
Collapse
Affiliation(s)
- 逍 容
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 茜 向
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 羿丞 赵
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 逦 邱
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 方雪 杜
- 四川大学华西医院 超声医学科 (成都 610041)Department of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
13
|
Di Carlo E, Sorrentino C. Oxidative Stress and Age-Related Tumors. Antioxidants (Basel) 2024; 13:1109. [PMID: 39334768 PMCID: PMC11428699 DOI: 10.3390/antiox13091109] [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] [Received: 07/19/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Oxidative stress is the result of the imbalance between reactive oxygen and nitrogen species (RONS), which are produced by several endogenous and exogenous processes, and antioxidant defenses consisting of exogenous and endogenous molecules that protect biological systems from free radical toxicity. Oxidative stress is a major factor in the aging process, contributing to the accumulation of cellular damage over time. Oxidative damage to cellular biomolecules, leads to DNA alterations, lipid peroxidation, protein oxidation, and mitochondrial dysfunction resulting in cellular senescence, immune system and tissue dysfunctions, and increased susceptibility to age-related pathologies, such as inflammatory disorders, cardiovascular and neurodegenerative diseases, diabetes, and cancer. Oxidative stress-driven DNA damage and mutations, or methylation and histone modification, which alter gene expression, are key determinants of tumor initiation, angiogenesis, metastasis, and therapy resistance. Accumulation of genetic and epigenetic damage, to which oxidative stress contributes, eventually leads to unrestrained cell proliferation, the inhibition of cell differentiation, and the evasion of cell death, providing favorable conditions for tumorigenesis. Colorectal, breast, lung, prostate, and skin cancers are the most frequent aging-associated malignancies, and oxidative stress is implicated in their pathogenesis and biological behavior. Our aim is to shed light on the molecular and cellular mechanisms that link oxidative stress, aging, and cancers, highlighting the impact of both RONS and antioxidants, provided by diet and exercise, on cellular senescence, immunity, and development of an antitumor response. The dual role of ROS as physiological regulators of cell signaling responsible for cell damage and diseases, as well as its use for anti-tumor therapeutic purposes, will also be discussed. Managing oxidative stress is crucial for promoting healthy aging and reducing the risk of age-related tumors.
Collapse
Affiliation(s)
- Emma Di Carlo
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| | - Carlo Sorrentino
- Department of Medicine and Sciences of Aging, "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
- Anatomic Pathology and Immuno-Oncology Unit, Center for Advanced Studies and Technology (CAST), "G. d'Annunzio" University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
14
|
Siripruekpong W, Praparatana R, Issarachot O, Wiwattanapatapee R. Simultaneous Delivery of Curcumin and Resveratrol via In Situ Gelling, Raft-Forming, Gastroretentive Formulations. Pharmaceutics 2024; 16:641. [PMID: 38794303 PMCID: PMC11124977 DOI: 10.3390/pharmaceutics16050641] [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] [Received: 03/21/2024] [Revised: 05/05/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Curcumin and resveratrol are polyphenolic compounds that have been shown to exhibit synergistic therapeutic properties including anti-inflammatory, anticancer, and antiulcer activities, which may be exploited for the treatment of gastric diseases. However, both compounds have poor aqueous solubility and rapid metabolism, resulting in a low oral bioavailability. In situ gelling, liquid formulations were developed to produce a gastroretentive, raft-forming delivery vehicle to improve bioavailability. Solid dispersions containing a mixture of curcumin and resveratrol with Eudragit® EPO (Cur/Res-SD) were first prepared using solvent evaporation, to improve the solubility and dissolution of the compounds. Solid dispersions of a weight ratio of 1:10 curcumin/resveratrol to Eudragit® EPO were subsequently incorporated into in situ gelling, liquid formulations based on the gelling polymers, sodium alginate (low viscosity and medium viscosity), pectin, and gellan gum, respectively. Calcium carbonate and sodium bicarbonate were included to produce carbon dioxide bubbles in the gel matrix, on exposure to gastric fluid, and to achieve flotation. Moreover, the calcium ions acted as a crosslinking agent for the hydrogels. Optimized formulations floated rapidly (<60 s) in simulated gastric fluid (pH = 1.2) and remained buoyant, resulting in the gradual release of more than 80% of the curcumin and resveratrol content within 8 h. The optimized formulation based on medium-viscosity sodium alginate exhibited enhanced cytotoxic activity toward human gastric adenocarcinoma cell lines (AGS), compared with unformulated curcumin and resveratrol compounds, and increased anti-inflammatory activity against RAW 264.7 macrophage cells compared with the NSAID, indomethacin. These findings demonstrate that in situ gelling, liquid formulations, loaded with a combination of curcumin and resveratrol in the form of solid dispersions, show potential as gastroretentive delivery systems for local and systemic effects.
Collapse
Affiliation(s)
- Worrawee Siripruekpong
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hatyai, Songkhla 90112, Thailand;
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hatyai, Songkhla 90112, Thailand
| | - Rachanida Praparatana
- Faculty of Medical Technology, Prince of Songkla University, Hatyai, Songkhla 90112, Thailand;
| | - Ousanee Issarachot
- Department of Pharmacy Technician, Faculty of Public Health and Allied Health Sciences, Sirindhorn College of Public Health Trang, Praboromarajchanok Institute, Kantang, Trang 92110, Thailand;
| | - Ruedeekorn Wiwattanapatapee
- Department of Pharmaceutical Technology, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hatyai, Songkhla 90112, Thailand;
- Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hatyai, Songkhla 90112, Thailand
| |
Collapse
|
15
|
Ganie SA, Naik RA, Dar OA, Rather LJ, Assiri MA, Li Q. Design and fabrication of functionalized curdlan-curcumin delivery system to facilitate the therapeutic effects of curcumin on breast cancer. Int J Biol Macromol 2024; 267:131388. [PMID: 38608982 DOI: 10.1016/j.ijbiomac.2024.131388] [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: 12/04/2023] [Revised: 03/23/2024] [Accepted: 04/03/2024] [Indexed: 04/14/2024]
Abstract
We developed a facile method for the fabrication of a biodegradable delivery system composed of two blocks: curdlan and curcumin. This was achieved by chemical functionalization of curdlan through tosylation, amination followed by complexation with curcumin. A comprehensive evaluation of structural characterization and component stability showed that cur-cum complex exhibited better anticancer properties with enhanced thermal properties. The cur-cum complex shows pH sensitive sustained release behaviour with higher release at acidic pH and kinetic data of drug release follows the Korsmeyer-Peppas model. The cur-cum complex has ability to block the proliferation of the MCF-7 cell line as revealed by MTT assay which showed increased toxicity of cur-cum complex against these cell lines. The results obtained from western blot analysis demonstrated that the co-administration of cur and cum effectively induced apoptosis in MCF-7 cells. This effect was observed by a considerable upregulation of the Bcl-2/Bax ratio, a decline in mRNA expression of LDHA, level of lactate and LDH activity. The results clearly depict the role of functionalized curdlan as efficient carrier for curcumin delivery with prolonged, sustained release and enhanced bioavailability, thereby improving the overall anticancer activity.
Collapse
Affiliation(s)
- Showkat Ali Ganie
- State Key Laboratory of Resource Insects, Chongqing Engineering Research Centre for Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, 400715 Chongqing, PR China.
| | - Rayees Ahmad Naik
- Department of Zoology, Dr. Harisingh Gour Vishwavidyalaya Sagar, Madhya Pradesh 470003, India
| | - Ovas Ahmad Dar
- College of Pharmaceutical Sciences, Southwest University, 400715 Chongqing, PR China.
| | - Luqman Jameel Rather
- State Key Laboratory of Resource Insects, Chongqing Engineering Research Centre for Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, 400715 Chongqing, PR China
| | - Mohammed A Assiri
- Department of Chemistry, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia.
| | - Qing Li
- State Key Laboratory of Resource Insects, Chongqing Engineering Research Centre for Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, 400715 Chongqing, PR China.
| |
Collapse
|
16
|
Otun S, Achilonu I, Odero-Marah V. Unveiling the potential of Muscadine grape Skin extract as an innovative therapeutic intervention in cancer treatment. J Funct Foods 2024; 116:106146. [PMID: 38817632 PMCID: PMC11139022 DOI: 10.1016/j.jff.2024.106146] [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] [Indexed: 06/01/2024] Open
Abstract
The use of muscadine grape extracts (MGSE). in cancer treatment has gained attention due to its distinctive composition of polyphenols and antioxidants. This review analyses the reported anti-cancer properties of MGSE. The study commences by reviewing the phytochemical composition of MGSE, highlighting the presence of resveratrol and ellagic acid. Furthermore, the review underscores the mechanism of action of these active compounds in MGSE in combating cancer cells. The anti-cancer potential of MGSE compared to other plant extracts is also discussed. In addition, it highlights MGSE's superiority and distinct phytochemical composition in preventing cancer growth by comparing its anti-cancer compounds with those of other anti-cancer medicinal plants. Lastly, the combinatory approaches of MGSE with traditional cancer therapies, its safety, and its possible side effects were highlighted. This work provides an understanding of the anti-cancer properties of MGSE, positioning it as a valuable and unique challenge within the field of cancer therapy.
Collapse
Affiliation(s)
- Sarah Otun
- School of Molecular and Cell Biology, Faculty of Science, Protein Structure-Function and Research Unit, University of the Witwatersrand, Braamfontein, Johannesburg, South Africa
| | - Ikechukwu Achilonu
- School of Molecular and Cell Biology, Faculty of Science, Protein Structure-Function and Research Unit, University of the Witwatersrand, Braamfontein, Johannesburg, South Africa
| | - Valerie Odero-Marah
- Center for Urban Health Disparities Research and Innovation, Department of Biology, Morgan State University, Baltimore MD 21251, United States
| |
Collapse
|
17
|
Kaur G, Kaur R, Sodhi GK, George N, Rath SK, Walia HK, Dwibedi V, Saxena S. Stilbenes: a journey from folklore to pharmaceutical innovation. Arch Microbiol 2024; 206:229. [PMID: 38647675 DOI: 10.1007/s00203-024-03939-z] [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: 01/11/2024] [Revised: 03/03/2024] [Accepted: 03/22/2024] [Indexed: 04/25/2024]
Abstract
In modern times, medicine is predominantly based on evidence-based practices, whereas in ancient times, indigenous people relied on plant-based medicines with factual evidence documented in ancient books or folklore that demonstrated their effectiveness against specific infections. Plants and microbes account for 70% of drugs approved by the USFDA (U.S. Food and Drug Administration). Stilbenes, polyphenolic compounds synthesized by plants under stress conditions, have garnered significant attention for their therapeutic potential, bridging ancient wisdom with modern healthcare. Resveratrol, the most studied stilbene, initially discovered in grapes, red wine, peanuts, and blueberries, exhibits diverse pharmacological properties, including cardiovascular protection, antioxidant effects, anticancer activity, and neuroprotection. Traditional remedies, documented in ancient texts like the Ayurvedic Charak Samhita, foreshadowed the medicinal properties of stilbenes long before their modern scientific validation. Today, stilbenes are integral to the booming wellness and health supplement market, with resveratrol alone projected to reach a market value of 90 million US$ by 2025. However, challenges in stilbene production persist due to limited natural sources and costly extraction methods. Bioprospecting efforts reveal promising candidates for stilbene production, particularly endophytic fungi, which demonstrate high-yield capabilities and genetic modifiability. However, the identification of optimal strains and fermentation processes remains a critical consideration. The current review emphasizes the knowledge of the medicinal properties of Stilbenes (i.e., cardiovascular, antioxidant, anticancer, anti-inflammatory, etc.) isolated from plant and microbial sources, while also discussing strategies for their commercial production and future research directions. This also includes examples of novel stilbenes compounds reported from plant and endophytic fungi.
Collapse
Affiliation(s)
- Gursharan Kaur
- University Institute of Biotechnology, Chandigarh University, Mohali, 140413, Punjab, India
| | - Rajinder Kaur
- Department of Plant Sciences, University of Idaho Moscow, Idaho, ID, 83844, USA
| | - Gurleen Kaur Sodhi
- Department of Biotechnology, Thapar Institute of Engineering and Technology Patiala, Patiala, Punjab, 147004, India
| | - Nancy George
- University Institute of Biotechnology, Chandigarh University, Mohali, 140413, Punjab, India
| | - Santosh Kumar Rath
- School of Pharmaceuticals and Population Health Informatics, Faculty of Pharmacy, DIT University, Dehradun, Uttarakhand, 248009, India
| | - Harleen Kaur Walia
- Department of Biotechnology, Thapar Institute of Engineering and Technology Patiala, Patiala, Punjab, 147004, India
| | - Vagish Dwibedi
- University Institute of Biotechnology, Chandigarh University, Mohali, 140413, Punjab, India.
- Institute of Soil, Water and Environmental Sciences, Volcani Research Center, Agricultural Research Organization, 7505101, Rishon LeZion, Israel.
| | - Sanjai Saxena
- Department of Biotechnology, Thapar Institute of Engineering and Technology Patiala, Patiala, Punjab, 147004, India
| |
Collapse
|
18
|
Entezari M, Tayari A, Paskeh MDA, Kheirabad SK, Naeemi S, Taheriazam A, Dehghani H, Salimimoghadam S, Hashemi M, Mirzaei S, Samarghandian S. Curcumin in treatment of hematological cancers: Promises and challenges. J Tradit Complement Med 2024; 14:121-134. [PMID: 38481552 PMCID: PMC10927384 DOI: 10.1016/j.jtcme.2023.10.004] [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: 03/31/2023] [Revised: 09/16/2023] [Accepted: 10/19/2023] [Indexed: 11/01/2024] Open
Abstract
Hematological cancers include leukemia, myeloma and lymphoma and up to 178.000 new cases are diagnosed with these tumors each year. Different kinds of treatment including radiotherapy, chemotherapy, immunotherapy and stem cell transplantation have been employed in the therapy of hematological cancers. However, they are still causing death among patients. On the other hand, curcumin as an anti-cancer agent for the suppression of human cancers has been introduced. The treatment of hematological cancers using curcumin has been followed. Curcumin diminishes viability and survival rate of leukemia, myeloma and lymphoma cells. Curcumin stimulates apoptosis and G2/M arrest to impair progression of tumor. Curcumin decreases levels of matrix metalloproteinases in suppressing cancer metastasis. A number of downstream targets including VEGF, Akt and STAT3 undergo suppression by curcumin in suppressing progression of hematological cancers. Curcumin stimulates DNA damage and reduces resistance of cancer cells to irradiation. Furthermore, curcumin causes drug sensitivity of hematological tumors, especially myeloma. For targeted delivery of curcumin and improving its pharmacokinetic and anti-cancer features, nanostructures containing curcumin and other anti-cancer agents have been developed.
Collapse
Affiliation(s)
- Maliheh Entezari
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Armita Tayari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Simin Khorsand Kheirabad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sahar Naeemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Orthopedics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Dehghani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Medical Laboratory Sciences, Islamic Azad University, Tehran Medical Sciences, Tehran, Iran
| | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Mehrdad Hashemi
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
19
|
Chang Z, Wu Y, Hu P, Jiang J, Quan G, Wu C, Pan X, Huang Z. The Necessity to Investigate In Vivo Fate of Nanoparticle-Loaded Dissolving Microneedles. Pharmaceutics 2024; 16:286. [PMID: 38399340 PMCID: PMC10892231 DOI: 10.3390/pharmaceutics16020286] [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] [Received: 12/28/2023] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Transdermal drug delivery systems are rapidly gaining prominence and have found widespread application in the treatment of numerous diseases. However, they encounter the challenge of a low transdermal absorption rate. Microneedles can overcome the stratum corneum barrier to enhance the transdermal absorption rate. Among various types of microneedles, nanoparticle-loaded dissolving microneedles (DMNs) present a unique combination of advantages, leveraging the strengths of DMNs (high payload, good mechanical properties, and easy fabrication) and nanocarriers (satisfactory solubilization capacity and a controlled release profile). Consequently, they hold considerable clinical application potential in the precision medicine era. Despite this promise, no nanoparticle-loaded DMN products have been approved thus far. The lack of understanding regarding their in vivo fate represents a critical bottleneck impeding the clinical translation of relevant products. This review aims to elucidate the current research status of the in vivo fate of nanoparticle-loaded DMNs and elaborate the necessity to investigate the in vivo fate of nanoparticle-loaded DMNs from diverse aspects. Furthermore, it offers insights into potential entry points for research into the in vivo fate of nanoparticle-loaded DMNs, aiming to foster further advancements in this field.
Collapse
Affiliation(s)
- Ziyao Chang
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (Y.W.); (X.P.)
| | - Yuhuan Wu
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (Y.W.); (X.P.)
| | - Ping Hu
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Junhuang Jiang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou 510006, China; (Z.C.); (Y.W.); (X.P.)
| | - Zhengwei Huang
- College of Pharmacy, Jinan University, Guangzhou 511443, China; (P.H.); (G.Q.); (C.W.)
| |
Collapse
|
20
|
Yamashita M, Nakanishi A, Chang C, Tsurushima K, Nakamoto K, Iida A. Evaluation of STAT3 Inhibition by Cancer Chemopreventive Trichothecenes Derived from Metabolites of Trichothecium roseum. Chem Pharm Bull (Tokyo) 2024; 72:693-699. [PMID: 39048375 DOI: 10.1248/cpb.c24-00300] [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] [Indexed: 07/27/2024]
Abstract
This study evaluated the ability of isolated or semisynthesized trichothecene sesquiterpenes to prevent cancer emergence and proliferation and inhibit signal transducer and activator of transcription-3 (STAT3) phosphorylation through in vitro assays. Trichothecinol A (TTC-A), which bears a hydroxy group at C3, exhibited greater cancer prevention, antiproliferation, and STAT3 phosphorylation inhibition effects than trichothecin (TTC), which lacks a hydroxy group at C3. Furthermore, trichothecinol B (TTC-B), which is a reduced derivative of TTC and has similar cytotoxic effect, showed substantially weaker chemoprotection and STAT3 phosphorylation inhibition effects than TTC. These results clearly indicate that the hydroxy group at C3 and carbonyl group at C8 are crucial for inducing both potent chemoprevention and STAT3 phosphorylation inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | - Akira Iida
- Graduate School of Agriculture, Kindai University
| |
Collapse
|
21
|
Jurczyk M, Musiał-Kulik M, Foryś A, Godzierz M, Kaczmarczyk B, Kasperczyk J, Wrześniok D, Beberok A, Jelonek K. Comparison of PLLA-PEG and PDLLA-PEG micelles for co-encapsulation of docetaxel and resveratrol. J Biomed Mater Res B Appl Biomater 2024; 112:e35318. [PMID: 37650461 DOI: 10.1002/jbm.b.35318] [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: 05/08/2023] [Revised: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 09/01/2023]
Abstract
The interest in combining chemosensitizers with cytostatics in cancer therapy is growing, which causes also a need to develop their delivery systems. Example of the combination with beneficial therapeutic effects is docetaxel (Dtx) and resveratrol (Res). Although poly(lactide)-co-poly(ethylene glycol) (PLA-PEG) micelles have been considered as one of the most promising platforms for drug delivery, their properties may depend on the stereoisomeric form of hydrophobic block. Therefore, the aim of this study was evaluation of the effect of PLA block on co-encapsulation and release rate of Dtx and Res, which has not been studied so far. This article presents a comparison of single- (Dtx or Res) and dual-drug (Dtx and Res) loaded micelles obtained from poly(l,l-lactide)-co-poly(ethylene glycol) (PLLA-PEG) and poly(d,l-lactide)-co-poly(ethylene glycol) (PDLLA-PEG). The analyzes of the micelles have been conducted including morphology, drug(s) encapsulation efficiency, intermolecular interactions, in vitro drug release, and cytotoxicity. Differences in drug loading ability and release profile have been observed between Res and Dtx but also depending on the polymer and number of drugs in micelles (single vs. dual loaded). The PLLA-PEG micelles have a significantly higher Dtx encapsulation capacity than PDLLA-PEG micelles. The highest cytotoxicity was shown for Dtx and Res dual-loaded micelles, regardless of the polymer. The findings may be used for selection of PLA-based drug delivery systems containing Dtx and Res.
Collapse
Affiliation(s)
- Magdalena Jurczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Sosnowiec, Poland
| | - Monika Musiał-Kulik
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Aleksander Foryś
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Marcin Godzierz
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Bożena Kaczmarczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| | - Janusz Kasperczyk
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
- Department of Biopharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Sosnowiec, Poland
| | - Dorota Wrześniok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Sosnowiec, Poland
| | - Artur Beberok
- Department of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Sosnowiec, Poland
| | - Katarzyna Jelonek
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, Zabrze, Poland
| |
Collapse
|
22
|
Sun X, Li F, Yuan L, Bing Z, Li X, Yang K. pH-responsive resveratrol-loaded ZIF-8 nanoparticles modified with tannic acid for promoting colon cancer cell apoptosis. J Biomed Mater Res B Appl Biomater 2024; 112:e35320. [PMID: 37702969 DOI: 10.1002/jbm.b.35320] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 07/30/2023] [Accepted: 08/16/2023] [Indexed: 09/14/2023]
Abstract
Resveratrol (Res) is known for its potential in treating various types of cancers, with a particular advantage of causing minimal toxic side effects. However, its clinical application is constrained by challenges such as poor bioavailability, low water solubility, and chemical instability in neutral and alkaline environments. In light of these limitations, we have developed a pH-responsive drug delivery nanoplatform, Res@ZIF-8/TA NPs, which exhibits good biocompatibility and shows promise for in vitro cancer therapy. Benefiting from the mild reaction conditions provided by zeolitic imidazolate frameworks (ZIFs), a "one-pot method" was used for drug synthesis and loading, resulting in a satisfactory loading capacity. Notably, Res@ZIF-8/TA NPs respond to acidic environments, leading to an improved drug release profile with a controlled release effect. Our cell-based experiments indicated that tannic acid (TA) modification enhances the biocompatibility of ZIFs. 3-(4,5)-dimethylthiahiazo (-z-y1)-3,5-di- phenytetrazoliumromide (MTT assay), Hoechst 33342/PI staining, cell scratch assay, Transwell and Reverse Transcription quantitative PCR (RT-qPCR) assays further demonstrated that Res@ZIF-8/TA NPs inhibited colon cancer cell migration and invasion, and promoted apoptosis of colon cancer cells, suggesting a therapeutic potential and demonstrating anti-cancer properties. In conclusion, the Res@ZIF-8/TA NPs pH-responsive drug delivery systems we developed may offer a promising avenue for cancer therapy. By addressing some of the challenges associated with Res-based treatments, this system could contribute to advancements in cancer therapeutics.
Collapse
Affiliation(s)
- Xueqiang Sun
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China
- Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Fuxin Li
- The People's Hospital of Hezhou Hepatobiliary, Pancreatic and Spleen Surgery, Hezhou, China
| | - Lingyan Yuan
- Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Zhitong Bing
- Department of Computational Physics, Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou, China
| | - Xun Li
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Kehu Yang
- The First Hospital of Lanzhou University, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
- Evidence Based Medicine Center, School of Basic Medical Science of Lanzhou University, Lanzhou, China
- Key Laboratory of Evidence Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
- Institution of Clinical Research and Evidence Based Medicine, Gansu Provincial Hospital, Lanzhou, China
- School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
Focaccetti C, Palumbo C, Benvenuto M, Carrano R, Melaiu O, Nardozi D, Angiolini V, Lucarini V, Kërpi B, Masuelli L, Cifaldi L, Bei R. The Combination of Bioavailable Concentrations of Curcumin and Resveratrol Shapes Immune Responses While Retaining the Ability to Reduce Cancer Cell Survival. Int J Mol Sci 2023; 25:232. [PMID: 38203402 PMCID: PMC10779126 DOI: 10.3390/ijms25010232] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
The polyphenols Curcumin (CUR) and Resveratrol (RES) are widely described for their antitumoral effects. However, their low bioavailability is a drawback for their use in therapy. The aim of this study was to explore whether CUR and RES, used at a bioavailable concentration, could modulate immune responses while retaining antitumor activity and to determine whether CUR and RES effects on the immune responses of peripheral blood mononuclear cells (PBMCs) and tumor growth inhibition could be improved by their combination. We demonstrate that the low-dose combination of CUR and RES reduced the survival of cancer cell lines but had no effect on the viability of PBMCs. Although following CUR + RES treatment T lymphocytes showed an enhanced activated state, RES counteracted the increased IFN-γ expression induced by CUR in T cells and the polyphenol combination increased IL-10 production by T regulatory cells. On the other hand, the combined treatment enhanced NK cell activity through the up- and downregulation of activating and inhibitory receptors and increased CD68 expression levels on monocytes/macrophages. Overall, our results indicate that the combination of CUR and RES at low doses differentially shapes immune cells while retaining antitumor activity, support the use of this polyphenol combinations in anticancer therapy and suggest its possible application as adjuvant for NK cell-based immunotherapies.
Collapse
Affiliation(s)
- Chiara Focaccetti
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
| | - Camilla Palumbo
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
| | - Monica Benvenuto
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
- Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
| | - Raffaele Carrano
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
| | - Ombretta Melaiu
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
| | - Daniela Nardozi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
| | - Valentina Angiolini
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (V.A.); (V.L.); (L.M.)
| | - Valeria Lucarini
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (V.A.); (V.L.); (L.M.)
| | - Bora Kërpi
- Department of Biomedicine, Catholic University ‘Our Lady of Good Counsel’, 1000 Tirana, Albania;
| | - Laura Masuelli
- Department of Experimental Medicine, University of Rome “Sapienza”, 00161 Rome, Italy; (V.A.); (V.L.); (L.M.)
| | - Loredana Cifaldi
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
| | - Roberto Bei
- Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy; (C.F.); (C.P.); (M.B.); (R.C.); (O.M.); (D.N.); (L.C.)
- Faculty of Medicine and Surgery, Catholic University ‘Our Lady of Good Counsel’, 1000 Tirana, Albania
| |
Collapse
|
24
|
Yan S, Gao Z, Ding J, Chen S, Wang Z, Jin W, Qu B, Zhang Y, Yang L, Guo D, Yin T, Yang Y, Zhang Y, Yang J. Nanocomposites based on nanoceria regulate the immune microenvironment for the treatment of polycystic ovary syndrome. J Nanobiotechnology 2023; 21:412. [PMID: 37936120 PMCID: PMC10631133 DOI: 10.1186/s12951-023-02182-w] [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: 08/22/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
The immune system is closely associated with the pathogenesis of polycystic ovary syndrome (PCOS). Macrophages are one of the important immune cell types in the ovarian proinflammatory microenvironment, and ameliorate the inflammatory status mainly through M2 phenotype polarization during PCOS. Current therapeutic approaches lack efficacy and immunomodulatory capacity, and a new therapeutic method is needed to prevent inflammation and alleviate PCOS. Here, octahedral nanoceria nanoparticles with powerful antioxidative ability were bonded to the anti-inflammatory drug resveratrol (CeO2@RSV), which demonstrates a crucial strategy that involves anti-inflammatory and antioxidative efficacy, thereby facilitating the proliferation of granulosa cells during PCOS. Notably, our nanoparticles were demonstrated to possess potent therapeutic efficacy via anti-inflammatory activities and effectively alleviated endocrine dysfunction, inflammation and ovarian injury in a dehydroepiandrosterone (DHEA)-induced PCOS mouse model. Collectively, this study revealed the tremendous potential of the newly developed nanoparticles in ameliorating the proinflammatory microenvironment and promoting the function of granulosa cells, representing the first attempt to treat PCOS by using CeO2@RSV nanoparticles and providing new insights in combating clinical PCOS.
Collapse
Affiliation(s)
- Sisi Yan
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Zhipeng Gao
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China
| | - Jinli Ding
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Suming Chen
- The Institute for Advanced Studies, Wuhan University, Wuhan, China
| | - Zehao Wang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Wenyi Jin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Bing Qu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yi Zhang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Lian Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China
| | - Duanying Guo
- Longgang District People's Hospital of Shenzhen, Shenzhen, China.
| | - Tailang Yin
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China.
| | - Yanbing Yang
- Key Laboratory of Biomedical Polymers of Ministry of Education, College of Chemistry and Molecular Sciences, Wuhan University, Wuhan, 430072, People's Republic of China.
| | - Yan Zhang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| | - Jing Yang
- Reproductive Medical Center, Renmin Hospital of Wuhan University and Hubei Clinic Research Center for Assisted Reproductive Technology and Embryonic Development, Wuhan, 430060, China.
| |
Collapse
|
25
|
Zhao C, Zhou X, Cao Z, Ye L, Cao Y, Pan J. Curcumin and analogues against head and neck cancer: From drug delivery to molecular mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:154986. [PMID: 37506572 DOI: 10.1016/j.phymed.2023.154986] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/05/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is one of the most life-threatening diseases which also causes economic burden worldwide. To overcome the limitations of traditional therapies, investigation into alternative adjuvant treatments is crucial. PURPOSE Curcumin, a turmeric-derived compound, demonstrates significant therapeutic potential in diverse diseases, including cancer. Furthermore, research focuses on curcumin analogues and novel drug delivery systems, offering approaches for improved efficacy. This review aims to provide a comprehensive overview of curcumin's current findings, emphasizing its mechanisms of anti-HNSCC effects and potential for clinical application. METHOD An electronic search of Web of Science, MEDLINE, and Embase was conducted to identify literature about the application of curcumin or analogues in HNSCC. Titles and abstracts were screened to identify potentially eligible studies. Full-text articles will be obtained and independently evaluated by two authors to make the decision of inclusion in the review. RESULTS Curcumin's clinical application is hindered by poor bioavailability, prompting the exploration of methods to enhance it, such as curcumin analogues and novel drug delivery systems. Curcumin could exhibit anti-cancer effects by targeting cancer cells and modulating the tumor microenvironment in HNSCC. Mechanisms of action include cell cycle arrest, apoptosis promotion, reactive oxygen species induction, endoplasmic reticulum stress, inhibition of epithelial-mesenchymal transition, attenuation of extracellular matrix degradation, and modulation of tumor metabolism in HNSCC cells. Curcumin also targets various components of the tumor microenvironment, including cancer-associated fibroblasts, innate and adaptive immunity, and lymphovascular niches. Furthermore, curcumin enhances the anti-cancer effects of other drugs as adjunctive therapy. Two clinical trials report its potential clinical applications in treating HNSCC. CONCLUSION Curcumin has demonstrated therapeutic potential in HNSCC through in vitro and in vivo studies. Its effectiveness is attributed to its ability to modulate cancer cells and interact with the intricate tumor microenvironment. The development of curcumin analogues and novel drug delivery systems has shown promise in improving its bioavailability, thereby expanding its clinical applications. Further research and exploration in this area hold great potential for harnessing the full therapeutic benefits of curcumin in HNSCC treatment.
Collapse
Affiliation(s)
- Chengzhi Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 1 Section 3rd, Renmin Nan Road, Chengdu 610041, PR China
| | - Xueer Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 1 Section 3rd, Renmin Nan Road, Chengdu 610041, PR China
| | - Zhiwei Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 1 Section 3rd, Renmin Nan Road, Chengdu 610041, PR China
| | - Li Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 1 Section 3rd, Renmin Nan Road, Chengdu 610041, PR China
| | - Yubin Cao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 1 Section 3rd, Renmin Nan Road, Chengdu 610041, PR China.
| | - Jian Pan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, No. 1 Section 3rd, Renmin Nan Road, Chengdu 610041, PR China.
| |
Collapse
|
26
|
Loos JA, Franco M, Chop M, Rodriguez Rodrigues C, Cumino AC. Resveratrol against Echinococcus sp.: Discrepancies between In Vitro and In Vivo Responses. Trop Med Infect Dis 2023; 8:460. [PMID: 37888588 PMCID: PMC10610609 DOI: 10.3390/tropicalmed8100460] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023] Open
Abstract
In an attempt to find new anti-echinococcal drugs, resveratrol (Rsv) effectiveness against the larval stages of Echinococcus granulosus and E. multilocularis was evaluated. The in vitro effect of Rsv on parasites was assessed via optical and electron microscopy, RT-qPCR and immunohistochemistry. In vivo efficacy was evaluated in murine models of cystic (CE) and alveolar echinococcosis (AE). The impact of infection and drug treatment on the mouse bone marrow hematopoietic stem cell (HSC) population and its differentiation into dendritic cells (BMDCs) was investigated via flow cytometry and RT-qPCR. In vitro treatment with Rsv reduced E. granulosus metacestode and protoscolex viability in a concentration-dependent manner, caused ultrastructural damage, increased autophagy gene transcription, and raised Eg-Atg8 expression while suppressing Eg-TOR. However, the intraperitoneal administration of Rsv was not only ineffective, but also promoted parasite development in mice with CE and AE. In the early infection model of AE treated with Rsv, an expansion of HSCs was observed followed by their differentiation towards BMCDs. The latter showed an anti-inflammatory phenotype and reduced LPS-stimulated activation compared to control BMDCs. We suggest that Rsv ineffectiveness could have been caused by the low intracystic concentration achieved in vivo and the drug's hormetic effect, with opposite anti-parasitic and immunomodulatory responses in different doses.
Collapse
Affiliation(s)
- Julia A. Loos
- Instituto de Investigaciones en Producción, Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, Mar del Plata 7600, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
| | - Micaela Franco
- Hospital Interzonal General de Agudos “Dr. Oscar E Alende”, Mar del Plata 7600, Argentina;
| | - Maia Chop
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, Mar del Plata 7600, Argentina
| | - Christian Rodriguez Rodrigues
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, Mar del Plata 7600, Argentina
| | - Andrea C. Cumino
- Instituto de Investigaciones en Producción, Sanidad y Ambiente (IIPROSAM), Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel Cero, Mar del Plata 7600, Argentina;
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mar del Plata 7600, Argentina; (M.C.); (C.R.R.)
- Departamento de Química, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata (UNMdP), Funes 3350, Nivel 2, Mar del Plata 7600, Argentina
| |
Collapse
|
27
|
Sarfraz M, Arafat M, Zaidi SHH, Eltaib L, Siddique MI, Kamal M, Ali A, Asdaq SMB, Khan A, Aaghaz S, Alshammari MS, Imran M. Resveratrol-Laden Nano-Systems in the Cancer Environment: Views and Reviews. Cancers (Basel) 2023; 15:4499. [PMID: 37760469 PMCID: PMC10526844 DOI: 10.3390/cancers15184499] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The genesis of cancer is a precisely organized process in which normal cells undergo genetic alterations that cause the cells to multiply abnormally, colonize, and metastasize to other organs such as the liver, lungs, colon, and brain. Potential drugs that could modify these carcinogenic pathways are the ones that will be used in clinical trials as anti-cancer drugs. Resveratrol (RES) is a polyphenolic natural antitoxin that has been utilized for the treatment of several diseases, owing to its ability to scavenge free radicals, control the expression and activity of antioxidant enzymes, and have effects on inflammation, cancer, aging, diabetes, and cardioprotection. Although RES has a variety of pharmacological uses and shows promising applications in natural medicine, its unpredictable pharmacokinetics compromise its therapeutic efficacy and prevent its use in clinical settings. RES has been encapsulated into various nanocarriers, such as liposomes, polymeric nanoparticles, lipidic nanocarriers, and inorganic nanoparticles, to address these issues. These nanocarriers can modulate drug release, increase bioavailability, and reach therapeutically relevant plasma concentrations. Studies on resveratrol-rich nano-formulations in various cancer types are compiled in the current article. Studies relating to enhanced drug stability, increased therapeutic potential in terms of pharmacokinetics and pharmacodynamics, and reduced toxicity to cells and tissues are the main topics of this research. To keep the readers informed about the current state of resveratrol nano-formulations from an industrial perspective, some recent and significant patent literature has also been provided. Here, the prospects for nano-formulations are briefly discussed, along with machine learning and pharmacometrics methods for resolving resveratrol's pharmacokinetic concerns.
Collapse
Affiliation(s)
- Muhammad Sarfraz
- College of Pharmacy, Al Ain University, Al Ain Campus, Al Ain P.O. Box 64141, United Arab Emirates
| | - Mosab Arafat
- College of Pharmacy, Al Ain University, Al Ain Campus, Al Ain P.O. Box 64141, United Arab Emirates
| | - Syeda Huma H. Zaidi
- Department of Chemistry, Faculty of Science, Northern Border University, Arar 91431, Saudi Arabia
| | - Lina Eltaib
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Muhammad Irfan Siddique
- Department of Pharmaceutics, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia;
| | - Abuzer Ali
- Department of Pharmacognosy, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | | | - Abida Khan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia (M.I.)
| | - Shams Aaghaz
- Department of Pharmacy, School of Medical & Allied Sciences, Galgotias University, Greater Noida 203201, India
| | - Mohammed Sanad Alshammari
- Department of Computer Science, Faculty of Computing and Information Technology, Northern Border University, Rafha 91911, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia (M.I.)
| |
Collapse
|
28
|
Patra S, Praharaj PP, Singh A, Bhutia SK. Targeting SIRT1-regulated autophagic cell death as a novel therapeutic avenue for cancer prevention. Drug Discov Today 2023; 28:103692. [PMID: 37379905 DOI: 10.1016/j.drudis.2023.103692] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/15/2023] [Accepted: 06/21/2023] [Indexed: 06/30/2023]
Abstract
Cellular localization and deacetylation activity of sirtuin 1 (SIRT1) has a significant role in cancer regulation. The multifactorial role of SIRT1 in autophagy regulates several cancer-associated cellular phenotypes, aiding cellular survival and cell death induction. SIRT1-mediated deacetylation of autophagy-related genes (ATGs) and associated signaling mediators control carcinogenesis. The hyperactivation of bulk autophagy, disrupted lysosomal and mitochondrial biogenesis, and excessive mitophagy are key mechanism for SIRT1-mediated autophagic cell death (ACD). In terms of the SIRT1-ACD nexus, identifying SIRT1-activating small molecules and understanding the possible mechanism triggering ACD could be a potential therapeutic avenue for cancer prevention. In this review, we provide an update on the structural and functional intricacy of SIRT1 and SIRT1-mediated autophagy activation as an alternative cell death modality for cancer prevention.
Collapse
Affiliation(s)
- Srimanta Patra
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha-769008, India
| | - Prakash P Praharaj
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha-769008, India
| | - Amruta Singh
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha-769008, India
| | - Sujit K Bhutia
- Cancer and Cell Death Laboratory, Department of Life Science, National Institute of Technology Rourkela, Odisha-769008, India.
| |
Collapse
|
29
|
Vieira IRS, Tessaro L, Lima AKO, Velloso IPS, Conte-Junior CA. Recent Progress in Nanotechnology Improving the Therapeutic Potential of Polyphenols for Cancer. Nutrients 2023; 15:3136. [PMID: 37513554 PMCID: PMC10384266 DOI: 10.3390/nu15143136] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/06/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Polyphenols derived from fruits, vegetables, and plants are bioactive compounds potentially beneficial to human health. Notably, compounds such as quercetin, curcumin, epigallocatechin-3-gallate (EGCG), and resveratrol have been highlighted as antiproliferative agents for cancer. Due to their low solubility and limited bioavailability, some alternative nanotechnologies have been applied to encapsulate these compounds, aiming to improve their efficacy against cancer. In this comprehensive review, we evaluate the main nanotechnology approaches to improve the therapeutic potential of polyphenols against cancer using in vitro studies and in vivo preclinical models, highlighting recent advancements in the field. It was found that polymeric nanomaterials, lipid-based nanomaterials, inorganic nanomaterials, and carbon-based nanomaterials are the most used classes of nanocarriers for encapsulating polyphenols. These delivery systems exhibit enhanced antitumor activity and pro-apoptotic effects, particularly against breast, lung, prostate, cervical, and colorectal cancer cells, surpassing the performance of free bioactive compounds. Preclinical trials in xenograft animal models have revealed decreased tumor growth after treatment with polyphenol-loaded delivery systems. Moreover, the interaction of polyphenol co-delivery systems and polyphenol-drug delivery systems is a promising approach to increase anticancer activity and decrease chemotherapy side effects. These innovative approaches hold significant implications for the advancement of clinical cancer research.
Collapse
Affiliation(s)
- Italo Rennan Sousa Vieira
- Analytical and Molecular Laboratorial Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, RJ, Brazil
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Bio-Chemistry, Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Graduate Program in Chemistry (PGQu), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Graduate Program in Food Science (PPGCAL), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| | - Leticia Tessaro
- Analytical and Molecular Laboratorial Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, RJ, Brazil
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Bio-Chemistry, Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Graduate Program in Chemistry (PGQu), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| | - Alan Kelbis Oliveira Lima
- Nanobiotechnology Laboratory, Institute of Biology (IB), Department of Genetics and Morphology, University of Brasilia, Brasilia 70910-900, DF, Brazil
| | - Isabela Portella Silva Velloso
- Analytical and Molecular Laboratorial Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, RJ, Brazil
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Bio-Chemistry, Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| | - Carlos Adam Conte-Junior
- Analytical and Molecular Laboratorial Center (CLAn), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Center for Food Analysis (NAL), Technological Development Support Laboratory (LADETEC), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-598, RJ, Brazil
- Laboratory of Advanced Analysis in Biochemistry and Molecular Biology (LAABBM), Department of Bio-Chemistry, Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Graduate Program in Chemistry (PGQu), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
- Graduate Program in Food Science (PPGCAL), Institute of Chemistry (IQ), Federal University of Rio de Janeiro (UFRJ), Cidade Universitária, Rio de Janeiro 21941-909, RJ, Brazil
| |
Collapse
|
30
|
Pradhan B, Bhuyan PP, Ki JS. Immunomodulatory, Antioxidant, Anticancer, and Pharmacokinetic Activity of Ulvan, a Seaweed-Derived Sulfated Polysaccharide: An Updated Comprehensive Review. Mar Drugs 2023; 21:md21050300. [PMID: 37233494 DOI: 10.3390/md21050300] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 05/27/2023] Open
Abstract
Cancer is one of the most worldwide spread diseases and causes maximum death. Treatment of cancer depends on the host immune system and the type of drugs. The inefficiency of conventional cancer treatments as a result of drug resistance, nontargeted delivery, and chemotherapy-related negative side effects has caused bioactive phytochemicals to come into focus. As a result, recent years have seen an increase in research into screening and identifying natural compounds with anticancer properties. Recent studies on the isolation and use of polysaccharides derived from various marine algal species have revealed a variety of biological activities, including antioxidant and anticancer properties. Ulvan is a polysaccharide derived from various green seaweeds of the Ulva species in the family Ulvaceae. It has been demonstrated to have potent anticancer and anti-inflammatory properties through the modulation of antioxidants. It is vital to understand the mechanisms underlying the biotherapeutic activities of Ulvan in cancer and its role in immunomodulation. In this context, we reviewed the anticancer effects of ulvan based on its apoptotic effects and immunomodulatory activity. Additionally, we also focused on its pharmacokinetic studies in this review. Ulvan is the most conceivable candidate for use as a cancer therapeutic agent and could be used to boost immunity. Moreover, it may be established as an anticancer drug once its mechanisms of action are understood. Due to its high food and nutritive values, it can be used as a possible dietary supplement for cancer patients in the near future. This review may provide fresh perspectives on the potential novel role of ulvan, reveal a brand-new cancer-prevention strategy, and improve human health.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Biotechnology, Sangmyung University, Seoul 03016, Republic of Korea
- School of Biological Sciences, AIPH University, Bhubaneswar 752101, Odisha, India
| | - Prajna Paramita Bhuyan
- Department of Botany, Maharaja Sriram Chandra Bhanja Deo University, Baripada 757003, Odisha, India
| | - Jang-Seu Ki
- Department of Biotechnology, Sangmyung University, Seoul 03016, Republic of Korea
| |
Collapse
|
31
|
Pradhan B, Ki JS. Antioxidant and chemotherapeutic efficacies of seaweed-derived phlorotannins in cancer treatment: A review regarding novel anticancer drugs. Phytother Res 2023; 37:2067-2091. [PMID: 36971337 DOI: 10.1002/ptr.7809] [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: 10/28/2022] [Revised: 02/17/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023]
Abstract
The ineffectiveness of traditional cancer therapies due to drug resistance, nontargeted delivery, and chemotherapy-associated adverse side effects has shifted attention to bioactive phytochemicals. Consequently, research efforts toward screening and identification of natural compounds with anticancer properties have increased in recent years. Marine seaweed-derived bioactive compounds, such as polyphenolic compounds, have exhibited anticancer properties. Phlorotannins (PTs), a major group of seaweed-derived polyphenolic compounds, have emerged as powerful chemopreventive and chemoprotective compounds, regulating apoptotic cell death pathways both in vitro and in vivo. In this context, this review focuses on the anticancer activity of polyphenols isolated from brown algae, with a special reference to PTs. Furthermore, we highlight the antioxidant effects of PTs and discuss how they can impact cell survival and tumor development and progression. Moreover, we discussed the potential therapeutic application of PTs as anticancer agents, having molecular mechanisms involving oxidative stress reduction. We have also discussed patents or patent applications that apply PTs as major components of antioxidant and antitumor products. With this review, researcher may gain new insights into the potential novel role of PTs, as well as uncover a novel cancer-prevention mechanism and improve human health.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Biotechnology, Sangmyung University, Seoul, 03016, South Korea
- School of Biological Sciences, AIPH University, Bhubaneswar, 752101, India
| | - Jang-Seu Ki
- Department of Biotechnology, Sangmyung University, Seoul, 03016, South Korea
| |
Collapse
|
32
|
Pradhan B, Ki JS. Biological activity of algal derived carrageenan: A comprehensive review in light of human health and disease. Int J Biol Macromol 2023; 238:124085. [PMID: 36948331 DOI: 10.1016/j.ijbiomac.2023.124085] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023]
Abstract
Carrageenans are a family of natural linear sulfated polysaccharides derived from red seaweeds and used as a common food additive. Carrageenan's properties, impact on health, and aesthetic benefits have all been studied for a long time; however, the mechanisms are still unclear. In pharmaceutical aspects, carrageenan displayed potential antioxidant and immunomodulatory properties in both in vivo and in vitro action. It also contributes to potential disease-preventive activities through dynamic modulation of important intracellular signaling pathways, regulation of ROS buildup, and preservation of major cell survival and death processes which leads to potential drug development. Furthermore, the chemical synthesis of the current bioactive medicine with confirmational rearrangement may increase availability and bioactivity needs diligent examination. In this review, we give an up-to-date overview of recent research on Carrageenan with reference to health and therapeutic advantages. In addition, we have focused on structural conformation and its primary strategic deployment in disease prevention, as well as the mechanistic investigation of how it functions to combat various disease-preventive employed for future therapeutic interventions. This review may get new insights into the possible novel role of carrageenan and open up a novel disease-preventive mechanism and enhance human health.
Collapse
Affiliation(s)
- Biswajita Pradhan
- Department of Biotechnology, Sangmyung University, Seoul 03016, Republic of Korea; School of Biological Sciences, AIPH University, Bhubaneswar 752101, Odisha, India
| | - Jang-Seu Ki
- Department of Biotechnology, Sangmyung University, Seoul 03016, Republic of Korea.
| |
Collapse
|
33
|
Han H, Alsayed AMM, Wang Y, Yan Q, Shen A, Zhang J, Ye Y, Liu Z, Wang K, Zheng X. Discovery of β-cyclocitral-derived mono-carbonyl curcumin analogs as anti-hepatocellular carcinoma agents via suppression of MAPK signaling pathway. Bioorg Chem 2023; 132:106358. [PMID: 36642021 DOI: 10.1016/j.bioorg.2023.106358] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/22/2022] [Accepted: 01/08/2023] [Indexed: 01/12/2023]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors with a high recurrence and mortality rate. In this study, a series of β-cyclocitral-derived mono-carbonyl curcumin analogs were synthesized and their anticancer properties were evaluated. Among the series, A19 exhibited the strongest cytotoxic activity by inhibiting cell viability and colony formation, inducing cell cycle G2/M phase arrest and cell apoptosis of HCC HepG2 and Huh-7 cells, while having almost no cytotoxicity on normal liver MIHA cells. Mechanistically, our results demonstrated that A19 triggered intense DNA damage via suppression of the ERK/JNK/p38 MAPK signaling pathway. Additionally, a combination of A19 with sorafenib significantly induced synergistic cytotoxicity in HCC cells. Overall, our results indicate the potential of A19 as a novel chemotherapeutic drug administered either separately or in combined therapy for HCC treatment.
Collapse
Affiliation(s)
- Haoyi Han
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Ali Mohammed Mohammed Alsayed
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yi Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Qi Yan
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Ancheng Shen
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Jianxia Zhang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Yanfei Ye
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China
| | - Zhiguo Liu
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| | - Kun Wang
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| | - Xiaohui Zheng
- Chemical Biology Research Center at School of Pharmaceutical Sciences, Wenzhou Medical University, 1210 University Town, Wenzhou, Zhejiang 325035, China.
| |
Collapse
|
34
|
Gahtori R, Tripathi AH, Kumari A, Negi N, Paliwal A, Tripathi P, Joshi P, Rai RC, Upadhyay SK. Anticancer plant-derivatives: deciphering their oncopreventive and therapeutic potential in molecular terms. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2023. [DOI: 10.1186/s43094-023-00465-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2023] Open
Abstract
Abstract
Background
Over the years, phytomedicines have been widely used as natural modalities for the treatment and prevention of various diseases by different ethnic groups across the globe. Although, 25% of drugs in the USA contain at least one plant-derived therapeutic compound, currently there is a paucity of plant-derived active medicinal ingredients in the pharmaceutical industry. Scientific evidence-based translation of plant-derived ethnomedicines for their clinical application is an urgent need. The anticancer and associated properties (antioxidative, anti-inflammatory, pro-apoptotic and epithelial-mesenchymal transition (EMT) inhibition) of various plant extracts and phytochemicals have been elucidated earlier. Several of the plant derivatives are already in use under prophylactic/therapeutic settings against cancer and many are being investigated under different phases of clinical trials.
Main body
The purpose of this study is to systematically comprehend the progress made in the area of prophylactic and therapeutic potential of the anticancerous plant derivatives. Besides, we aim to understand their anticancer potential in terms of specific sub-phenomena, such as anti-oxidative, anti-inflammatory, pro-apoptotic and inhibition of EMT, with an insight of the molecules/pathways associated with them. The study also provides details of classes of anticancer compounds, their plant source(s) and the molecular pathway(s) targeted by them. In addition to the antioxidative and antiproliferative potentials of anticancer plant derivatives, this study emphasizes on their EMT-inhibition potential and other ‘anticancer related’ properties. The EMT is highlighted as a phenomenon of choice for targeting cancer due to its role in the induction of metastasis and drug resistance. Different phytochemicals in pre-clinical or clinical trials, with promising chemopreventive/anticancer activities have been enlisted and the plant compounds showing synergistic anticancer activity in combination with the existing drugs have been discussed. The review also unravels the need of carrying out pan-signalome studies for identifying the cardinal pathways modulated by phytomedicine(s), as in many cases, the molecular pathway(s) has/have been randomly studied.
Conclusion
This review systematically compiles the studies regarding the impact of various plant derivatives in different cancers and oncogenic processes, as tested in diverse experimental model systems. Availability of more comprehensive information on anticancer phyto-constituents, their relative abundance in crude drugs, pathways/molecules targeted by phytomedicines, their long-term toxicity data and information regarding their safe use under the combinatorial settings, would open greater avenues of their utilization in future against this dreaded disease.
Graphical Abstract
Collapse
|
35
|
Sain A, Kandasamy T, Naskar D. Targeting UNC-51-like kinase 1 and 2 by lignans to modulate autophagy: possible implications in metastatic colorectal cancer. Mol Divers 2023; 27:27-43. [PMID: 35192112 DOI: 10.1007/s11030-022-10399-4] [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] [Received: 12/17/2021] [Accepted: 01/31/2022] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC), especially metastatic (mCRC) form, becomes a major reason behind cancer morbidity worldwide, whereas the treatment strategy is not optimum. Several novel targets are under investigation for mCRC including the autophagy pathway. Natural compounds including dietary lignans are sparsely reported as autophagy modulators. Nonetheless, the interaction between dietary lignans and core autophagy complexes are yet to be characterised. We aimed to describe the interaction between the dietary lignans from flaxseed (Linum usitatissimum) and sesame seeds (Sesamum indicum) along with the enterolignans (enterodiol and enterolactone) and the UNC-51-like kinase 1 and 2 (ULK1/2), important kinases required for the autophagy. A range of in-silico technologies viz. molecular docking, drug likeness, and ADME/T was employed to select the best fit modulator and/or inhibitor of the target kinases from the list of selected lignans. Drug likeness and ADME/T studied further selected the best-suited lignans as potential autophagy inhibitor. Molecular dynamic simulation (MDS) analyses were used to validate the molecular docking results. Binding free energies of the protein-ligand interactions by MM-PBSA method further confirmed best-selected lignans as ULK1 and/or ULK2 inhibitor. In conclusion, three dietary lignans pinoresinol, medioresinol, and lariciresinol successfully identified as dual ULK1/2 inhibitor/modifier, whereas enterodiol emerged as a selective ULK2 inhibitor/modifier.
Collapse
Affiliation(s)
- Arindam Sain
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, NH-12, Haringhata, Nadia, West Bengal, 741249, India
| | - Thirukumaran Kandasamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, India
| | - Debdut Naskar
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, NH-12, Haringhata, Nadia, West Bengal, 741249, India.
| |
Collapse
|
36
|
Chen Z, Wen T, Wang X, Yang L, Wang Z, Qin Y, Hu Y, Zhang T, Wang D, Liu A, Zhang L, Lei M, Zhu Y. Co-delivery of immunochemotherapeutic by classified targeting based on chitosan and cyclodextrin derivatives. Int J Biol Macromol 2023; 226:1396-1410. [PMID: 36442558 DOI: 10.1016/j.ijbiomac.2022.11.253] [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] [Received: 09/14/2022] [Revised: 11/17/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Herein, a cyclodextrin derivative (R6RGD-CMβCD) with tumor target and a carboxymethyl chitosan derivative (M2pep-CMCS) with tumor-associated macrophages 2 (TAM2) target were successfully synthesized, respectively. DOX-loaded nanoparticles (R6RGD-CMβCD@DOX NPs, RCNPDOX) and R848-loaded nanoparticles (M2pep-CMCS@R848 NPs, MCNPR848) were prepared. Furthermore, the RCNPDOX and MCNPR848 exhibited good DOX and R848 absorption. Meanwhile, the synergetic cell toxicity of RCNPDOX and MCNPR848 was found. Additionally, RCNPDOX + MCNPR848 nanoparticles greatly promoted the expression levels of cleaved Caspase3, which indicated that the nanoparticles could induce cell apoptosis. At the same time, the immunohistochemical images exhibited that RCNPDOX + MCNPR848 group could effectively transform the phenotype of tumor-associated macrophages. Importantly, in vivo experiments revealed that RCNPDOX + MCNPR848 NPs exerted excellent anticancer effects in tumor-bearing mice. To summarize, RCNPDOX + MCNPR848 NPs are effective anticancer treatment combining chemotherapy and immunotherapy, M2pep-CMCS and R6RGD-CMβCD are good delivery materials.
Collapse
Affiliation(s)
- Zhimeng Chen
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Tiantian Wen
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Xueyuan Wang
- College of Life Science, Nanjing Normal University, Nanjing 210023, China
| | - Lin Yang
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Zhongjie Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yanru Qin
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Yixue Hu
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Tianyu Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Dongna Wang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Amin Liu
- College of Science, Nanjing Forestry University, Nanjing 210037, China
| | - Liefeng Zhang
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| | - Meng Lei
- College of Science, Nanjing Forestry University, Nanjing 210037, China.
| | - Yongqiang Zhu
- School of Food and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China; College of Life Science, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
37
|
Hu H, Liao Z, Xu M, Wan S, Wu Y, Zou W, Wu J, Fan Q. Fabrication, Optimization, and Evaluation of Paclitaxel and Curcumin Coloaded PLGA Nanoparticles for Improved Antitumor Activity. ACS OMEGA 2023; 8:976-986. [PMID: 36643566 PMCID: PMC9835547 DOI: 10.1021/acsomega.2c06359] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
Codelivery of chemotherapeutic drugs in nanoparticles can enhance the therapeutic effects against tumors. However, their anticancer properties and physiochemical characteristics can be severely influenced by many formulation parameters during the preparation process. It is a complicated development phase to select the optimal parameters for preparation of nanoparticles based on the commonly used one single parameter method, which consumes a lot of money, time, and effort, and sometimes even fails. Therefore, the statistical analysis based on Box-Behnken design (BBD) has attracted much attention in bioengineering fields because it can illustrate the influence of parameters, build mathematical models, and predict the optimal combinational factors in a decreased number of experiments. In this study, we used a three-factor three-level BBD design to optimize the preparation of poly(lactic-co-glycolic acid) (PLGA) nanoparticles coloaded with two anticancer drugs curcumin and paclitaxel (PLGA-CUR-PTX nanoparticles). The surfactant concentration, polymer concentration, and oil-water ratio were selected as independent variables. An optimized model of the formulation for PLGA-CUR-PTX nanoparticles was validated. The optimal nanoparticles possessed a uniform spherical shape, with an average size of 99.94 nm, and the drug encapsulation efficiencies of CUR and PTX were 63.53 and 80.64%, respectively. The drug release from nanoparticles showed a biphasic release behavior, with a release mechanism via diffusion and fundamentally quasi-Fickian diffusion. The optimized nanoparticles demonstrated an enhanced cytotoxicity effect with lower IC50 values to 4T1 and MCF-7 breast cancer cell lines compared to free drugs. In summary, BBD optimization of CUR and PTX coloaded nanoparticles yielded a favorable drug carrier that holds potential as an alternative treatment for anticancer therapy.
Collapse
Affiliation(s)
- Haiyang Hu
- Department
of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan610075, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| | - Zuyue Liao
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
| | - Mengyao Xu
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
| | - Shengli Wan
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| | - Yuesong Wu
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| | - Wenjun Zou
- Department
of Chinese Materia Medica, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan610075, China
| | - Jianming Wu
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
- School
of Basic Medical Sciences, Southwest Medical
University, Luzhou646000, China
| | - Qingze Fan
- Department
of Pharmacy, The Affiliated Hospital of
Southwest Medical University, Luzhou, Sichuan646099, China
- Sichuan
Key Medical Laboratory of New Drug Discovery and Druggability Evaluation,
Luzhou Key Laboratory of Activity Screening and Druggability Evaluation
for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou646000, China
| |
Collapse
|
38
|
Asam Raza M, Farwa U, Waseem Mumtaz M, Kainat J, Sabir A, Al-Sehemi AG. Green synthesis of gold and silver nanoparticles as antidiabetic and anticancerous agents. GREEN CHEMISTRY LETTERS AND REVIEWS 2023; 16. [DOI: 10.1080/17518253.2023.2275666] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 10/21/2023] [Indexed: 01/05/2025]
Affiliation(s)
| | - Umme Farwa
- Department of Chemistry, University of Gujrat, Gujrat, Pakistan
| | | | - Javeria Kainat
- Department of Chemistry, University of Gujrat, Gujrat, Pakistan
| | - Areej Sabir
- Department of Chemistry, University of Gujrat, Gujrat, Pakistan
| | | |
Collapse
|
39
|
Bhatiya M, Pathak S, Jothimani G, Duttaroy AK, Banerjee A. A Comprehensive Study on the Anti-cancer Effects of Quercetin and Its Epigenetic Modifications in Arresting Progression of Colon Cancer Cell Proliferation. Arch Immunol Ther Exp (Warsz) 2023; 71:6. [PMID: 36807774 PMCID: PMC9941246 DOI: 10.1007/s00005-023-00669-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 12/02/2022] [Indexed: 02/23/2023]
Abstract
Colon cancer etiology involves a wide spectrum of genetic and epigenetic alterations, finding it challenging to find effective therapeutic strategies. Quercetin exhibits potent anti-proliferative/apoptotic properties. In the present study, we aimed to elucidate the anti-cancer and anti-aging effect of quercetin in colon cancer cell lines. The anti-proliferative effect of quercetin was assessed in vitro by CCK-8 in normal and colon cancer cell lines. To check the anti-aging potential of quercetin, collagenase, elastase, and hyaluronidase inhibitory activity assays were performed. The epigenetic and DNA damage assays were performed using the human NAD-dependent deacetylase Sirtuin-6, proteasome 20S, Klotho, Cytochrome-C, and telomerase ELISA kits. Furthermore, the aging-associated miRNA expression profiling was performed on colon cancer cells. The treatment with quercetin inhibited cell proliferation of colon cancer cells in a dose-dependent manner. Quercetin arrested colon cancer cell growth by modulating expression of aging proteins including Sirtuin-6 and Klotho and also by inhibiting telomerase activity to restrict the telomere length which is evident from qPCR analysis. Quercetin also exhibited DNA damage protection by reducing proteasome 20S levels. The miRNA expression profiling results displayed differential expression of miRNA in colon cancer cell, and in addition, the highly upregulated miRNA was involved in the regulation of cell cycle, proliferation, and transcription. Our data suggest that quercetin treatment inhibited cell proliferation in colon cancer cells through regulating the anti-aging protein expression and provides better understanding for quercetin's potential use in colon cancer treatment.
Collapse
Affiliation(s)
- Meenu Bhatiya
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603 103 India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603 103 India
| | - Ganesan Jothimani
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu 603 103 India
| | - Asim K. Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Tamil Nadu, 603 103, India.
| |
Collapse
|
40
|
Han Z, Song B, Yang J, Wang B, Ma Z, Yu L, Li Y, Xu H, Qiao M. Curcumin-Encapsulated Fusion Protein-Based Nanocarrier Demonstrated Highly Efficient Epidermal Growth Factor Receptor-Targeted Treatment of Colorectal Cancer. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:15464-15473. [PMID: 36454954 DOI: 10.1021/acs.jafc.2c04668] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Curcumin, a polyphenol derived from turmeric, has multiple biological functions, such as anti-inflammatory, antioxidant, antibacterial and, above all, antitumor activity. Colorectal cancer is a common malignancy of the gastrointestinal tract with an extremely high mortality rate. However, the low bioavailability and poor targeting properties of curcumin generally limit its clinical application. In the present study, we designed a fusion protein GE11-HGFI as a nanodrug delivery system. The protein was connected by flexible linkers, inheriting the self-assembly properties of hydrophobin HGFI and the targeting ability of GE11. The data show that the encapsulation of curcumin by fusion protein GE11-HGFI can form uniform and stable nanoparticles with a size of only 80 nm. In addition, the nanocarrier had high encapsulation efficiency for curcumin and made it to release sustainably. Notably, the drug-loaded nanosystem selectively targeted colorectal cancer cells with high epidermal growth factor receptor expression, resulting in high aggregated concentrations of curcumin at tumor sites, thus showing a significant anticancer effect. These results suggest that the nanocarrier fusion protein has the potential to be a novel strategy for enhancing molecular bioactivity and drug targeting in cancer therapy.
Collapse
Affiliation(s)
- Zhiqiang Han
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Bo Song
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Jiyuan Yang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Bo Wang
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Zhongqiang Ma
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Long Yu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Yuanhao Li
- Remegen Co., Ltd., Shandong 264000, P.R. China
| | - Haijin Xu
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
| | - Mingqiang Qiao
- The Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, P.R. China
- School of Life Science, Shanxi University, Shanxi 030000, P.R. China
| |
Collapse
|
41
|
Exploiting Polyphenol-Mediated Redox Reorientation in Cancer Therapy. Pharmaceuticals (Basel) 2022; 15:ph15121540. [PMID: 36558995 PMCID: PMC9787032 DOI: 10.3390/ph15121540] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Polyphenol, one of the major components that exert the therapeutic effect of Chinese herbal medicine (CHM), comprises several categories, including flavonoids, phenolic acids, lignans and stilbenes, and has long been studied in oncology due to its significant efficacy against cancers in vitro and in vivo. Recent evidence has linked this antitumor activity to the role of polyphenols in the modulation of redox homeostasis (e.g., pro/antioxidative effect) in cancer cells. Dysregulation of redox homeostasis could lead to the overproduction of reactive oxygen species (ROS), resulting in oxidative stress, which is essential for many aspects of tumors, such as tumorigenesis, progression, and drug resistance. Thus, investigating the ROS-mediated anticancer properties of polyphenols is beneficial for the discovery and development of novel pharmacologic agents. In this review, we summarized these extensively studied polyphenols and discussed the regulatory mechanisms related to the modulation of redox homeostasis that are involved in their antitumor property. In addition, we discussed novel technologies and strategies that could promote the development of CHM-derived polyphenols to improve their versatile anticancer properties, including the development of novel delivery systems, chemical modification, and combination with other agents.
Collapse
|
42
|
Li C, Zhu P, Xiang H, Jin Y, Lu B, Shen Y, Wang W, Huang B, Chen Y. 3D-CEUS tracking of injectable chemo-sonodynamic therapy-enabled mop-up of residual renal cell carcinoma after thermal ablation. Mater Today Bio 2022; 18:100513. [PMID: 36569591 PMCID: PMC9771734 DOI: 10.1016/j.mtbio.2022.100513] [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: 10/13/2022] [Revised: 12/01/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022]
Abstract
Thermal ablation (TA), as a minimally invasive therapeutic technique, has been extensively used to the treatment of solid tumors, such as renal cell carcinoma (RCC), which, unfortunately, still fails to overcome the high risk of local recurrence and distant metastasis since the incomplete ablation cannot be ignored due to various factors such as the indistinguishable tumor margins and limited ablation zone. Herein, we report the injectable thermosensitive hydrogel by confining curcumin (Cur)-loaded hollow mesoporous organosilica nanoparticles (Cur@HMON@gel) which can locate in tumor site more than half a month and mop up the residual RCC under ultrasound (US) irradiation after transforming from colloidal sol status to elastic gel matrix at physiological temperature. Based on the US-triggered accelerated diffusion of the model chemotherapy drug with multi-pharmacologic functions, the sustained and controlled release of Cur has been demonstrated in vitro. Significantly, US is employed as an external energy to trigger Cur, as a sonosensitizer also, to generate reactive oxygen species (ROS) for sonodynamic tumor therapy (SDT) in parallel. Tracking by the three-dimensional contrast-enhanced ultrasound (3D-CEUS) imaging, the typical decreased blood perfusions have been observed since the residual xenograft tumor after incomplete TA were effectively suppressed during the chemo-sonodynamic therapy process. The high in vivo biocompatibility and biodegradability of the multifunctional nanoplatform confined by thermogel provide the potential of their further clinical translation for the solid tumor eradication under the guidance and monitoring of 3D-CEUS.
Collapse
Affiliation(s)
- Cuixian Li
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Piao Zhu
- Shanghai Tenth People's Hospital, Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai 200331, PR China,Corresponding author.
| | - Huijing Xiang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Yunjie Jin
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Beilei Lu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Yujia Shen
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China,Corresponding author. Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| | - Beijian Huang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China,Shanghai Institute of Medical Imaging, Fudan University, Shanghai 200032, PR China,Corresponding author. Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai 200032, PR China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China,Corresponding author.
| |
Collapse
|
43
|
Immunomodulatory and anti-inflammatory and anticancer activities of porphyran, a sulfated galactan. Carbohydr Polym 2022; 301:120326. [DOI: 10.1016/j.carbpol.2022.120326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/13/2022]
|
44
|
Jit BP, Pattnaik S, Arya R, Dash R, Sahoo SS, Pradhan B, Bhuyan PP, Behera PK, Jena M, Sharma A, Agrawala PK, Behera RK. Phytochemicals: A potential next generation agent for radioprotection. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 106:154188. [PMID: 36029645 DOI: 10.1016/j.phymed.2022.154188] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/13/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Radiation hazards are accountable for extensive damage in the biological system and acts as a public health burden. Owing to the rapid increasing in radiation technology, both Ionizing radiation (IR) from natural and man made source poses detrimental outcome to public health. IR releases free radicals which induces oxidative stress and deleterious biological damage by modulating radiation induced signalling intermediates. The efficacy of existing therapeutic approach and treatment strategy are limited owing to their toxicity and associated side effects. Indian system of traditional medicine is enriched with prospective phytochemicals with potential radioprotection ability. PURPOSE The present review elucidated and summarized the potential role of plant derived novel chemical compound with prospective radioprotective potential. METHOD So far as the traditional system of Indian medicine is concerned, plant kingdom is enriched with potential bioactive molecules with diverse pharmacological activities. We reviewed several compounds mostly secondary metabolites from plant origin using various search engines. RESULTS Both compounds from land plants and marine source exhibited antioxidant antiinflammatory, free radical scavenging ability. These compounds have tremendous potential in fine-tuning of several signalling intermediates, which are actively participated in the progression and development of a pathological condition associated with radiation stress. CONCLUSION Development and explore of an operational radioprotective agent from originated from plant source that can be used as a novel molecular tool to eliminate the widespread damage caused by space exploration, ionizing radiation, nuclear war and radiotherapy has been significantly appreciated. Through extensive literature search we highlighted several compounds from both land plant and marine origin can be implemented for a better therapeutic potential against radiation induced injury. Furthermore, extensive clinical trials must be carried out in near future for better therapeutic modality and clinical efficacy.
Collapse
Affiliation(s)
- Bimal Prasad Jit
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India; School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India
| | - Subhaswaraj Pattnaik
- Department of Microbiology, School of Life Sciences, Pondicherry University, Puducherry 605014, India; Centre of Excellence in Natural Products and Therapeutics, Department of Biotechnology and Bioinformatics, Sambalpur University, Jyoti Vihar, Burla, Odisha 768019, India
| | - Rakesh Arya
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India; School of Life Sciences, Sambalpur University, Jyoti Vihar, Burla 768019, India
| | - Rutumbara Dash
- Departement of Gastroenterology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | | | - Biswajita Pradhan
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India; Department of Biotechnology, Sangmyung University, Seoul 03016, South Korea
| | - Prajna Paramita Bhuyan
- Department of Botany, Maharaja Sriram Chandra Bhanja Deo University, Baripada, Odisha 757003, India
| | - Pradyota Kumar Behera
- Department of Chemistry, Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India
| | - Mrutyunjay Jena
- Algal Biotechnology and Molecular Systematic Laboratory, Post Graduate Department of Botany, Berhampur University, Bhanja Bihar, Berhampur, Odisha 760007, India
| | - Ashok Sharma
- Department of Biochemistry, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| | - Paban Kumar Agrawala
- Institute of Nuclear Medicine and Allied Science, Defence Research and Development Organization, New Delhi 110054, India
| | | |
Collapse
|
45
|
The Utilization of Physiologically Active Molecular Components of Grape Seeds and Grape Marc. Int J Mol Sci 2022; 23:ijms231911165. [PMID: 36232467 PMCID: PMC9570270 DOI: 10.3390/ijms231911165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/01/2022] [Accepted: 09/16/2022] [Indexed: 11/18/2022] Open
Abstract
Nutritional interventions may highly contribute to the maintenance or restoration of human health. Grapes (Vitis vinifera) are one of the oldest known beneficial nutritional components of the human diet. Their high polyphenol content has been proven to enhance human health beyond doubt in statistics-based public health studies, especially in the prevention of cardiovascular disease and cancer. The current review concentrates on presenting and classifying polyphenol bioactive molecules (resveratrol, quercetin, catechin/epicatechin, etc.) available in high quantities in Vitis vinifera grapes or their byproducts. The molecular pathways and cellular signaling cascades involved in the effects of these polyphenol molecules are also presented in this review, which summarizes currently available in vitro and in vivo experimental literature data on their biological activities mostly in easily accessible tabular form. New molecules for different therapeutic purposes can also be synthesized based on existing polyphenol compound classes available in high quantities in grape, wine, and grape marc. Therefore an overview of these molecular structures is provided. Novel possibilities as dendrimer nanobioconjugates are reviewed, too. Currently available in vitro and in vivo experimental literature data on polyphenol biological activities are presented in easily accessible tabular form. The scope of the review details the antidiabetic, anticarcinogenic, antiviral, vasoprotective, and neuroprotective roles of grape-origin flavonoids. The novelty of the study lies in the description of the processing of agricultural by-products (grape seeds and skins) of industrial relevance, and the detailed description of the molecular mechanisms of action. In addition, the review of the clinical therapeutic applications of polyphenols is unique as no summary study has yet been done.
Collapse
|
46
|
Sufianova G, Gareev I, Beylerli O, Wu J, Shumadalova A, Sufianov A, Chen X, Zhao S. Modern aspects of the use of natural polyphenols in tumor prevention and therapy. Front Cell Dev Biol 2022; 10:1011435. [PMID: 36172282 PMCID: PMC9512088 DOI: 10.3389/fcell.2022.1011435] [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: 08/04/2022] [Accepted: 08/26/2022] [Indexed: 11/13/2022] Open
Abstract
Polyphenols are secondary plant metabolites or organic compounds synthesized by them. In other words, these are molecules that are found in plants. Due to the wide variety of polyphenols and the plants in which they are found, these compounds are divided according to the source of origin, the function of the polyphenols, and their chemical structure; where the main ones are flavonoids. All the beneficial properties of polyphenols have not yet been studied, since this group of substances is very extensive and diverse. However, most polyphenols are known to be powerful antioxidants and have anti-inflammatory effects. Polyphenols help fight cell damage caused by free radicals and immune system components. In particular, polyphenols are credited with a preventive effect that helps protect the body from certain forms of cancer. The onset and progression of tumors may be related directly to oxidative stress, or inflammation. These processes can increase the amount of DNA damage and lead to loss of control over cell division. A number of studies have shown that oxidative stress uncontrolled by antioxidants or an uncontrolled and prolonged inflammatory process increases the risk of developing sarcoma, melanoma, and breast, lung, liver, and prostate cancer. Therefore, a more in-depth study of the effect of polyphenolic compounds on certain signaling pathways that determine the complex cascade of oncogenesis is a promising direction in the search for new methods for the prevention and treatment of tumors.
Collapse
Affiliation(s)
- Galina Sufianova
- Department of Pharmacology, Tyumen State Medical University, Tyumen, Russia
| | - Ilgiz Gareev
- Educational and Scientific Institute of Neurosurgery, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Ozal Beylerli
- Educational and Scientific Institute of Neurosurgery, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
| | - Jianing Wu
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, China
| | - Alina Shumadalova
- Department of General Chemistry, Bashkir State Medical University, Ufa, Russia
| | - Albert Sufianov
- Educational and Scientific Institute of Neurosurgery, Peoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
- *Correspondence: Albert Sufianov, ; Xin Chen, ; Shiguang Zhao,
| | - Xin Chen
- Department of Neurosurgical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Albert Sufianov, ; Xin Chen, ; Shiguang Zhao,
| | - Shiguang Zhao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen, China
- Department of Neurosurgical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin, China
- *Correspondence: Albert Sufianov, ; Xin Chen, ; Shiguang Zhao,
| |
Collapse
|
47
|
Peri S, Ruzzolini J, Urciuoli S, Versienti G, Biagioni A, Andreucci E, Peppicelli S, Bianchini F, Bottari A, Calorini L, Nediani C, Magnelli L, Papucci L. An Oleocanthal-Enriched EVO Oil Extract Induces the ROS Production in Gastric Cancer Cells and Potentiates the Effect of Chemotherapy. Antioxidants (Basel) 2022; 11:antiox11091762. [PMID: 36139836 PMCID: PMC9495378 DOI: 10.3390/antiox11091762] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Oleocanthal, a minor polar compound in extra-virgin olive (EVO) oil, contains anticancer properties, which should be encouraged in its use in oncology. Gastric Cancer (GC), a very aggressive human cancer, is often diagnosed at advanced stages, when surgery is substituted or supported by chemotherapy (CT). However, CT frequently fails due to the patient’s resistance to the treatment. Thus, the aim of this study is to verify whether an OC-enriched EVO oil extract fraction (OCF) may be useful in order to overcome a resistance to GC. We evaluated the OCF effects on an AGS gastric adenocarcinoma cell line wild type (AGS wt) and on its subpopulations resistant to 5-fluorouracil (5FUr), Paclitaxel (TAXr) or cisplatin (CISr). We found that a 60 µM dose of the OCF acts on the AGS wt, 5FUr and TAXr, leading to the cell cycle inhibition and to a ROS production, but not on CISr cells. Resistance of CISr to the OCF seems to be due to higher levels of antioxidant-enzymes that can counteract the OCF-induced ROS production. Moreover, using the OCF plus 5-fluorouracil, Paclitaxel or cisplatin, we found a potentiating effect compared with a mono-treatment in all resistant GC cells, including CISr. In conclusion, the use of the OCF in the management of GC has shown very interesting advantages, opening-up the possibility to evaluate the efficacy of the OCF in vivo, as a valid adjuvant in the treatment of resistant GC.
Collapse
Affiliation(s)
- Sara Peri
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Jessica Ruzzolini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Silvia Urciuoli
- PHYTOLAB (Pharmaceutical, Cosmetic, Food Supplement, Technology and Analysis)-DiSIA, University of Florence, Via U. Schiff, 6, 50019 Florence, Italy
| | - Giampaolo Versienti
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Alessio Biagioni
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Andrea Bottari
- Digestive Surgery Unit, AOU Careggi University Hospital, 50134 Florence, Italy
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
- Center of Excellence for Research, Transfer, and High Education (DENOTHE), University of Florence, 50134 Florence, Italy
| | - Chiara Nediani
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
- Correspondence: ; Tel.: +39-0552751203
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, 50134 Florence, Italy
| |
Collapse
|
48
|
Combination Therapy Using Polyphenols: An Efficient Way to Improve Antitumoral Activity and Reduce Resistance. Int J Mol Sci 2022; 23:ijms231810244. [PMID: 36142147 PMCID: PMC9499610 DOI: 10.3390/ijms231810244] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
Polyphenols represent a structural class of mainly natural organic chemicals that contain multiple phenol structural units. The beneficial properties of polyphenols have been extensively studied for their antitumor, anti-inflammatory, and antibacterial effects, but nowadays, their medical applications are starting to be extended to many other applications due to their prebiotic role and their impact on the microbiota. This review focused on the use of polyphenols in cancer treatment. Their antineoplastic effects have been demonstrated in various studies when they were tested on numerous cancer lines and some in in vivo models. A431 and SCC13 human skin cancer cell lines treated with EGCG presented a reduced cell viability and enhanced cell death due to the inactivation of β-catenin signaling. Additionally, resveratrol showed a great potential against breast cancer mainly due to its ability to exert both anti-estrogenic and estrogenic effects (based on the concentration) and because it has a high affinity for estrogen receptors ERα and Erβ. Polyphenols can be combined with different classical cytostatic agents to enhance their therapeutic effects on cancer cells and to also protect healthy cells from the aggressiveness of antitumor drugs due to their anti-inflammatory properties. For instance, curcumin has been reported to reduce the gastrointestinal toxicity associated with chemotherapy. In the case of 5-FU-induced, it reduced the gastrointestinal toxicity by increasing the intestinal permeability and inhibiting mucosal damage. Co-administration of EGCG and doxorubicin induced the death of liver cancer cells. EGCG has the ability to inhibit autophagic activity and stop hepatoma Hep3B cell proliferation This symbiotic approach is well-known in medical practice including in multiple chemotherapy.
Collapse
|
49
|
A state-of-the-art review on fucoidan as an antiviral agent to combat viral infections. Carbohydr Polym 2022; 291:119551. [PMID: 35698330 PMCID: PMC9057937 DOI: 10.1016/j.carbpol.2022.119551] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 04/13/2022] [Accepted: 04/26/2022] [Indexed: 01/17/2023]
Abstract
As a significant public health hazard with several drug side effects during medical treatment, searching for novel therapeutic natural medicines is promising. Sulfated polysaccharides from algae, such as fucoidan, have been discovered to have a variety of medical applications, including antibacterial and immunomodulatory properties. The review emphasized on the utilization of fucoidan as an antiviral agent against viral infections by inhibiting their attachment and replication. Moreover, it can also trigger immune response against viral infection in humans. This review suggested to be use the fucoidan for the potential protective remedy against COVID-19 and addressing the antiviral activities of sulfated polysaccharide, fucoidan derived from marine algae that could be used as an anti-COVID19 drug in near future.
Collapse
|
50
|
Peng F, Liao M, Qin R, Zhu S, Peng C, Fu L, Chen Y, Han B. Regulated cell death (RCD) in cancer: key pathways and targeted therapies. Signal Transduct Target Ther 2022; 7:286. [PMID: 35963853 PMCID: PMC9376115 DOI: 10.1038/s41392-022-01110-y] [Citation(s) in RCA: 376] [Impact Index Per Article: 125.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/04/2022] [Accepted: 07/05/2022] [Indexed: 02/07/2023] Open
Abstract
Regulated cell death (RCD), also well-known as programmed cell death (PCD), refers to the form of cell death that can be regulated by a variety of biomacromolecules, which is distinctive from accidental cell death (ACD). Accumulating evidence has revealed that RCD subroutines are the key features of tumorigenesis, which may ultimately lead to the establishment of different potential therapeutic strategies. Hitherto, targeting the subroutines of RCD with pharmacological small-molecule compounds has been emerging as a promising therapeutic avenue, which has rapidly progressed in many types of human cancers. Thus, in this review, we focus on summarizing not only the key apoptotic and autophagy-dependent cell death signaling pathways, but the crucial pathways of other RCD subroutines, including necroptosis, pyroptosis, ferroptosis, parthanatos, entosis, NETosis and lysosome-dependent cell death (LCD) in cancer. Moreover, we further discuss the current situation of several small-molecule compounds targeting the different RCD subroutines to improve cancer treatment, such as single-target, dual or multiple-target small-molecule compounds, drug combinations, and some new emerging therapeutic strategies that would together shed new light on future directions to attack cancer cell vulnerabilities with small-molecule drugs targeting RCD for therapeutic purposes.
Collapse
Affiliation(s)
- Fu Peng
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Minru Liao
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rui Qin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Shiou Zhu
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Leilei Fu
- Sichuan Engineering Research Center for Biomimetic Synthesis of Natural Drugs, School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, China.
| | - Yi Chen
- West China School of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Bo Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Hospital of Chengdu University of Traditional Chinese Medicine, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|