1
|
Chen L, Cai B, Ni X, Lin Q, Ke R, Wan X, Huang T, Shan X, Wang B. Temozolomide monotherapy versus combination therapies in melanoma: a meta-analysis of efficacy and safety. Melanoma Res 2025; 35:87-101. [PMID: 39874124 DOI: 10.1097/cmr.0000000000001021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
Temozolomide is used in melanoma therapy, but the comparative efficacy and safety of monotherapy vs combination therapies are unclear. This meta-analysis evaluates temozolomide monotherapy vs combination therapies in melanoma patients. PubMed, Embase, and Cochrane Library were searched up to August 2024 for studies comparing temozolomide monotherapy with combination therapies in melanoma. Primary outcomes were 1-year survival and objective response rates (RR); secondary outcomes included hematologic and non-hematologic toxicities. Data were pooled using risk ratios with 95% confidence intervals (CIs). Seven studies were included. Combination therapies improved objective RR over temozolomide monotherapy (risk ratio 0.68, 95% CI: 0.53-0.88). One-year survival did not differ significantly between groups (risk ratio 0.81, 95% CI: 0.59-1.12). Temozolomide monotherapy was associated with reduced incidence of leukopenia (risk ratio 0.54, 95% CI: 0.30-0.95). Adding interferon-alpha (IFN-α) to temozolomide significantly improved 1-year survival (risk ratio 0.54, 95% CI: 0.35-0.84) and objective RR (risk ratio 0.57, 95% CI: 0.42-0.78) compared to temozolomide alone, without significantly increasing toxicity. Combination therapies enhance objective RR over temozolomide monotherapy, with similar 1-year survival. Temozolomide monotherapy offers a better hematologic safety profile. Combining temozolomide with IFN-α significantly improves survival and RR without increasing toxicity. Clinicians should balance efficacy and safety when choosing melanoma treatments.
Collapse
Affiliation(s)
- Lu Chen
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Beichen Cai
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xuejun Ni
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qian Lin
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ruonan Ke
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiaofen Wan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Tao Huang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Xiuying Shan
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Biao Wang
- Department of Plastic Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Plastic Surgery, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
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
|
3
|
Nowacka A, Śniegocka M, Smuczyński W, Liss S, Ziółkowska E, Bożiłow D, Śniegocki M, Wiciński M. The Potential Application of Resveratrol and Its Derivatives in Central Nervous System Tumors. Int J Mol Sci 2024; 25:13338. [PMID: 39769099 PMCID: PMC11728356 DOI: 10.3390/ijms252413338] [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: 11/23/2024] [Revised: 12/07/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Resveratrol, a naturally occurring polyphenolic compound found in various plants, has been extensively studied for its broad spectrum of beneficial biological effects. These encompass its potent antioxidant properties, anti-inflammatory activities, anti-aging capabilities, cardioprotective functions, and neuroprotective potential. The diverse biological actions of resveratrol extend beyond these well-established properties. It also exerts a significant impact on metabolic processes and bioavailability, and critically, it demonstrates the ability to effectively traverse the blood-brain barrier. This capacity to penetrate the central nervous system renders resveratrol a promising therapeutic agent for the management of central nervous system malignancies, as it has been shown to inhibit tumor cell proliferation, induce apoptosis, and modulate key signaling cascades, such as PI3K/Akt, JAK/STAT, and NF-kB. The multifaceted nature of resveratrol's biological effects, including its influence on diverse physiological processes, underscores its potential as a valuable therapeutic option for the treatment of central nervous system tumors.
Collapse
Affiliation(s)
- Agnieszka Nowacka
- Department of Neurosurgery, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Martyna Śniegocka
- Department of Anatomical, Histological, Forensic & Orthopedic Sciences, Section of Histology & Medical Embryology, Sapienza University of Rome, Via A. Scarpa, 14-16, 00161 Rome, Italy
| | - Wojciech Smuczyński
- Department of Physiotherapy, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Techników 3, 85-801 Bydgoszcz, Poland
| | - Sara Liss
- Department of Pharmacology and Therapeutics, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-090 Bydgoszcz, Poland
| | - Ewa Ziółkowska
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Dominika Bożiłow
- Anaesthesiology and Intensive Care Clinical Ward, The 10th Military Research Hospital and Polyclinic, ul. Powstańców Warszawy 5, 85-681 Bydgoszcz, Poland
| | - Maciej Śniegocki
- Department of Neurosurgery, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-094 Bydgoszcz, Poland
| | - Michał Wiciński
- Department of Pharmacology and Therapeutics, Nicolas Copernicus University in Toruń, Collegium Medicum in Bydgoszcz, ul. Curie Skłodowskiej 9, 85-090 Bydgoszcz, Poland
| |
Collapse
|
4
|
Sheida A, Farshadi M, Mirzaei A, Najjar Khalilabad S, Zarepour F, Taghavi SP, Hosseini Khabr MS, Ravaei F, Rafiei S, Mosadeghi K, Yazdani MS, Fakhraie A, Ghattan A, Zamani Fard MM, Shahyan M, Rafiei M, Rahimian N, Talaei Zavareh SA, Mirzaei H. Potential of Natural Products in the Treatment of Glioma: Focus on Molecular Mechanisms. Cell Biochem Biophys 2024; 82:3157-3208. [PMID: 39150676 DOI: 10.1007/s12013-024-01447-x] [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] [Accepted: 07/17/2024] [Indexed: 08/17/2024]
Abstract
Despite the waning of traditional treatments for glioma due to possible long-term issues, the healing possibilities of substances derived from nature have been reignited in the scientific community. These natural substances, commonly found in fruits and vegetables, are considered potential alternatives to pharmaceuticals, as they have been shown in prior research to impact pathways surrounding cancer progression, metastases, invasion, and resistance. This review will explore the supposed molecular mechanisms of different natural components, such as berberine, curcumin, coffee, resveratrol, epigallocatechin-3-gallate, quercetin, tanshinone, silymarin, coumarin, and lycopene, concerning glioma treatment. While the benefits of a balanced diet containing these compounds are widely recognized, there is considerable scope for investigating the efficacy of these natural products in treating glioma.
Collapse
Affiliation(s)
- Amirhossein Sheida
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | | | - Amirhossein Mirzaei
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shakiba Najjar Khalilabad
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Fatemeh Zarepour
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Pouya Taghavi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Sadat Hosseini Khabr
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Ravaei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Sara Rafiei
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Kimia Mosadeghi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Sepehr Yazdani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Ali Fakhraie
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Ghattan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Masoud Zamani Fard
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Maryam Shahyan
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Moein Rafiei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Neda Rahimian
- Endocrine Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Internal Medicine, School of Medicine, Firoozgar Hospital, Iran University of Medical Sciences, Tehran, Iran.
| | | | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
5
|
Gautam M, Gabrani R. Current Combinatorial Therapeutic Aspects: The Future Prospect for Glioblastoma Treatment. Curr Med Sci 2024; 44:1175-1184. [PMID: 39695017 DOI: 10.1007/s11596-024-2950-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 03/21/2024] [Indexed: 12/20/2024]
Abstract
There are several types of brain tumors but glioblastoma (GBM) is one of the highly malignant tumors. A primary concern with GBM is that the treatment is inadequate. Even after giving many multi-stacked combinations of therapies to patients, inclusive of chemotherapy, radiation, and surgery, the median survival rate remains poor. Due to its heterogeneous nature, the use of selective therapy for specific targeting of tumor cells is of particular importance. Although many treatment alternatives which include surgery with adjuvant chemotherapy and radiotherapy are available, the prognosis of the disease is very poor. Combination therapy is becoming the foundation of modern antitumor therapy and it is continuously evolving and developing innovative drug regimens as evidenced by ongoing preclinical and clinical trials. In this review, we discuss the current treatment options and emerging therapeutic approaches for the treatment of GBM. The prospects for alternative glioblastoma therapy are also discussed.
Collapse
Affiliation(s)
- Megha Gautam
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, 201309, India
| | - Reema Gabrani
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, 201309, India.
| |
Collapse
|
6
|
Dewanjee S, Bhattacharya H, Bhattacharyya C, Chakraborty P, Fleishman J, Alexiou A, Papadakis M, Jha SK. Nrf2/Keap1/ARE regulation by plant secondary metabolites: a new horizon in brain tumor management. Cell Commun Signal 2024; 22:497. [PMID: 39407193 PMCID: PMC11476647 DOI: 10.1186/s12964-024-01878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
Brain cancer is regarded as one of the most life-threatening forms of cancer worldwide. Oxidative stress acts to derange normal brain homeostasis, thus is involved in carcinogenesis in brain. The Nrf2/Keap1/ARE pathway is an important signaling cascade responsible for the maintenance of redox homeostasis, and regulation of anti-inflammatory and anticancer activities by multiple downstream pathways. Interestingly, Nrf2 plays a somewhat, contradictory role in cancers, including brain cancer. Nrf2 has traditionally been regarded as a tumor suppressor since its cytoprotective functions are considered to be the principle cellular defense mechanism against exogenous and endogenous insults, such as xenobiotics and oxidative stress. However, hyperactivation of the Nrf2 pathway supports the survival of normal as well as malignant cells, protecting them against oxidative stress, and therapeutic agents. Plants possess a pool of secondary metabolites with potential chemotherapeutic/chemopreventive actions. Modulation of Nrf2/ARE and downstream activities in a Keap1-dependant manner, with the aid of plant-derived secondary metabolites exhibits promise in the management of brain tumors. Current article highlights the effects of Nrf2/Keap1/ARE cascade on brain tumors, and the potential role of secondary metabolites regarding the management of the same.
Collapse
Affiliation(s)
- Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India.
| | - Hiranmoy Bhattacharya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Chiranjib Bhattacharyya
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Pratik Chakraborty
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, West Bengal, India
| | - Joshua Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY, 11439, USA
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Chandigarh-Ludhiana Highway, Mohali, Punjab, India
- Department of Research & Development, Funogen, Athens, 11741, Greece
- Department of Research & Development, AFNP Med, Wien, 1030, Austria
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, 2770, Australia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India.
| |
Collapse
|
7
|
Liao X, Zhang S, Li X, Qian W, Li M, Chen S, Wu X, Yu X, Li Z, Tang M, Xu Y, Yu R, Zhang Q, Wu G, Zhang N, Song L, Li J. Dynamic structural remodeling of LINC01956 enhances temozolomide resistance in MGMT-methylated glioblastoma. Sci Transl Med 2024; 16:eado1573. [PMID: 39356744 DOI: 10.1126/scitranslmed.ado1573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 07/02/2024] [Accepted: 09/11/2024] [Indexed: 10/04/2024]
Abstract
The mechanisms underlying stimuli-induced dynamic structural remodeling of RNAs for the maintenance of cellular physiological function and survival remain unclear. Here, we showed that in MGMT promoter-methylated glioblastoma (GBM), the RNA helicase DEAD-box helicase 46 (DDX46) is phosphorylated by temozolomide (TMZ)-activated checkpoint kinase 1 (CHK1), resulting in a dense-to-loose conformational change and an increase in DDX46 helicase activity. DDX46-mediated tertiary structural remodeling of LINC01956 exposes the binding motifs of LINC01956 to the 3' untranslated region of O6-methylguanine DNA methyltransferase (MGMT). This accelerates recruitment of MGMT mRNA to the RNA export machinery and transportation of MGMT mRNA from the nucleus to the cytoplasm, leading to increased MGMT abundance and TMZ resistance. Using patient-derived xenograft (PDX) and tumor organoid models, we found that treatment with the CHK1 inhibitor SRA737abolishes TMZ-induced structural remodeling of LINC01956 and subsequent MGMT up-regulation, resensitizing TMZ-resistant MGMT promoter-methylated GBM to TMZ. In conclusion, these findings highlight a mechanism underlying temozolomide-induced RNA structural remodeling and may represent a potential therapeutic strategy for patients with TMZ-resistant MGMT promoter-methylated GBM.
Collapse
Affiliation(s)
- Xinyi Liao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong 510060, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Shuxia Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Xincheng Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Wanying Qian
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Man Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Suwen Chen
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Xingui Wu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Xuexin Yu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Ziwen Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Miaoling Tang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Yingru Xu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Ruyuan Yu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Qiliang Zhang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Geyan Wu
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| | - Nu Zhang
- Department of Neurosurgery, First Affiliated Hospital, Sun Yat-sen University, Guangdong 510080, China
| | - Libing Song
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong 510060, China
| | - Jun Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangdong 510060, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangdong 510080, China
| |
Collapse
|
8
|
Reyes-Soto CY, Ramírez-Carreto RJ, Ortíz-Alegría LB, Silva-Palacios A, Zazueta C, Galván-Arzate S, Karasu Ç, Túnez I, Tinkov AA, Aschner M, López-Goerne T, Chavarría A, Santamaría A. S-allyl-cysteine triggers cytotoxic events in rat glioblastoma RG2 and C6 cells and improves the effect of temozolomide through the regulation of oxidative responses. Discov Oncol 2024; 15:272. [PMID: 38977545 PMCID: PMC11231126 DOI: 10.1007/s12672-024-01145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/03/2024] [Indexed: 07/10/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive form of cancer affecting the Central Nervous System (CNS) of thousands of people every year. Redox alterations have been shown to play a key role in the development and progression of these tumors as Reactive Oxygen Species (ROS) formation is involved in the modulation of several signaling pathways, transcription factors, and cytokine formation. The second-generation oral alkylating agent temozolomide (TMZ) is the first-line chemotherapeutic drug used to treat of GBM, though patients often develop primary and secondary resistance, reducing its efficacy. Antioxidants represent promising and potential coadjutant agents as they can reduce excessive ROS formation derived from chemo- and radiotherapy, while decreasing pharmacological resistance. S-allyl-cysteine (SAC) has been shown to inhibit the proliferation of several types of cancer cells, though its precise antiproliferative mechanisms remain poorly investigated. To date, SAC effects have been poorly explored in GBM cells. Here, we investigated the effects of SAC in vitro, either alone or in combination with TMZ, on several toxic and modulatory endpoints-including oxidative stress markers and transcriptional regulation-in two glioblastoma cell lines from rats, RG2 and C6, to elucidate some of the biochemical and cellular mechanisms underlying its antiproliferative properties. SAC (1-750 µM) decreased cell viability in both cell lines in a concentration-dependent manner, although C6 cells were more resistant to SAC at several of the tested concentrations. TMZ also produced a concentration-dependent effect, decreasing cell viability of both cell lines. In combination, SAC (1 µM or 100 µM) and TMZ (500 µM) enhanced the effects of each other. SAC also augmented the lipoperoxidative effect of TMZ and reduced cell antioxidant resistance in both cell lines by decreasing the TMZ-induced increase in the GSH/GSSG ratio. In RG2 and C6 cells, SAC per se had no effect on Nrf2/ARE binding activity, while in RG2 cells TMZ and the combination of SAC + TMZ decreased this activity. Our results demonstrate that SAC, alone or in combination with TMZ, exerts antitumor effects mediated by regulatory mechanisms of redox activity responses. SAC is also a safe drug for testing in other models as it produces non-toxic effects in primary astrocytes. Combined, these effects suggest that SAC affords antioxidant properties and potential antitumor efficacy against GBM.
Collapse
Affiliation(s)
- Carolina Y Reyes-Soto
- Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 06726, Mexico City, Mexico
| | - Ricardo J Ramírez-Carreto
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 06726, Mexico City, Mexico
- Facultad de Química, Universidad Nacional Autónoma de México, 04510, Mexico, Mexico
- Programa de Doctorado en Ciencias Biomédicas, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico
| | - Luz Belinda Ortíz-Alegría
- Laboratorio de Inmunología Experimental, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, 04530, Mexico City, Mexico
| | - Alejandro Silva-Palacios
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, SSA, 14080, Mexico City, Mexico
| | - Cecilia Zazueta
- Departamento de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, SSA, 14080, Mexico City, Mexico
| | - Sonia Galván-Arzate
- Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, S.S, 14269, Mexico City, Mexico
| | - Çimen Karasu
- Department of Medical Pharmacology, Cellular Stress Response and Signal Transduction Research Laboratory, Faculty of Medicine, Gazi University, 06500, Ankara, Turkey
| | - Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina y Enfermería, Instituto de Investigaciones Biomédicas Maimónides de Córdoba (IMIBIC)Universidad de CórdobaRed Española de Excelencia en Estimulación Cerebral (REDESTIM), 14071, Córdoba, Spain
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119435, Russia
- Departament of Elementology, and Department of Human Ecology and Bioelementology, Peoples' Friendship University of Russia (RUDN University), Moscow, 117198, Russia
- Laboratory of Molecular Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150003, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Tessy López-Goerne
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico
| | - Anahí Chavarría
- Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, 06726, Mexico City, Mexico.
| | - Abel Santamaría
- Laboratorio de Nanotecnología y Nanomedicina, Departamento de Atención a la Salud, Universidad Autónoma Metropolitana-Xochimilco, 04960, Mexico City, Mexico.
- Facultad de Ciencias, Universidad Nacional Autónoma de México, 04510, Mexico City, Mexico.
| |
Collapse
|
9
|
Majchrzak-Celińska A, Studzińska-Sroka E. New Avenues and Major Achievements in Phytocompounds Research for Glioblastoma Therapy. Molecules 2024; 29:1682. [PMID: 38611962 PMCID: PMC11013944 DOI: 10.3390/molecules29071682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/14/2024] Open
Abstract
Phytocompounds have been evaluated for their anti-glioblastoma actions for decades, with promising results from preclinical studies but only limited translation into clinics. Indeed, by targeting multiple signaling pathways deregulated in cancer, they often show high efficacy in the in vitro studies, but their poor bioavailability, low tumor accumulation, and rapid clearance compromise their efficacy in vivo. Here, we present the new avenues in phytocompound research for the improvement of glioblastoma therapy, including the ways to enhance the response to temozolomide using phytochemicals, the current focus on phytocompound-based immunotherapy, or the use of phytocompounds as photosensitizers in photodynamic therapy. Moreover, we present new, intensively evaluated approaches, such as chemical modifications of phytochemicals or encapsulation into numerous types of nanoformulations, to improve their bioavailability and delivery to the brain. Finally, we present the clinical trials evaluating the role of phytocompounds or phytocompound-derived drugs in glioblastoma therapy and the less studied phytocompounds or plant extracts that have only recently been found to possess promising anti-glioblastoma properties. Overall, recent advancements in phytocompound research are encouraging; however, only with more 3D glioblastoma models, in vivo studies, and clinical trials it is possible to upgrade the role of phytocompounds in glioblastoma treatment to a satisfactory level.
Collapse
Affiliation(s)
- Aleksandra Majchrzak-Celińska
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland
| | - Elżbieta Studzińska-Sroka
- Department of Pharmacognosy and Biomaterials, Poznan University of Medical Sciences, Rokietnicka 3 Str., 60-806 Poznan, Poland;
| |
Collapse
|
10
|
Prakash V, Bose C, Sunilkumar D, Cherian RM, Thomas SS, Nair BG. Resveratrol as a Promising Nutraceutical: Implications in Gut Microbiota Modulation, Inflammatory Disorders, and Colorectal Cancer. Int J Mol Sci 2024; 25:3370. [PMID: 38542344 PMCID: PMC10970219 DOI: 10.3390/ijms25063370] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/21/2024] [Accepted: 02/25/2024] [Indexed: 12/20/2024] Open
Abstract
Natural products have been a long-standing source for exploring health-beneficial components from time immemorial. Modern science has had a renewed interest in natural-products-based drug discovery. The quest for new potential secondary metabolites or exploring enhanced activities for existing molecules remains a pertinent topic for research. Resveratrol belongs to the stilbenoid polyphenols group that encompasses two phenol rings linked by ethylene bonds. Several plant species and foods, including grape skin and seeds, are the primary source of this compound. Resveratrol is known to possess potent anti-inflammatory, antiproliferative, and immunoregulatory properties. Among the notable bioactivities associated with resveratrol, its pivotal role in safeguarding the intestinal barrier is highlighted for its capacity to prevent intestinal inflammation and regulate the gut microbiome. A better understanding of how oxidative stress can be controlled using resveratrol and its capability to protect the intestinal barrier from a gut microbiome perspective can shed more light on associated physiological conditions. Additionally, resveratrol exhibits antitumor activity, proving its potential for cancer treatment and prevention. Moreover, cardioprotective, vasorelaxant, phytoestrogenic, and neuroprotective benefits have also been reported. The pharmaceutical industry continues to encounter difficulties administering resveratrol owing to its inadequate bioavailability and poor solubility, which must be addressed simultaneously. This report summarizes the currently available literature unveiling the pharmacological effects of resveratrol.
Collapse
Affiliation(s)
- Vidhya Prakash
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Chinchu Bose
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Damu Sunilkumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
- Department of Cancer Biology and Genetics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Robin Mathew Cherian
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Shwetha Susan Thomas
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Bipin G. Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| |
Collapse
|
11
|
Li HY, Feng YH, Lin CL, Hsu TI. Mitochondrial Mechanisms in Temozolomide Resistance: Unraveling the Complex Interplay and Therapeutic Strategies in Glioblastoma. Mitochondrion 2024; 75:101836. [PMID: 38158149 DOI: 10.1016/j.mito.2023.101836] [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: 03/28/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/03/2024]
Abstract
Glioblastoma (GBM) is a highly aggressive and lethal brain tumor, with temozolomide (TMZ) being the standard chemotherapeutic agent for its treatment. However, TMZ resistance often develops, limiting its therapeutic efficacy and contributing to poor patient outcomes. Recent evidence highlights the crucial role of mitochondria in the development of TMZ resistance through various mechanisms, including alterations in reactive oxygen species (ROS) production, metabolic reprogramming, apoptosis regulation, biogenesis, dynamics, stress response, and mtDNA mutations. This review article aims to provide a comprehensive overview of the mitochondrial mechanisms involved in TMZ resistance and discuss potential therapeutic strategies targeting these mechanisms to overcome resistance in GBM. We explore the current state of clinical trials targeting mitochondria or related pathways in primary GBM or recurrent GBM, as well as the challenges and future perspectives in this field. Understanding the complex interplay between mitochondria and TMZ resistance will facilitate the development of more effective therapeutic strategies and ultimately improve the prognosis for GBM patients.
Collapse
Affiliation(s)
- Hao-Yi Li
- Department of Biochemistry, Ludwig-Maximilians-University, Munich 81377, Germany; Gene Center, Ludwig-Maximilians-University, Munich 81377, Germany
| | | | | | - Tsung-I Hsu
- Ph.D. Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University and National Health Research Institutes, Taipei 110, Taiwan; International Master Program in Medical Neuroscience, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Neuroscience, Taipei Medical University, Taipei 110, Taiwan; TMU Research Center of Cancer Translational Medicine, Taipei 110, Taiwan.
| |
Collapse
|
12
|
Mittal S, Shah S, Yadav HN, Ali J, Gupta MM, Baboota S. Quality by design engineered, enhanced anticancer activity of temozolomide and resveratrol coloaded NLC and brain targeting via lactoferrin conjugation in treatment of glioblastoma. Eur J Pharm Biopharm 2023; 191:175-188. [PMID: 37648174 DOI: 10.1016/j.ejpb.2023.08.018] [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: 05/28/2023] [Revised: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/01/2023]
Abstract
The most dangerous type of high-grade astrocytoma is glioblastoma multiforme. The objective of the work was to engineer lactoferrin conjugated temozolomide and resveratrol co-loaded NLC for the treatment of glioblastoma using intranasal delivery for brain targeting. Synergistic activity of temozolomide and resveratrol was determined using combination index method and 1:1 ratio was selected. QbD approach was used to formulate and optimize NLC, with minimum particle size, maximum transmittance and entrapment efficiency using Central Composite Rotable Design (CCRD) method. The optimized LTR-NLC had desired average particle size (209.3 nm), narrow PDI along, high percentage transmittance (>95%) and better entrapment efficiency (95.26% of TEM and 87.59% of RES). From ex-vivo permeation studies it was found that the permeation at 24 h was 77.43 %, and 88.55 % from LTR-NLC and 25.76 % and 31.10% from suspension for resveratrol and temozolomide respectively. In comparison to drug suspension, NLC had nearly 3-fold increase in drug penetration. IC50 value was also significantly better in the groups treated with LTR-NLC. Hence it can be concluded that LTR-NLC may be an effective formulation for the treatment of glioblastoma, according to the findings of this investigation.
Collapse
Affiliation(s)
- Saurabh Mittal
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India; Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Noida 201303, U.P., India.
| | - Sadia Shah
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Harlokesh Narayan Yadav
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Madan Mohan Gupta
- School of Pharmacy, Faculty of Medical Science, The University of the West Indies, St. Augustine, Trinidad & Tobago.
| | - Sanjula Baboota
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
13
|
Kumari S, Gupta R, Ambasta RK, Kumar P. Multiple therapeutic approaches of glioblastoma multiforme: From terminal to therapy. Biochim Biophys Acta Rev Cancer 2023; 1878:188913. [PMID: 37182666 DOI: 10.1016/j.bbcan.2023.188913] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 05/10/2023] [Indexed: 05/16/2023]
Abstract
Glioblastoma multiforme (GBM) is an aggressive brain cancer showing poor prognosis. Currently, treatment methods of GBM are limited with adverse outcomes and low survival rate. Thus, advancements in the treatment of GBM are of utmost importance, which can be achieved in recent decades. However, despite aggressive initial treatment, most patients develop recurrent diseases, and the overall survival rate of patients is impossible to achieve. Currently, researchers across the globe target signaling events along with tumor microenvironment (TME) through different drug molecules to inhibit the progression of GBM, but clinically they failed to demonstrate much success. Herein, we discuss the therapeutic targets and signaling cascades along with the role of the organoids model in GBM research. Moreover, we systematically review the traditional and emerging therapeutic strategies in GBM. In addition, we discuss the implications of nanotechnologies, AI, and combinatorial approach to enhance GBM therapeutics.
Collapse
Affiliation(s)
- Smita Kumari
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, India
| | - Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University, India.
| |
Collapse
|
14
|
Pizzimenti C, Fiorentino V, Franchina M, Martini M, Giuffrè G, Lentini M, Silvestris N, Di Pietro M, Fadda G, Tuccari G, Ieni A. Autophagic-Related Proteins in Brain Gliomas: Role, Mechanisms, and Targeting Agents. Cancers (Basel) 2023; 15:cancers15092622. [PMID: 37174088 PMCID: PMC10177137 DOI: 10.3390/cancers15092622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
The present review focuses on the phenomenon of autophagy, a catabolic cellular process, which allows for the recycling of damaged organelles, macromolecules, and misfolded proteins. The different steps able to activate autophagy start with the formation of the autophagosome, mainly controlled by the action of several autophagy-related proteins. It is remarkable that autophagy may exert a double role as a tumour promoter and a tumour suppressor. Herein, we analyse the molecular mechanisms as well as the regulatory pathways of autophagy, mainly addressing their involvement in human astrocytic neoplasms. Moreover, the relationships between autophagy, the tumour immune microenvironment, and glioma stem cells are discussed. Finally, an excursus concerning autophagy-targeting agents is included in the present review in order to obtain additional information for the better treatment and management of therapy-resistant patients.
Collapse
Affiliation(s)
- Cristina Pizzimenti
- Translational Molecular Medicine and Surgery, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy
| | - Vincenzo Fiorentino
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Mariausilia Franchina
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Giuseppe Giuffrè
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Maria Lentini
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Nicola Silvestris
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Oncology Section, University of Messina, 98125 Messina, Italy
| | - Martina Di Pietro
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Oncology Section, University of Messina, 98125 Messina, Italy
| | - Guido Fadda
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Giovanni Tuccari
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| | - Antonio Ieni
- Department of Human Pathology in Adult and Developmental Age "Gaetano Barresi", Pathology Section, University of Messina, 98125 Messina, Italy
| |
Collapse
|
15
|
Wang M, Zhang Y, Liu M, Jia Y, He J, Xu X, Shi H, Zhang Y, Zhang J, Liu Y. Inhibition of STAT3 signaling as critical molecular event in HUC-MSCs suppressed Glioblastoma Cells. J Cancer 2023; 14:611-627. [PMID: 37057281 PMCID: PMC10088538 DOI: 10.7150/jca.77905] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/17/2023] [Indexed: 03/13/2023] Open
Abstract
Objective: We investigated the effect of human umbilical cord mesenchymal stem cells (HUC-MSCs) supernatants on proliferation, migration, invasion, and apoptosis in glioblastoma (GBM) cell lines RG-2, U251, U87-MG, and LN-428, as well as their apoptosis and autophagy-mediated through IL-6/JAK2/STAT3 signaling pathway to explore the molecular mechanisms. Methods: In this study, RG-2, U251, U87-MG, and LN-428 cells were treated with 9 mg/ml HUC-MSCs supernatants. Their responses to HUC-MSCs supernatants treatment and the status of STAT3 signaling were analyzed by multiple experimental approaches to elucidate the importance of HUC-MSCs supernatants for GBM. Results: The results demonstrated that after treatment with HUC-MSCs supernatants, in vitro proliferation of RG-2, U251, U87-MG, and LN-428 cells were inhibited, and their sustained growth was also blocked. RG-2, U251, and U87-MG cells showed significant S phase accumulation, while LN-428 cells were blocked in G0/G1 phase. Their migratory invasive capacities were inhibited, and their apoptosis and autophagy ratios were increased. These effects were mediated through the IL-6/JAK2/STAT3 and its downstream signaling pathway. Conclusion: Our data showed that HUC-MSCs supernatants had anti-tumor effects on GBM cells. It inhibited the proliferation, migration, and invasion of GBM cells and promoted their apoptosis. Negative regulation of the IL-6/JAK2/STAT3 signaling pathway enhanced apoptosis and autophagy in tumor cells, thereby improving the therapeutic effect on GBM.
Collapse
Affiliation(s)
- Mingming Wang
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Yufu Zhang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Min Liu
- Department of Pathology, Affiliated Hospital of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Yuna Jia
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Jing He
- Laboratory of Obstetrics and Gynecology, Affiliated Hospital of Yan'an University, Yan'an, 716000 Shaanxi Province, China
| | - Xiangrong Xu
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Haiyan Shi
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Yunqing Zhang
- Laboratory of Obstetrics and Gynecology, Affiliated Hospital of Yan'an University, Yan'an, 716000 Shaanxi Province, China
| | - Jing Zhang
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an 716000, Shaanxi Province, China
| | - Yusi Liu
- Department of Cell Biology and Genetics, Medical College of Yan'an University, Yan'an 716000, Shaanxi Province, China
| |
Collapse
|
16
|
Liu G, Guan Y, Liu Y, Wang Y, Zhang J, Liu Y, Liu X. Saikosaponin D Inducing Apoptosis and Autophagy through the Activation of Endoplasmic Reticulum Stress in Glioblastoma. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1-12. [PMID: 36467888 PMCID: PMC9715330 DOI: 10.1155/2022/5489553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2024]
Abstract
Saikosaponin D (SSD), a saponin derivative, is extracted from Bupleurum falcatum. It exhibits an inhibitory effect on a number of tumor cells and is relatively safe when used at therapeutic doses. However, its effects on glioblastoma multiforme (GBM) have not been fully explored. This study is aimed at investigating the cytotoxic effects of SSD in GBM cell lines. SSD induces apoptosis and autophagy by activating endoplasmic reticulum (ER) stress in GBM cells. GBM cell proliferation activity and morphology were observed using the Cell Counting Kit-8 assay and hematoxylin and eosin staining. Hoechst 33258 fluorescence staining and flow cytometry were performed to assess apoptosis. Western blotting and immunocytochemical staining were used to detect protein expression and distribution. SSD significantly inhibited the proliferation of RG-2, U87-MG, and U251 cells in a dose-dependent manner, and the proportion of apoptotic cells increased significantly. Additionally, the expressions of ER-, apoptosis-, and autophagy-related proteins were significantly upregulated and distributed in the cytoplasm and nucleus. Therefore, SSD may be considered a novel treatment option for GBM. This study demonstrated the anti-GBM effect of SSD from the perspectives of cell apoptosis and autophagy.
Collapse
Affiliation(s)
- Guimei Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yuehong Guan
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yongxian Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yaping Wang
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Jing Zhang
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Yusi Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| | - Xiaobin Liu
- Department of Medical Immunology, College of Basic Medical Sciences, Yan’an University, Yan’an 716000, China
| |
Collapse
|
17
|
Karthika C, Najda A, Klepacka J, Zehravi M, Akter R, Akhtar MF, Saleem A, Al-Shaeri M, Mondal B, Ashraf GM, Tagde P, Ramproshad S, Ahmad Z, Khan FS, Rahman MH. Involvement of Resveratrol against Brain Cancer: A Combination Strategy with a Pharmaceutical Approach. Molecules 2022; 27:4663. [PMID: 35889532 PMCID: PMC9320031 DOI: 10.3390/molecules27144663] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022] Open
Abstract
A brain tumor (BT) is a condition in which there is growth or uncontrolled development of the brain cells, which usually goes unrecognized or is diagnosed at the later stages. Since the mechanism behind BT is not clear, and the various physiological conditions are difficult to diagnose, the success rate of BT is not very high. This is the central issue faced during drug development and clinical trials with almost all types of neurodegenerative disorders. In the first part of this review, we focus on the concept of brain tumors, their barriers, and the types of delivery possible to target the brain cells. Although various treatment methods are available, they all have side effects or toxic effects. Hence, in the second part, a correlation was made between the use of resveratrol, a potent antioxidant, and its advantages for brain diseases. The relationship between brain disease and the blood-brain barrier, multi-drug resistance, and the use of nanomedicine for treating brain disorders is also mentioned. In short, a hypothetical concept is given with a background investigation into the use of combination therapy with resveratrol as an active ingredient, the possible drug delivery, and its formulation-based approach.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, India;
| | - Agnieszka Najda
- Department of Vegetable and Herbal Crops, University of Life Science in Lublin, Doświadczalna Street 51A, 20280 Lublin, Poland
| | - Joanna Klepacka
- Department of Commodity Science and Food Analysis, Faculty of Food Science, University of Warmia and Mazury in Olsztyn, Oczapowskiego 2, 10719 Olsztyn, Poland;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Lahore Campus, Riphah International University, Lahore 54950, Pakistan;
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Majed Al-Shaeri
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (B.M.); (S.R.)
| | - Ghulam Md. Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Priti Tagde
- Amity Institute of Pharmacy, Amity University, Noida 201301, India;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (B.M.); (S.R.)
| | - Zubair Ahmad
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
- Biology Department, College of Arts and Sciences, Dehran Al-Junub, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Farhat S. Khan
- Biology Department, College of Arts and Sciences, Dehran Al-Junub, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Korea;
| |
Collapse
|
18
|
Ma W, Liu Y, Lei P, Zhu M, Pan X. Novel Compound, ND-17, Regulates the JAK/STAT, PI3K/AKT, and MAPK Pathways and Restrains Human T-Lymphoid Leukemia Development. Curr Cancer Drug Targets 2022; 22:404-413. [DOI: 10.2174/1568009622666220304202116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/27/2021] [Accepted: 01/21/2022] [Indexed: 11/22/2022]
Abstract
Background:
T cell acute lymphoblastic leukemia (T-ALL) is an invasive hematological malignant disorder of T cell progenitors. The Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling pathway plays an important role in the development of T-ALL and in the inhibition of the key molecule, JAK2, and could suppress T-ALL cell proliferation.
Objective:
The objective of this study was to investigate the in vitro anti-tumor effects of a novel nilotinib derivative, ND-17, on cancer cell lines via its interactions with JAK2.
Methods:
The effects of ND-17 on cell proliferation and on cell cycle and apoptosis were evaluated using the tetrazolium assay and flow cytometry, respectively. In addition, the ND-17/JAK2 binding interactions were evaluated using surface plasmon resonance and western blot analyses.
Results:
ND-17 exerted the greatest inhibitory effects on T-ALL cells amongst all hematological cancer cell lines tested. Flow cytometric analysis indicated that ND-17 blocked the cell cycle at the S phase in T-ALL cells. Nilotinib did not significantly inhibit T-ALL cell growth or regulate cell cycle. Preliminary investigations revealed that the regulation of cyclin-dependent kinases/cyclins was attributed to ND-17-induced cell cycle arrest. Furthermore, ND-17 could bind to JAK2 with strong affinity and more importantly, ND-17 bound to the ATP pocket of JAK2 in a manner similar to the potent inhibitor. Thus, ND-17 treatment exhibited a prominent effect in inhibiting the phosphorylation of JAK2 in T-ALL cells. An increase in the phosphorylation of JAK2 was observed in interleukin-6-stimulated Jurkat cells, which was reversed by ND-17 treatment. Meanwhile, the combination of TG-101348 and ND-17 led to further improvement in inhibiting phosphorylation of JAK2. Moreover, the transfection and knockdown of JAK2 altered the inhibitory effect of ND-17 on Jurkat cell viability. In addition, ND-17 treatment suppressed the JAK/STAT, phosphatidylinositol-3-kinase/protein kinase B/mechanistic target of rapamycin, and mitogen-activated protein kinase/extracellular signal-regulated protein kinases 1 and 2 signaling pathways.
Conclusion:
These findings suggest that ND-17 could be a promising JAK2 inhibitor for the treatment of T-ALL.
Collapse
Affiliation(s)
- Weina Ma
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Yanhong Liu
- State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi’an 710061, P.R. China
- State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi’an 710061, P.R. China
| | - Panpan Lei
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, P.R. China
| | - Man Zhu
- State Key Laboratory of Shaanxi for Natural Medicines Research and Engineering, Xi’an 710061, P.R. China
| | - Xiaoyan Pan
- School of Pharmacy, Health Science Center, Xi’an Jiaotong University, Xi’an 710061, P.R. China
| |
Collapse
|
19
|
Peter K, Kar SK, Gothalwal R, Gandhi P. Curcumin in Combination with Other Adjunct Therapies for Brain Tumor Treatment: Existing Knowledge and Blueprint for Future Research. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2022; 10:163-181. [PMID: 35178355 PMCID: PMC8800460 DOI: 10.22088/ijmcm.bums.10.3.163] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 01/03/2022] [Indexed: 12/12/2022]
Abstract
Malignant brain tumors proliferate aggressively and have a debilitating outcome. Surgery followed by chemo-radiotherapy has been the standard procedure of care since 2005 but issues of therapeutic toxicity and relapse still remain unaddressed. Repurposing of drugs to develop novel combinations that can augment existing treatment regimens for brain tumors is the need of the hour. Herein, we discuss studies documenting the use of curcumin as an adjuvant to conventional and alternative therapies for brain tumors. Comprehensive analysis of data suggests that curcumin together with available therapies can generate a synergistic action achieved through multiple molecular targeting, which results in simultaneous inhibition of tumor growth, and reduced treatment-induced toxicity as well as resistance. The review also highlights approaches to increase bioavailability and bioaccumulation of drugs when co-delivered with curcumin using nano-cargos. Despite substantial preclinical work on radio-chemo sensitizing effects of curcumin, to date, there is only a single clinical report on brain tumors. Based on available lab evidence, it is proposed that antibody-conjugated nano-curcumin in combination with sub-toxic doses of conventional or repurposed therapeutics should be designed and tested in clinical studies. This will increase tumor targeting, the bioavailability of the drug combination, reduce therapy resistance, and tumor recurrence through modulation of aberrant signaling cascades; thus improving clinical outcomes in brain malignancies.
Collapse
Affiliation(s)
- Kavita Peter
- Department of Biotechnology, Barkatullah University, Bhopal, M.P, India
| | | | - Ragini Gothalwal
- Department of Biotechnology, Barkatullah University, Bhopal, M.P, India
| | - Puneet Gandhi
- Department of Research, Bhopal Memorial Hospital and Research Centre, Bhopal, M.P, India
| |
Collapse
|
20
|
OUP accepted manuscript. J AOAC Int 2022; 105:1258-1267. [DOI: 10.1093/jaoacint/qsac045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 11/13/2022]
|
21
|
Liu Y, Chen Y, Zhu R, Xu L, Xie HQ, Zhao B. Rutaecarpine Inhibits U87 Glioblastoma Cell Migration by Activating the Aryl Hydrocarbon Receptor Signaling Pathway. Front Mol Neurosci 2021; 14:765712. [PMID: 34955744 PMCID: PMC8696176 DOI: 10.3389/fnmol.2021.765712] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/10/2021] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma is the most frequent and aggressive primary astrocytoma in adults. The high migration ability of the tumor cells is an important reason for the high recurrence rate and poor prognosis of glioblastoma. Recently, emerging evidence has shown that the migration ability of glioblastoma cells was inhibited upon the activation of aryl hydrocarbon receptor (AhR), suggesting potential anti-tumor effects of AhR agonists. Rutaecarpine is a natural compound with potential tumor therapeutic effects which can possibly bind to AhR. However, its effect on the migration of glioblastoma is unclear. Therefore, we aim to explore the effects of rutaecarpine on the migration of human glioblastoma cells U87 and the involvement of the AhR signaling pathway. The results showed that: (i) compared with other structural related alkaloids, like evodiamine and dehydroevodiamine, rutaecarpine was a more potent AhR activator, and has a stronger inhibitory effect on the glioblastoma cell migration; (ii) rutaecarpine decreased the migration ability of U87 cells in an AhR-dependent manner; (iii) AhR mediated the expression of a tumor suppressor interleukin 24 (IL24) induced by rutaecarpine, and AhR-IL24 axis was involved in the anti-migratory effects of rutaecarpine on the glioblastoma. Besides IL24, other candidates AhR downstream genes both associated with cancer and migration were proposed to participate in the migration regulation of rutaecarpine by RNA-Seq and bioinformatic analysis. These data indicate that rutaecarpine is a naturally-derived AhR agonist that could inhibit the migration of U87 human glioblastoma cells mostly via the AhR-IL24 axis.
Collapse
Affiliation(s)
- Yiyun Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yangsheng Chen
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Ruihong Zhu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
22
|
Medeiros M, Candido MF, Valera ET, Brassesco MS. The multifaceted NF-kB: are there still prospects of its inhibition for clinical intervention in pediatric central nervous system tumors? Cell Mol Life Sci 2021; 78:6161-6200. [PMID: 34333711 PMCID: PMC11072991 DOI: 10.1007/s00018-021-03906-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/16/2022]
Abstract
Despite advances in the understanding of the molecular mechanisms underlying the basic biology and pathogenesis of pediatric central nervous system (CNS) malignancies, patients still have an extremely unfavorable prognosis. Over the years, a plethora of natural and synthetic compounds has emerged for the pharmacologic intervention of the NF-kB pathway, one of the most frequently dysregulated signaling cascades in human cancer with key roles in cell growth, survival, and therapy resistance. Here, we provide a review about the state-of-the-art concerning the dysregulation of this hub transcription factor in the most prevalent pediatric CNS tumors: glioma, medulloblastoma, and ependymoma. Moreover, we compile the available literature on the anti-proliferative effects of varied NF-kB inhibitors acting alone or in combination with other therapies in vitro, in vivo, and clinical trials. As the wealth of basic research data continues to accumulate, recognizing NF-kB as a therapeutic target may provide important insights to treat these diseases, hopefully contributing to increase cure rates and lower side effects related to therapy.
Collapse
Affiliation(s)
- Mariana Medeiros
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Marina Ferreira Candido
- Department of Cell Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elvis Terci Valera
- Department of Pediatrics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - María Sol Brassesco
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, FFCLRP-USP, University of São Paulo, Av. Bandeirantes, 3900, Bairro Monte Alegre, Ribeirão Preto, São Paulo, CEP 14040-901, Brazil.
| |
Collapse
|
23
|
Human astrocytes and astrocytoma respond differently to resveratrol. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 37:102441. [PMID: 34302989 DOI: 10.1016/j.nano.2021.102441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 06/01/2021] [Accepted: 07/06/2021] [Indexed: 12/17/2022]
Abstract
A fundamental problem in oncology is that anticancer chemotherapeutics kill both cancer and healthy cells in the surrounding tissues. Resveratrol is a natural antioxidant with intriguing and opposing biological properties: it reduces viability of some cancer cells but not of non-transformed ones (in equimolar concentrations). Therefore, we examined resveratrol in human non-transformed primary astrocytes and astrocytoma. Resveratrol reduced reactive oxygen species in astrocytes, but not in astrocytoma. Such cell-type dependent response is particularly evident with analyses at the single cell level showing clear population difference in high and low glutathione levels. Due to resveratrol's poor aqueous solubility that limits its use in clinics, we incorporated it into stimulus-responsive micelles assembled from miktoarm polymers. This could be an attractive chemotherapeutic delivery strategy in nano-oncology. As a proof of principle, we show that these formulations containing resveratrol markedly decrease astrocytoma viability, particularly in combination with temozolomide, a first line chemotherapeutic for astrocytoma.
Collapse
|
24
|
Arabzadeh A, Mortezazadeh T, Aryafar T, Gharepapagh E, Majdaeen M, Farhood B. Therapeutic potentials of resveratrol in combination with radiotherapy and chemotherapy during glioblastoma treatment: a mechanistic review. Cancer Cell Int 2021; 21:391. [PMID: 34289841 PMCID: PMC8296583 DOI: 10.1186/s12935-021-02099-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 07/15/2021] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma, WHO grade IV astrocytoma, is the most aggressive type of brain tumors. These cancerous cells have a rapid growth rate, tendency to penetrate vital brain structures, molecular heterogeneity, etc. and this cancer is associated with a poor prognosis and low survival rate. Due to the resistance of glioblastoma cells to conventional therapeutic modalities (such as radiation therapy and chemotherapy) as well as the adverse effects of these modalities, the researchers have attempted to discover an appropriate alternative or adjuvant treatment for glioblastoma. Resveratrol, as an herbal and natural polyphenolic compound, has anti-tumoral property and has shown to be effective in GBM treatment. Resveratrol exerts its anti-tumoral effect through various mechanisms such as regulation of cell cycle progression and cell proliferation, autophagy, oxidant system, apoptosis pathways, and so on. Resveratrol in combination with radiation therapy and chemotherapy has also been used. In the present study, we summarized the current findings on therapeutic potentials of resveratrol in glioblastoma radiotherapy and chemotherapy.
Collapse
Affiliation(s)
- AmirAhmad Arabzadeh
- Department of Surgery, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Tohid Mortezazadeh
- Department of Medical Physics, School of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Tayebeh Aryafar
- Department of Radiation Sciences, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Esmaeil Gharepapagh
- Medical Radiation Sciences Research Team , Tabriz University of Medical Science, Tabriz, Iran
| | - Mehrsa Majdaeen
- Department of Radiotherapy and Oncology, Razi Hospital, Guilan University of Medical Sciences, Rasht, Iran.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
25
|
More MP, Pardeshi SR, Pardeshi CV, Sonawane GA, Shinde MN, Deshmukh PK, Naik JB, Kulkarni AD. Recent advances in phytochemical-based Nano-formulation for drug-resistant Cancer. MEDICINE IN DRUG DISCOVERY 2021. [DOI: 10.1016/j.medidd.2021.100082] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
26
|
Chelliah SS, Paul EAL, Kamarudin MNA, Parhar I. Challenges and Perspectives of Standard Therapy and Drug Development in High-Grade Gliomas. Molecules 2021; 26:1169. [PMID: 33671796 PMCID: PMC7927069 DOI: 10.3390/molecules26041169] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/18/2022] Open
Abstract
Despite their low incidence rate globally, high-grade gliomas (HGG) remain a fatal primary brain tumor. The recommended therapy often is incapable of resecting the tumor entirely and exclusively targeting the tumor leads to tumor recurrence and dismal prognosis. Additionally, many HGG patients are not well suited for standard therapy and instead, subjected to a palliative approach. HGG tumors are highly infiltrative and the complex tumor microenvironment as well as high tumor heterogeneity often poses the main challenges towards the standard treatment. Therefore, a one-fit-approach may not be suitable for HGG management. Thus, a multimodal approach of standard therapy with immunotherapy, nanomedicine, repurposing of older drugs, use of phytochemicals, and precision medicine may be more advantageous than a single treatment model. This multimodal approach considers the environmental and genetic factors which could affect the patient's response to therapy, thus improving their outcome. This review discusses the current views and advances in potential HGG therapeutic approaches and, aims to bridge the existing knowledge gap that will assist in overcoming challenges in HGG.
Collapse
Affiliation(s)
- Shalini Sundramurthi Chelliah
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
- School of Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Ervin Ashley Lourdes Paul
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Muhamad Noor Alfarizal Kamarudin
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| | - Ishwar Parhar
- Brain Research Institute Monash Sunway, Jeffrey Cheah School of Medicine and Health Science, Monash University Malaysia, Bandar Sunway 47500, Malaysia; (S.S.C.); (E.A.L.P.); (M.N.A.K.)
| |
Collapse
|