1
|
Hong Y, Peng J, Chen Q, Zhou Q, Xu F, Yao J, Zou Q, Yuan L, Li L, Long Q, Liao L, Liu M, Liu X, Zhang D, Wang S, Yi W. Comparison of neoadjuvant chemotherapy response and prognosis among pegylated liposomal doxorubicin, epirubicin and pirarubicin in HR ⩽ 10%/HER2-negative breast cancer: an exploratory real-world multicentre cohort study. Ther Adv Med Oncol 2024; 16:17588359241279695. [PMID: 39346118 PMCID: PMC11428166 DOI: 10.1177/17588359241279695] [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: 03/20/2024] [Accepted: 08/14/2024] [Indexed: 10/01/2024] Open
Abstract
Background Pegylated liposomal doxorubicin (PLD), epirubicin and pirarubicin are the main anthracyclines widely used in China. PLD demonstrates therapeutic response comparable to epirubicin and pirarubicin in neoadjuvant chemotherapy (NAC) of breast cancer. Objectives The objectives of our study were to retrospectively assess the real-world effectiveness and prognostic characteristics of PLD as NAC for HR ⩽ 10%/human epidermal growth factor receptor 2 (HER2)-negative breast cancer. Design This was a retrospective study. Methods Our study enrolled patients with HR ⩽ 10%/HER2-negative breast cancer who received PLD-, epirubicin- or pirarubicin-based NAC from three centres in Hunan Province, China, between 2015 and 2022. We employed inverse probability of treatment weighting to balance the differences in patients' characteristics among the PLD, epirubicin, and pirarubicin groups. The endpoints were pathological complete response (pCR), event-free survival (EFS), and overall survival (OS). Results A total of 267 patients were included. After NAC, the pCR rates in PLD group were superior to epirubicin group (PLD, 34.1%; epirubicin, 20.8%, p = 0.038). The differences in EFS (log-rank p = 0.99) and OS (log-rank p = 0.33) among the three groups were not statistically significant. Among the three groups, non-pCR patients had worse EFS than pCR patients (log-rank p = 0.014). For patients with pCR, the differences in EFS (log-rank p = 0.47) and OS (log-rank p = 0.38) were not statistically significant among the three groups, and the EFS (log-rank p = 0.59) and OS (log-rank p = 0.14) of non-pCR patients in the PLD group were similar to those in the epirubicin and pirarubicin groups. Conclusion PLD had a similar therapeutic response and prognosis compared to epirubicin or pirarubicin in NAC for patients with HR ⩽ 10%/HER2 negative breast cancer, which means that PLD represents a potential NAC option.
Collapse
Affiliation(s)
- Yue Hong
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Jing Peng
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Qin Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Feng Xu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Jia Yao
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Qiongyan Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Liqin Yuan
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Lun Li
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre For Breast Disease In Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Liqiu Liao
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Mingwen Liu
- Department of Breast Surgery of the First People's Hospital of Xiangtan City, Shuyuan Road 100#, Xiangtan City, Hunan Province, China
| | - Xuan Liu
- Department of Breast Surgery of the First People's Hospital of Xiangtan City, Shuyuan Road 100#, Xiangtan City, Hunan Province, China
| | - Danhua Zhang
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre for Breast Disease in Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| | - Shouman Wang
- Department of Breast Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No. 139, Renmin Central Road, Changsha 410011, China
- Clinical Research Centre for Breast Disease in Hunan Province, No. 139, Renmin Central Road, Changsha 410011, China
| |
Collapse
|
2
|
Jiang Y, Li Y. Nutrition Intervention and Microbiome Modulation in the Management of Breast Cancer. Nutrients 2024; 16:2644. [PMID: 39203781 PMCID: PMC11356826 DOI: 10.3390/nu16162644] [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: 07/18/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Breast cancer (BC) is one of the most common cancers worldwide and a leading cause of cancer-related deaths among women. The escalating incidence of BC underscores the necessity of multi-level treatment. BC is a complex and heterogeneous disease involving many genetic, lifestyle, and environmental factors. Growing evidence suggests that nutrition intervention is an evolving effective prevention and treatment strategy for BC. In addition, the human microbiota, particularly the gut microbiota, is now widely recognized as a significant player contributing to health or disease status. It is also associated with the risk and development of BC. This review will focus on nutrition intervention in BC, including dietary patterns, bioactive compounds, and nutrients that affect BC prevention and therapeutic responses in both animal and human studies. Additionally, this paper examines the impacts of these nutrition interventions on modulating the composition and functionality of the gut microbiome, highlighting the microbiome-mediated mechanisms in BC. The combination treatment of nutrition factors and microbes is also discussed. Insights from this review paper emphasize the necessity of comprehensive BC management that focuses on the nutrition-microbiome axis.
Collapse
Affiliation(s)
| | - Yuanyuan Li
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
3
|
Liu P, Zhang B, Li Y, Yuan Q. Potential mechanisms of cancer prevention and treatment by sulforaphane, a natural small molecule compound of plant-derived. Mol Med 2024; 30:94. [PMID: 38902597 PMCID: PMC11191161 DOI: 10.1186/s10020-024-00842-7] [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: 12/28/2023] [Accepted: 05/20/2024] [Indexed: 06/22/2024] Open
Abstract
Despite recent advances in tumor diagnosis and treatment technologies, the number of cancer cases and deaths worldwide continues to increase yearly, creating an urgent need to find new methods to prevent or treat cancer. Sulforaphane (SFN), as a member of the isothiocyanates (ITCs) family, which is the hydrolysis product of glucosinolates (GLs), has been shown to have significant preventive and therapeutic cancer effects in different human cancers. Early studies have shown that SFN scavenges oxygen radicals by increasing cellular defenses against oxidative damage, mainly through the induction of phase II detoxification enzymes by nuclear factor erythroid 2-related factor 2 (Nrf2). More and more studies have shown that the anticancer mechanism of SFN also includes induction of apoptotic pathway in tumor cells, inhibition of cell cycle progression, and suppression of tumor stem cells. Therefore, the application of SFN is expected to be a necessary new approach to treating cancer. In this paper, we review the multiple molecular mechanisms of SFN in cancer prevention and treatment in recent years, which can provide a new vision for cancer treatment.
Collapse
Affiliation(s)
- Pengtao Liu
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Bo Zhang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Yuanqiang Li
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, P. R. China.
| |
Collapse
|
4
|
Wang Q, Li D, Liu L, Shan Y, Bao Y. Dietary isothiocyanates and anticancer agents: exploring synergism for improved cancer management. Front Nutr 2024; 11:1386083. [PMID: 38919393 PMCID: PMC11196812 DOI: 10.3389/fnut.2024.1386083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
Human studies have shown the anticancer effects of dietary isothiocyanates (ITCs), but there are some inconsistencies, and more evidence supports that such anticancer effect is from higher doses of ITCs. The inconsistencies found in epidemiological studies may be due to many factors, including the biphasic dose-response (so called hormetic effect) of ITCs, which was found to be more profound under hypoxia conditions. In this comprehensive review, we aim to shed light on the intriguing synergistic interactions between dietary ITCs, focusing on sulforaphane (SFN) and various anticancer drugs. Our exploration is motivated by the potential of these combinations to enhance cancer management strategies. While the anticancer properties of ITCs have been recognized, our review delves deeper into understanding the mechanisms and emphasizing the significance of the hormetic effect of ITCs, characterized by lower doses stimulating both normal cells and cancer cells, whereas higher doses are toxic to cancer cells and inhibit their growth. We have examined a spectrum of studies unraveling the multifaceted interaction and combinational effects of ITCs with anticancer agents. Our analysis reveals the potential of these synergies to augment therapeutic efficacy, mitigate chemoresistance, and minimize toxic effects, thereby opening avenues for therapeutic innovation. The review will provide insights into the underlying mechanisms of action, for example, by spotlighting the pivotal role of Nrf2 and antioxidant enzymes in prevention. Finally, we glimpse ongoing research endeavors and contemplate future directions in this dynamic field. We believe that our work contributes valuable perspectives on nutrition and cancer and holds promise for developing novel and optimized therapeutic strategies.
Collapse
Affiliation(s)
- Qi Wang
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Dan Li
- Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, China
| | - Lihua Liu
- Department of Nutrition and Food Hygiene, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Yujuan Shan
- Department of Nutrition and Food Hygiene, School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Yongping Bao
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| |
Collapse
|
5
|
Baladia E, Moñino M, Pleguezuelos E, Russolillo G, Garnacho-Castaño MV. Broccoli Consumption and Risk of Cancer: An Updated Systematic Review and Meta-Analysis of Observational Studies. Nutrients 2024; 16:1583. [PMID: 38892516 PMCID: PMC11174709 DOI: 10.3390/nu16111583] [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: 04/24/2024] [Revised: 05/15/2024] [Accepted: 05/18/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND The scientific literature has reported an inverse association between broccoli consumption and the risk of suffering from several types of cancer; however, the results were not entirely consistent across studies. A systematic review and meta-analysis of observational studies were conducted to determine the association between broccoli consumption and cancer risk with the aim of clarifying the beneficial biological effects of broccoli consumption on cancer. METHODS PubMed/MEDLINE, Web of Science, Scopus, Cochrane Library (CENTRAL), and Epistemonikos databases were searched to identify all published papers that evaluate the impact of broccoli consumption on the risk of cancer. Citation chasing of included studies was conducted as a complementary search strategy. The risk of bias in individual studies was assessed using the Newcastle-Ottawa Scale. A random-effects model meta-analysis was employed to quantitatively synthesize results, with the I2 index used to assess heterogeneity. RESULTS Twenty-three case-control studies (n = 12,929 cases and 18,363 controls; n = 31,292 individuals) and 12 cohort studies (n = 699,482 individuals) were included in the meta-analysis. The results suggest an inverse association between broccoli consumption and the risk of cancer both in case-control studies (OR: 0.64, 95% CI from 0.58 to 0.70, p < 0.001; Q = 35.97, p = 0.072, I2 = 30.49%-moderate heterogeneity; τ2 = 0.016) and cohort studies (RR: 0.89, 95% CI from 0.82 to 0.96, p = 0.003; Q = 13.51, p = 0.333, I2 = 11.21%-low heterogeneity; τ2 = 0.002). Subgroup analysis suggested a potential benefit of broccoli consumption in site-specific cancers only in case-control studies. CONCLUSIONS In summary, the findings indicate that individuals suffering from some type of cancer consumed less broccoli, suggesting a protective biological effect of broccoli on cancer. More studies, especially cohort studies, are necessary to clarify the possible beneficial effect of broccoli on several types of cancer.
Collapse
Affiliation(s)
- Eduard Baladia
- Spanish Academy of Nutrition and Dietetics, 31006 Pamplona, Spain; (E.B.); (M.M.); (G.R.)
| | - Manuel Moñino
- Spanish Academy of Nutrition and Dietetics, 31006 Pamplona, Spain; (E.B.); (M.M.); (G.R.)
- Spanish Biomedical Research Center in Physiopathology of Obesity and Nutrition, Carlos III Health Institute, 28029 Madrid, Spain
| | - Eulogio Pleguezuelos
- Department of Physical Medicine and Rehabilitation, Mataró Hospital, Mataró, 08304 Barcelona, Spain;
| | - Giuseppe Russolillo
- Spanish Academy of Nutrition and Dietetics, 31006 Pamplona, Spain; (E.B.); (M.M.); (G.R.)
| | - Manuel Vicente Garnacho-Castaño
- DAFNiS Research Group, Pain, Physical Activity, Nutrition and Health, Campus Docent Sant Joan de Déu, Universitat de Barcelona, Sant Boi de Llobregat, 08830 Barcelona, Spain
- Facultad de Ciencias de la Salud, Universidad Internacional de Valencia (VIU), 46002 Valencia, Spain
| |
Collapse
|
6
|
Sailo BL, Liu L, Chauhan S, Girisa S, Hegde M, Liang L, Alqahtani MS, Abbas M, Sethi G, Kunnumakkara AB. Harnessing Sulforaphane Potential as a Chemosensitizing Agent: A Comprehensive Review. Cancers (Basel) 2024; 16:244. [PMID: 38254735 PMCID: PMC10814109 DOI: 10.3390/cancers16020244] [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: 10/18/2023] [Revised: 12/14/2023] [Accepted: 12/20/2023] [Indexed: 01/24/2024] Open
Abstract
Recent advances in oncological research have highlighted the potential of naturally derived compounds in cancer prevention and treatment. Notably, sulforaphane (SFN), an isothiocyanate derived from cruciferous vegetables including broccoli and cabbage, has exhibited potent chemosensitizing capabilities across diverse cancer types of bone, brain, breast, lung, skin, etc. Chemosensitization refers to the enhancement of cancer cell sensitivity to chemotherapy agents, counteracting the chemoresistance often developed by tumor cells. Mechanistically, SFN orchestrates this sensitization by modulating an array of cellular signaling pathways (e.g., Akt/mTOR, NF-κB, Wnt/β-catenin), and regulating the expression and activity of pivotal genes, proteins, and enzymes (e.g., p53, p21, survivin, Bcl-2, caspases). When combined with conventional chemotherapeutic agents, SFN synergistically inhibits cancer cell proliferation, invasion, migration, and metastasis while potentiating drug-induced apoptosis. This positions SFN as a potential adjunct in cancer therapy to augment the efficacy of standard treatments. Ongoing preclinical and clinical investigations aim to further delineate the therapeutic potential of SFN in oncology. This review illuminates the multifaceted role of this phytochemical, emphasizing its potential to enhance the therapeutic efficacy of anti-cancer agents, suggesting its prospective contributions to cancer chemosensitization and management.
Collapse
Affiliation(s)
- Bethsebie Lalduhsaki Sailo
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Le Liu
- Department of Gastroenterology, Shenzhen Hospital, Southern Medical University, Shenzhen 518001, China;
| | - Suravi Chauhan
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Sosmitha Girisa
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Mangala Hegde
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| | - Liping Liang
- Guangzhou Key Laboratory of Digestive Diseases, Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China;
| | - Mohammed S. Alqahtani
- Radiological Sciences Department, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia;
| | - Mohamed Abbas
- Electrical Engineering Department, College of Engineering, King Khalid University, Abha 61421, Saudi Arabia;
| | - Gautam Sethi
- Department of Pharmacology and NUS Centre for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Ajaikumar B. Kunnumakkara
- Cancer Biology Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, India; (B.L.S.); (S.C.); (S.G.); (M.H.)
| |
Collapse
|
7
|
Ajmeera D, Ajumeera R. Drug repurposing: A novel strategy to target cancer stem cells and therapeutic resistance. Genes Dis 2024; 11:148-175. [PMID: 37588226 PMCID: PMC10425757 DOI: 10.1016/j.gendis.2022.12.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 11/21/2022] [Accepted: 12/08/2022] [Indexed: 01/21/2023] Open
Abstract
Chemotherapy is an effortless and frequently used approach in cancer therapy. However, in most cases, it can only prolong life expectancy and does not guarantee a complete cure. Furthermore, chemotherapy is associated with severe adverse effects, one of the major complications of effective cancer therapy. In addition, newly published research outputs show that cancer stem cells are involved in cancer disease progression, drug resistance, metastasis, and recurrence and that they are functional in the trans-differentiation capacity of cancer stem cells to cancer cells in response to treatments. Novel strategies are therefore required for better management of cancer therapy. The prime approach would be to synthesize and develop novel drugs that need extensive resources, time, and endurance to be brought into therapeutic use. The subsequent approach would be to screen the anti-cancer activity of available non-cancerous drugs. This concept of repurposing non-cancer drugs as an alternative to current cancer therapy has become popular in recent years because using existing anticancer drugs has several adverse effects. Micronutrients have also been investigated for cancer therapy due to their significant anti-cancer effects with negligible or no side effects and availability in food sources. In this paper, we discuss an ideal hypothesis for screening available non-cancerous drugs with anticancer activity, with a focus on cancer stem cells and their clinical application for cancer treatment. Further, drug repurposing and the combination of micronutrients that can target both cancers and cancer stem cells may result in a better therapeutic approach leading to maximum tumor growth control.
Collapse
Affiliation(s)
- Divya Ajmeera
- Cell Biology Department, ICMR-National Institute of Nutrition (NIN), Hyderabad, Telangana 500007, India
| | - Rajanna Ajumeera
- Cell Biology Department, ICMR-National Institute of Nutrition (NIN), Hyderabad, Telangana 500007, India
| |
Collapse
|
8
|
Liao Y, Meng Q. Protection against cancer therapy-induced cardiovascular injury by planed-derived polyphenols and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 238:116896. [PMID: 37586453 DOI: 10.1016/j.envres.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Cancer therapy-induced heart injury is a significant concern for cancer patients undergoing chemotherapy, radiotherapy, immunotherapy, and also targeted molecular therapy. The use of these treatments can lead to oxidative stress and cardiomyocyte damage in the heart, which can result in heart failure and other cardiac complications. Experimental studies have revealed that chemotherapy drugs such as doxorubicin and cyclophosphamide can cause severe side effects such as cardiac fibrosis, electrophysiological remodeling, chronic oxidative stress and inflammation, etc., which may increase risk of cardiac disorders and attacks for patients that underwent chemotherapy. Similar consequences may also be observed for patients that undergo radiotherapy for left breast or lung malignancies. Polyphenols, a group of natural compounds with antioxidant and anti-inflammatory properties, have shown the potential in protecting against cancer therapy-induced heart injury. These compounds have been found to reduce oxidative stress, necrosis and apoptosis in the heart, thereby preserving cardiac function. In recent years, nanoparticles loaded with polyphenols have also provided for the delivery of these compounds and increasing their efficacy in different organs. These nanoparticles can improve the bioavailability and efficacy of polyphenols while minimizing their toxicity. This review article summarizes the current understanding of the protective effects of polyphenols and nanoparticles loaded with polyphenols against cancer therapy-induced heart injury. The article discusses the mechanisms by which polyphenols protect the heart, including antioxidant and anti-inflammation abilities. The article also highlights the potential benefits of using nanoparticles for the delivery of polyphenols.
Collapse
Affiliation(s)
- Yunshu Liao
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China
| | - Qinghua Meng
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
9
|
Wu S, Li L, Liang Q, Gao H, Tang T, Tang Y. A DFT study of sulforaphane adsorption on the group III nitrides (B12N12, Al12N12 and Ga12N12) nanocages. J Biomol Struct Dyn 2023:1-12. [PMID: 37882329 DOI: 10.1080/07391102.2023.2272755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 09/04/2023] [Indexed: 10/27/2023]
Abstract
In this paper, the adsorption behavior of group III nitrides (B12N12, Al12N12, and Ga12N12) nanocages to sulforaphane (SF) anticancer medicine were studied by density functional theory (DFT). The adsorption energy, solvation energy, desorption time and related quantum molecular descriptors were calculated in neutral and acidic solutions. When the drugs were adsorbed to nanocages, the structure of nanocages and drugs changed after adsorption, indicating that the process was effective adsorption. The adsorption energy and solvation energy of the complexes created after adsorption were negative values, which indicated that the structure of complexes formed by adsorption were stable. According to charge decomposition analysis (CDA) and natural bonding orbitals (NBO), drugs act as charge donors and nanocages act as charge acceptors, so that the charge flows from drugs to nanocages. Thermodynamic calculations demonstrate that drugs adsorption on nanocages is a spontaneous exothermic process. The calculation of quantum molecular descriptors confirmed that drugs adsorption on nanocages increased the chemical reactivity and solubility of drugs, which facilitated its transfer in biological fluids. Both interaction region index (IRI) and topological analysis of atom in molecule (AIM) revealed Van Der Waals interaction between drugs and nanocages. Protonation studies demonstrated that acidic circumstances could improve the polarity of complexes, increase the solvation effect, and boost drugs release in target cancer cells. The results of this work indicate that X12N12(X = B, Al, Ga) nanocages can be used as the delivery vehicle of SF drug.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- ShiQuan Wu
- School of Physics, Guizhou University, Guiyang, China
| | - Li Li
- School of Physics, Guizhou University, Guiyang, China
| | - QiQi Liang
- School of Physics, Guizhou University, Guiyang, China
| | - HuaXu Gao
- School of Physics, Guizhou University, Guiyang, China
| | - TianYu Tang
- School of Physics, Guizhou University, Guiyang, China
| | - YanLin Tang
- School of Physics, Guizhou University, Guiyang, China
| |
Collapse
|
10
|
Otoo RA, Allen AR. Sulforaphane's Multifaceted Potential: From Neuroprotection to Anticancer Action. Molecules 2023; 28:6902. [PMID: 37836745 PMCID: PMC10574530 DOI: 10.3390/molecules28196902] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 10/15/2023] Open
Abstract
Sulforaphane (SFN) is a naturally occurring compound found in cruciferous vegetables such as broccoli and cauliflower. It has been widely studied for its potential as a neuroprotective and anticancer agent. This review aims to critically evaluate the current evidence supporting the neuroprotective and anticancer effects of SFN and the potential mechanisms through which it exerts these effects. SFN has been shown to exert neuroprotective effects through the activation of the Nrf2 pathway, the modulation of neuroinflammation, and epigenetic mechanisms. In cancer treatment, SFN has demonstrated the ability to selectively induce cell death in cancer cells, inhibit histone deacetylase, and sensitize cancer cells to chemotherapy. SFN has also shown chemoprotective properties through inhibiting phase I metabolizing enzymes, modulating phase II xenobiotic-metabolizing enzymes, and targeting cancer stem cells. In addition to its potential as a therapeutic agent for neurological disorders and cancer treatment, SFN has shown promise as a potential treatment for cerebral ischemic injury and intracranial hemorrhage. Finally, the ongoing and completed clinical trials on SFN suggest potential therapeutic benefits, but more research is needed to establish its effectiveness. Overall, SFN holds significant promise as a natural compound with diverse therapeutic applications.
Collapse
Affiliation(s)
- Raymond A. Otoo
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA;
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
| | - Antiño R. Allen
- Division of Radiation Health, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA;
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
- Neurobiology & Developmental Sciences, University of Arkansas for Medical Sciences, 4301 West Markham, Suite 441B-2, Little Rock, AR 72205, USA
| |
Collapse
|
11
|
Milczarek M, Cierpiał T, Kiełbasiński P, Małecka-Giełdowska M, Świtalska M, Wietrzyk J, Mazur M, Wiktorska K. An Organofluorine Isoselenocyanate Analogue of Sulforaphane Affects Antimetabolite 5-Fluorouracil's Anticancer Activity: A Perspective for New Combinatory Therapy in Triple-Negative Breast Cancer. Molecules 2023; 28:5808. [PMID: 37570783 PMCID: PMC10420864 DOI: 10.3390/molecules28155808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/27/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Antimetabolites, especially 5-fluorouracil, are commonly used clinically to treat breast, colon, and other cancers. However, their side effects and inefficiency in monotherapy have prompted further searches for new combinations. Thus, the anticancer effect of 5-fluorouracil (5-FU) and the sulforaphane analogue, 4-isoselenocyanato-1-butyl 4'-fluorobenzyl sulfoxide (ISC), were tested in in vitro and in vivo models of triple-negative breast cancer (TNBC) as a new option for this treatment-resistant and aggressive type of breast cancer. A synergic interaction between 5-FU and ISC was observed in the TNBC in vitro model MDA-MB-231 cell line, which led to enhanced antiproliferative effects. The results of in vitro studies were confirmed by in vivo tests, which demonstrated stronger tumor growth inhibition and additive interactions between 5-FU and ISC in the murine TNBC model. Moreover, the results of the body mass and blood analysis showed the safety of the tested combination. The mechanistic study revealed that the combined treatment triggered apoptosis and necrosis, as well as inhibited cell migration.
Collapse
Affiliation(s)
- Małgorzata Milczarek
- Laboratory of Translation Research, Department of Biomedical Research, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
| | - Tomasz Cierpiał
- Division of Organic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Łódź, Poland; (T.C.); (P.K.)
| | - Piotr Kiełbasiński
- Division of Organic Chemistry, Centre of Molecular and Macromolecular Studies, Polish Academy of Sciences, Sienkiewicza 112, 90-363 Łódź, Poland; (T.C.); (P.K.)
| | - Milena Małecka-Giełdowska
- Department of Laboratory Medicine, Medical University of Warsaw, Stefana Banacha 1A, 02-097 Warsaw, Poland
- Central Laboratory, Central Teaching Hospital University Clinical Center, Medical University of Warsaw, Stefana Banacha 1A, 02-097 Warsaw, Poland
| | - Marta Świtalska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland; (M.Ś.); (J.W.)
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, 53-114 Wrocław, Poland; (M.Ś.); (J.W.)
| | - Maciej Mazur
- Faculty of Chemistry, University of Warsaw, Ludwika Pasteura 1, 02-093 Warsaw, Poland;
| | - Katarzyna Wiktorska
- Laboratory of Translation Research, Department of Biomedical Research, National Medicines Institute, Chełmska 30/34, 00-725 Warsaw, Poland
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences—SGGW, Nowoursynowska 166, 02-776 Warsaw, Poland
| |
Collapse
|
12
|
Boťanská B, Pecníková V, Fogarassyová M, Barančík M. The Role of Heat Shock Proteins and Autophagy in Mechanisms Underlying Effects of Sulforaphane on Doxorubicin-Induced Toxicity in HEK293 Cells. Physiol Res 2023; 72:S47-S59. [PMID: 37294118 DOI: 10.33549/physiolres.935107] [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: 06/23/2023] Open
Abstract
Doxorubicin (DOX) is a cytostatic agent belonging to anthracycline group. Important role in mechanism associated with negative effects of DOX plays an oxidative stress. Heat shock proteins (HSPs) are part of mechanisms initiated in response to stressful stimuli and play an important role in cellular responses to oxidative stress through interaction with components of redox signaling. The present work was aimed to study the role of HSPs and autophagy in mechanisms underlying effects of sulforaphane (SFN), a potential activator of Nrf-2, on doxorubicin-induced toxicity in human kidney HEK293 cells. We investigated effects of SFN and DOX on proteins associated with regulation of heat shock response, redox signaling, and autophagy. Results show that SFN significantly reduced cytotoxic effects of DOX. The positive effects of SFN on DOX-induced changes were associated with up-regulation of Nrf-2 and HSP60 protein levels. In the case of another heat shock protein HSP40, SFN increased its levels when was administered alone but not in conditions when cells were exposed to the effects of DOX. Sulforaphane also reversed negative effects of DOX on activities of superoxide dismutases (SODs) and up-regulation of autophagy markers (LC3A/B-II, Atg5, and Atg12). In conclusion, the changes observed in HSP60 are of particular importance in terms of protecting cells from the effects of DOX. Finding that under conditions where SFN reduced cytotoxic effects of DOX were significantly increased protein levels of both Nrf-2 and HSP60 point to the role of HSP60 in mechanisms of redox signaling underlying effects of SFN on DOX-induced toxicity in HEK293 cells. Moreover, data confirmed an important role of autophagy in effects of SFN on DOX-induced toxicity.
Collapse
Affiliation(s)
- B Boťanská
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | |
Collapse
|
13
|
Svolacchia F, Brongo S, Catalano A, Ceccarini A, Svolacchia L, Santarsiere A, Scieuzo C, Salvia R, Finelli F, Milella L, Saturnino C, Sinicropi MS, Fabrizio T, Giuzio F. Natural Products for the Prevention, Treatment and Progression of Breast Cancer. Cancers (Basel) 2023; 15:cancers15112981. [PMID: 37296944 DOI: 10.3390/cancers15112981] [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: 05/01/2023] [Revised: 05/23/2023] [Accepted: 05/26/2023] [Indexed: 06/12/2023] Open
Abstract
In this review, we summarize the most used natural products as useful adjuvants in BC by clarifying how these products may play a critical role in the prevention, treatment and progression of this disease. BC is the leading cancer, in terms of incidence, that affects women. The epidemiology and pathophysiology of BC were widely reported. Inflammation and cancer are known to influence each other in several tumors. In the case of BC, the inflammatory component precedes the development of the neoplasm through a slowly increasing and prolonged inflammation that also favors its growth. BC therapy involves a multidisciplinary approach comprising surgery, radiotherapy and chemotherapy. There are numerous observations that showed that the effects of some natural substances, which, in integration with the classic protocols, can be used not only for prevention or integration in order to prevent recurrences and induce a state of chemoquiescence but also as chemo- and radiosensitizers during classic therapy.
Collapse
Affiliation(s)
- Fabiano Svolacchia
- Department of Medical-Surgical Sciences and Biotechnologies, La Sapienza University, 00118 Rome, Italy
- Department of Medical Sciences, Policlinic Foundation Tor Vergata University, 00133 Rome, Italy
| | - Sergio Brongo
- Department of Plastic Surgery, University of Salerno, 84131 Campania, Italy
| | - Alessia Catalano
- Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", 70126 Bari, Italy
| | - Agostino Ceccarini
- U.O.C. Primary Care and Territorial Health, Social and Health Department, State Hospital, 47893 San Marino, San Marino
| | - Lorenzo Svolacchia
- Department of Medical-Surgical Sciences and Biotechnologies, La Sapienza University, 00118 Rome, Italy
| | - Alessandro Santarsiere
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- CNRS, UMR 7042-LIMA, ECPM, Université de Strasbourg, Université de Haute-Alsace, 67000 Strasbourg, France
| | - Carmen Scieuzo
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- Spinoff XFlies s.r.l., University of Basilicata, 85100 Potenza, Italy
| | - Rosanna Salvia
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- Spinoff XFlies s.r.l., University of Basilicata, 85100 Potenza, Italy
| | | | - Luigi Milella
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Carmela Saturnino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
| | - Maria Stefania Sinicropi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy
| | - Tommaso Fabrizio
- Department of Plastic Surgery, IRCCS, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture, Italy
| | - Federica Giuzio
- U.O.C. Primary Care and Territorial Health, Social and Health Department, State Hospital, 47893 San Marino, San Marino
- Department of Science, University of Basilicata, 85100 Potenza, Italy
- Spinoff TNcKILLERS s.r.l., University of Basilicata, 85100 Potenza, Italy
| |
Collapse
|
14
|
Shoaib S, Khan FB, Alsharif MA, Malik MS, Ahmed SA, Jamous YF, Uddin S, Tan CS, Ardianto C, Tufail S, Ming LC, Yusuf N, Islam N. Reviewing the Prospective Pharmacological Potential of Isothiocyanates in Fight against Female-Specific Cancers. Cancers (Basel) 2023; 15:cancers15082390. [PMID: 37190316 DOI: 10.3390/cancers15082390] [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: 11/23/2022] [Revised: 04/11/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Gynecological cancers are the most commonly diagnosed malignancies in females worldwide. Despite the advancement of diagnostic tools as well as the availability of various therapeutic interventions, the incidence and mortality of female-specific cancers is still a life-threatening issue, prevailing as one of the major health problems worldwide. Lately, alternative medicines have garnered immense attention as a therapeutic intervention against various types of cancers, seemingly because of their safety profiles and enhanced effectiveness. Isothiocyanates (ITCs), specifically sulforaphane, benzyl isothiocyanate, and phenethyl isothiocyanate, have shown an intriguing potential to actively contribute to cancer cell growth inhibition, apoptosis induction, epigenetic alterations, and modulation of autophagy and cancer stem cells in female-specific cancers. Additionally, it has been shown that ITCs plausibly enhance the chemo-sensitization of many chemotherapeutic drugs. To this end, evidence has shown enhanced efficacy in combinatorial regimens with conventional chemotherapeutic drugs and/or other phytochemicals. Reckoning with these, herein, we discuss the advances in the knowledge regarding the aspects highlighting the molecular intricacies of ITCs in female-specific cancers. In addition, we have also argued regarding the potential of ITCs either as solitary treatment or in a combinatorial therapeutic regimen for the prevention and/or treatment of female-specific cancers. Hopefully, this review will open new horizons for consideration of ITCs in therapeutic interventions that would undoubtedly improve the prognosis of the female-specific cancer clientele. Considering all these, it is reasonable to state that a better understanding of these molecular intricacies will plausibly provide a facile opportunity for treating these female-specific cancers.
Collapse
Affiliation(s)
- Shoaib Shoaib
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India
| | - Farheen Badrealam Khan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates
| | - Meshari A Alsharif
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - M Shaheer Malik
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Saleh A Ahmed
- Department of Chemistry, Faculty of Applied Sciences, Umm Al-Qura University, Makkah 21955, Saudi Arabia
- Department of Chemistry, Faculty of Applied Sciences, Assiut University, Assiut 71515, Egypt
| | - Yahya F Jamous
- Vaccines and Bioprocessing Center, King Abdulaziz City for Science and Technology (KACST), Riyadh 12354, Saudi Arabia
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
- Laboratory of Animal Center, Qatar University, Doha 2731, Qatar
| | - Ching Siang Tan
- School of Pharmacy, KPJ Healthcare University College, Nilai 71800, Malaysia
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Saba Tufail
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India
| | - Long Chiau Ming
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
- School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia
| | - Nabiha Yusuf
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Najmul Islam
- Department of Biochemistry, Jawaharlal Nehru Medical College, Aligarh Muslim University, Aligarh 202002, India
| |
Collapse
|
15
|
Pogorzelska A, Mazur M, Świtalska M, Wietrzyk J, Sigorski D, Fronczyk K, Wiktorska K. Anticancer effect and safety of doxorubicin and nutraceutical sulforaphane liposomal formulation in triple-negative breast cancer (TNBC) animal model. Biomed Pharmacother 2023; 161:114490. [PMID: 36931031 DOI: 10.1016/j.biopha.2023.114490] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/01/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Female breast cancer is the most deadly cancer in women worldwide. The triple-negative breast cancer subtype therapies, due to the lack of specific drug targets, are still based on systemic chemotherapy with doxorubicin, which is burdened with severe adverse effects. To enhance therapeutic success and protect against systemic toxicity, drug carriers or combination therapy are being developed. Thus, an innovative liposomal formulation containing doxorubicin and the main nutraceutical, sulforaphane, has been developed. The anticancer efficacy and safety of the proposed liposomal formulation was evaluated in vivo, in a 4T1 mouse model of triple-negative breast cancer, and the mechanism of action was determined in vitro, using triple-negative breast cancer MDA-MB-231 and non-tumorigenic breast MCF-10A cell line. The elaborated drug carriers were shown to efficiently deliver both compounds into the cancer cell and direct doxorubicin to the cell nucleus. Incorporation of sulforaphane resulted in a twofold inhibition of tumor growth and the potential of up to a fourfold reduction in doxorubicin concentration due to the synergistic interaction between the two compounds. Sulforaphane was shown to increase the accumulation of doxorubicin in the nuclei of cancer cells, accompanied by inhibition of mitosis, without affecting the reactive oxygen species status of the cell. In normal cells, an antagonistic effect resulting in less cytotoxicity was observed. In vivo results showed that sulforaphane incorporation yielded not only cardioprotective, but also nephro- and hepatoprotective effects. The results of the research revealed the prospects of applying sulforaphane as a component of liposomal doxorubicin in triple-negative breast cancer chemotherapy.
Collapse
Affiliation(s)
- Anna Pogorzelska
- Laboratory of Translation Research, Department of Biomedical Research, National Medicines Institute, Chełmska 30/34, Warsaw 00-725, Poland
| | - Maciej Mazur
- Faculty of Chemistry, University of Warsaw, Ludwika Pasteura 1, Warsaw 02-093, Poland
| | - Marta Świtalska
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, Wrocław 53-114, Poland
| | - Joanna Wietrzyk
- Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Rudolfa Weigla 12, Wrocław 53-114, Poland
| | - Dawid Sigorski
- Department of Oncology, Collegium Medicum, University of Warmia and Mazury, Al. Wojska Polskiego 37, Olsztyn 10-228, Poland; Department of Oncology and Immuno-Oncology, Warmian-Masurian Cancer Center of The Ministry of The Interior and Administration's Hospital, Al. Wojska Polskiego 37, Olsztyn 10-228, Poland
| | - Krzysztof Fronczyk
- Faculty of Psychology, University of Warsaw, Stawki 5/7, Warsaw 00-183, Poland
| | - Katarzyna Wiktorska
- Laboratory of Translation Research, Department of Biomedical Research, National Medicines Institute, Chełmska 30/34, Warsaw 00-725, Poland.
| |
Collapse
|
16
|
Coutinho LDL, Junior TCT, Rangel MC. Sulforaphane: An emergent anti-cancer stem cell agent. Front Oncol 2023; 13:1089115. [PMID: 36776295 PMCID: PMC9909961 DOI: 10.3389/fonc.2023.1089115] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 01/09/2023] [Indexed: 01/28/2023] Open
Abstract
Cancer is a major public health concern worldwide responsible for high morbidity and mortality rates. Alternative therapies have been extensively investigated, and plant-derived compounds have caught the attention of the scientific community due to their chemopreventive and anticancer effects. Sulforaphane (SFN) is one of these naturally occurring agents, and studies have shown that it is able to target a specific cancer cell population displaying stem-like properties, known as cancer stem cells (CSCs). These cells can self-renewal and differentiate to form highly heterogeneous tumor masses. Notably, most of the conventional chemotherapeutic agents cannot target CSCs once they usually exist in a quiescent state and overall, the available cytotoxic drugs focus on highly dividing cells. This is, at least in part, one of the reasons why some oncologic patients relapse after standard therapy. In this review we bring together studies supporting not only the chemopreventive and anticancer properties of SFN, but especially the emerging anti-CSCs effects of this natural product and its potential to be used with conventional antineoplastic drugs in the clinical setting.
Collapse
|
17
|
Cao W, Lu X, Zhong C, Wu J. Sulforaphane Suppresses MCF-7 Breast Cancer Cells Growth via miR-19/PTEN Axis to Antagonize the Effect of Butyl Benzyl Phthalate. Nutr Cancer 2023; 75:980-991. [PMID: 36542459 DOI: 10.1080/01635581.2022.2156555] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Sulforaphane (SFN), a major isothiocyanate found in cruciferous vegetables, reportedly exerts extensive antitumor effects. Butyl benzyl phthalate (BBP), a widely used plasticizer, plays a crucial role in the promotion of breast cancer. In the present study, we demonstrated that SFN inhibited proliferation, induced apoptosis, and suppressed the stemness of MCF-7 cells, whereas BBP exerted the opposite effects; microRNA-19 (miR-19) plays an important role in BBP-induced cell growth and dysregulation mediated via PTEN and p21. The growth-promoting effect of BBP could be mitigated by SFN, accompanied by a reversal of altered expression of miR-19a, miR-19b, PTEN, and p21. SFN also suppressed BBP-induced binding of upregulated miR-19 with PTEN, as determined using a dual-luciferase reporter assay. Collectively, these results demonstrated, for the first time, that SFN regulates the miR-19/PTEN axis to exert protective effects against BBP-mediated breast cancer promotion, suggesting a new potential role for SFN (or SFN-rich foods) in phthalate antagonism.
Collapse
Affiliation(s)
- Wanshuang Cao
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China.,Cancer Research Division, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiaomin Lu
- Department of Clinical Nutrition, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Caiyun Zhong
- Department of Nutrition and Food Hygiene, School of Public Health, Nanjing Medical University, Nanjing, China.,Cancer Research Division, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jieshu Wu
- Department of Maternal, Child and Adolescent Health, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
18
|
Krejbich P, Birringer M. The Self-Administered Use of Complementary and Alternative Medicine (CAM) Supplements and Antioxidants in Cancer Therapy and the Critical Role of Nrf-2-A Systematic Review. Antioxidants (Basel) 2022; 11:2149. [PMID: 36358521 PMCID: PMC9686580 DOI: 10.3390/antiox11112149] [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: 08/26/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 07/30/2023] Open
Abstract
Complementary and alternative medicine (CAM) supplements are widely used by cancer patients. Dietary supplements, vitamins and minerals, herbal remedies, and antioxidants are especially popular. In a systematic literature review, 37 studies, each including more than 1000 participants, on CAM, dietary supplement, and vitamin use among cancer patients were identified. Accordingly, cancer patients use antioxidants such as vitamin C (from 2.6% (United Kingdom) to 41.6% (United States)) and vitamin E (from 2.9% (China) to 48% (United States)). Dietary supplements and vitamins are taken for different reasons, but often during conventional cancer treatment involving chemotherapy or radiotherapy and in a self-decided manner without seeking medical advice from healthcare professionals. Drug-drug interactions with dietary supplements or vitamins involving multiple signaling pathways are well described. Since most of the anticancer drugs generate reactive oxygen species (ROS), an adaptive stress response of healthy and malignant cells, mainly driven by the Nrf-2-Keap I network, can be observed. On the one hand, healthy cells should be protected from ROS-overproducing chemotherapy and radiotherapy; on the other hand, ROS production in cancer cells is a "desirable side effect" during anticancer drug treatment. We here describe the paradoxical use of antioxidants and supplements during cancer therapy, possible interactions with anticancer drugs, and the involvement of the Nrf-2 transcription factor.
Collapse
Affiliation(s)
- Paula Krejbich
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Wissenschaftliches Zentrum für Ernährung, Lebensmittel und Nachhaltige Versorgungssysteme (ELVe), Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Public Health Zentrum Fulda, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
| | - Marc Birringer
- Department of Nutritional, Food and Consumer Sciences, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Wissenschaftliches Zentrum für Ernährung, Lebensmittel und Nachhaltige Versorgungssysteme (ELVe), Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
- Public Health Zentrum Fulda, Fulda University of Applied Sciences, Leipziger Straße 123, 36037 Fulda, Germany
| |
Collapse
|
19
|
Molecular Pathways Related to Sulforaphane as Adjuvant Treatment: A Nanomedicine Perspective in Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2022; 58:medicina58101377. [PMID: 36295538 PMCID: PMC9610969 DOI: 10.3390/medicina58101377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/23/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022]
Abstract
Because cancer is a multifactorial disease, it is difficult to identify the specific agents responsible for the disease's progression and development, but lifestyle and diet have been shown to play a significant role. Diverse natural compounds are demonstrating efficacy in the development of novel cancer therapies, including sulforaphane (1-isothiocyanate-4-(methylsulfinyl)butane), a compound found in broccoli and other cruciferous vegetables that promotes key biological processes such as apoptosis, cell cycle arrest, autophagy, and suppression of key signalling pathways such as the PI3K/AKT/mTOR pathway in breast cancer cells. However, one of the primary challenges with sulforaphane treatment is its low solubility in water and oral bioavailability. As a consequence, several investigations were conducted using this component complexed in nanoparticles, which resulted in superior outcomes when combined with chemotherapy drugs. In this study, we discuss the properties and benefits of sulforaphane in cancer therapy, as well as its ability to form complexes with nanomolecules and chemotherapeutic agents that synergize the antitumour response in breast cancer cells.
Collapse
|
20
|
Herbal Ingredients in the Prevention of Breast Cancer: Comprehensive Review of Potential Molecular Targets and Role of Natural Products. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6044640. [PMID: 36017236 PMCID: PMC9398845 DOI: 10.1155/2022/6044640] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/06/2022] [Accepted: 07/26/2022] [Indexed: 11/17/2022]
Abstract
Among various cancers, breast cancer is the most prevalent type in women throughout the world. Breast cancer treatment is challenging due to complex nature of the etiology of disease. Cell division cycle alterations are often encountered in a variety of cancer types including breast cancer. Common treatments include chemotherapy, surgery, radiotherapy, and hormonal therapy; however, adverse effects and multidrug resistance lead to complications and noncompliance. Accordingly, there is an increasing demand for natural products from medicinal plants and foods. This review summarizes molecular mechanisms of signaling pathways in breast cancer and identifies mechanisms by which natural compounds may exert their efficacy in the treatment of breast cancer.
Collapse
|
21
|
Gao Q, Feng J, Liu W, Wen C, Wu Y, Liao Q, Zou L, Sui X, Xie T, Zhang J, Hu Y. Opportunities and challenges for co-delivery nanomedicines based on combination of phytochemicals with chemotherapeutic drugs in cancer treatment. Adv Drug Deliv Rev 2022; 188:114445. [PMID: 35820601 DOI: 10.1016/j.addr.2022.114445] [Citation(s) in RCA: 78] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 02/08/2023]
Abstract
The therapeutic limitations such as insufficient efficacy, drug resistance, metastasis, and undesirable side effects are frequently caused by the long duration monotherapy based on chemotherapeutic drugs. multiple combinational anticancer strategies such as nucleic acids combined with chemotherapeutic agents, chemotherapeutic combinations, chemotherapy and tumor immunotherapy combinations have been embraced, holding great promise to counter these limitations, while still taking including some potential risks. Nowadays, an increasing number of research has manifested the anticancer effects of phytochemicals mediated by modulating cancer cellular events directly as well as the tumor microenvironment. Specifically, these natural compounds exhibited suppression of cancer cell proliferation, apoptosis, migration and invasion of cancer cells, P-glycoprotein inhibition, decreasing vascularization and activation of tumor immunosuppression. Due to the low toxicity and multiple modulation pathways of these phytochemicals, the combination of chemotherapeutic agents with natural compounds acts as a novel approach to cancer therapy to increase the efficiency of cancer treatments as well as reduce the adverse consequences. In order to achieve the maximized combination advantages of small-molecule chemotherapeutic drugs and natural compounds, a variety of functional nano-scaled drug delivery systems, such as liposomes, host-guest supramolecules, supramolecules, dendrimers, micelles and inorganic systems have been developed for dual/multiple drug co-delivery. These co-delivery nanomedicines can improve pharmacokinetic behavior, tumor accumulation capacity, and achieve tumor site-targeting delivery. In that way, the improved antitumor effects through multiple-target therapy and reduced side effects by decreasing dose can be implemented. Here, we present the synergistic anticancer outcomes and the related mechanisms of the combination of phytochemicals with small-molecule anticancer drugs. We also focus on illustrating the design concept, and action mechanisms of nanosystems with co-delivery of drugs to synergistically improve anticancer efficacy. In addition, the challenges and prospects of how these insights can be translated into clinical benefits are discussed.
Collapse
Affiliation(s)
- Quan Gao
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Jiao Feng
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wencheng Liu
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chengyong Wen
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Yihan Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qian Liao
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China
| | - Xinbing Sui
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Tian Xie
- School of Pharmacy and Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China.
| | - Jinming Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yichen Hu
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Chengdu University, No. 2025, Cheng Luo Road, Chengdu 610106, Sichuan, China.
| |
Collapse
|
22
|
Sharma M, Tollefsbol TO. Combinatorial epigenetic mechanisms of sulforaphane, genistein and sodium butyrate in breast cancer inhibition. Exp Cell Res 2022; 416:113160. [PMID: 35447103 DOI: 10.1016/j.yexcr.2022.113160] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 01/04/2023]
Abstract
Dietary phytochemicals are currently being studied with great interest due to their ability to regulate the epigenome resulting in prevention of cancer. Some natural botanicals have been reported to have enhanced and synergistic impact on cancer suppression when administered at optimum concentrations and in-conjunction. Sulforaphane (SFN) is an isothiocyanate found in cruciferous vegetables and sodium butyrate (NaB) is a short-chain fatty acid produced by gut microbiota. They have been intensively explored due to numerous anti-cancerous properties and ability to modulate epigenetic machinery by inhibition of histone deacetylase (HDAC). Genistein (GE), present in soy, is a known DNA methyltransferase (DNMT) inhibitor. While combined chemoprotective epigenetic effects induced by SFN and GE have been investigated, the key impact of combinatorial SFN-NaB, GE-NaB, and SFN-GE-NaB bioactive components in regulation of various mechanisms are poorly defined. In the present study, we found that combinations of dietary compounds had synergistic effects in decreasing cellular viability at lower dosages than their single dosages in breast cancer cell lines. The respective combinations limited growth and increased apoptosis and necrosis in cancerous cells among which the tri-combination displayed the most significant impact. Additionally, the respective combinations of compounds arrested MDA-MB-231 and MCF-7 breast cancer cells at G2/M phase. Our further mechanistic evaluation revealed that respective di-combinations and tri-combination had higher impact in down-regulation of DNMTs (DNMT3A and DNMT3B), HDACs (HDAC1, HDAC6 and HDAC11), histone methyltransferases (EZH2 and SUV39H1) and histone acetyltransferases (GCN5, PCAF, P300 and CBP) levels as compared to singly administered compounds. We also found that these combinations exhibited global epigenetic changes by inhibition of DNMT and HDAC activity, histone H3 at lysine 27 methylation (H3K27me) and histone H3 at lysine 9 methylation (H3K9me) levels, and by induction of histone acetyltransferases activity. Collectively, our investigation indicates that combined SFN, GE and NaB is highly effective in inhibiting breast cancer genesis by, at least in part, regulating epigenetic modifications, which may have implications in breast cancer therapy.
Collapse
Affiliation(s)
- Manvi Sharma
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States.
| | - Trygve O Tollefsbol
- Department of Biology, University of Alabama at Birmingham, Birmingham, AL, United States; O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States; Integrative Center for Aging Research, University of Alabama at Birmingham, Birmingham, AL, United States; Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States; Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States; University Wide Microbiome Center, University of Alabama at Birmingham, Birmingham, AL, United States.
| |
Collapse
|
23
|
Stern S, Liang D, Li L, Kurian R, Lynch C, Sakamuru S, Heyward S, Zhang J, Kareem KA, Chun YW, Huang R, Xia M, Hong CC, Xue F, Wang H. Targeting CAR and Nrf2 improves cyclophosphamide bioactivation while reducing doxorubicin-induced cardiotoxicity in triple-negative breast cancer treatment. JCI Insight 2022; 7:e153868. [PMID: 35579950 PMCID: PMC9309041 DOI: 10.1172/jci.insight.153868] [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: 08/04/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Cyclophosphamide (CPA) and doxorubicin (DOX) are key components of chemotherapy for triple-negative breast cancer (TNBC), although suboptimal outcomes are commonly associated with drug resistance and/or intolerable side effects. Through an approach combining high-throughput screening and chemical modification, we developed CN06 as a dual activator of the constitutive androstane receptor (CAR) and nuclear factor erythroid 2-related factor 2 (Nrf2). CN06 enhances CAR-induced bioactivation of CPA (a prodrug) by provoking hepatic expression of CYP2B6, while repressing DOX-induced cytotoxicity in cardiomyocytes in vitro via stimulating Nrf2-antioxidant signaling. Utilizing a multicellular coculture model incorporating human primary hepatocytes, TNBC cells, and cardiomyocytes, we show that CN06 increased CPA/DOX-mediated TNBC cell death via CAR-dependent CYP2B6 induction and subsequent conversion of CPA to its active metabolite 4-hydroxy-CPA, while protecting against DOX-induced cardiotoxicity by selectively activating Nrf2-antioxidant signaling in cardiomyocytes but not in TNBC cells. Furthermore, CN06 preserves the viability and function of human iPSC-derived cardiomyocytes by modulating antioxidant defenses, decreasing apoptosis, and enhancing the kinetics of contraction and relaxation. Collectively, our findings identify CAR and Nrf2 as potentially novel combined therapeutic targets whereby CN06 holds the potential to improve the efficacy/toxicity ratio of CPA/DOX-containing chemotherapy.
Collapse
Affiliation(s)
- Sydney Stern
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Dongdong Liang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Ritika Kurian
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Caitlin Lynch
- National Center for Advancing Translational Science (NCATS), NIH, Rockville, Maryland, USA
| | - Srilatha Sakamuru
- National Center for Advancing Translational Science (NCATS), NIH, Rockville, Maryland, USA
| | - Scott Heyward
- Bioreclamation In Vitro Technologies, Halethorpe, Maryland, USA
| | - Junran Zhang
- Department of Radiation Oncology, The Ohio State University James Comprehensive Cancer Center and College of Medicine, Columbus, Ohio, USA
| | - Kafayat Ajoke Kareem
- Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Young Wook Chun
- Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Ruili Huang
- National Center for Advancing Translational Science (NCATS), NIH, Rockville, Maryland, USA
| | - Menghang Xia
- National Center for Advancing Translational Science (NCATS), NIH, Rockville, Maryland, USA
| | - Charles C. Hong
- Division of Cardiovascular Medicine, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Fengtian Xue
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland, USA
| |
Collapse
|
24
|
Assessing Drug-Induced Mitochondrial Toxicity in Cardiomyocytes: Implications for Preclinical Cardiac Safety Evaluation. Pharmaceutics 2022; 14:pharmaceutics14071313. [PMID: 35890211 PMCID: PMC9319223 DOI: 10.3390/pharmaceutics14071313] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 02/07/2023] Open
Abstract
Drug-induced cardiotoxicity not only leads to the attrition of drugs during development, but also contributes to the high morbidity and mortality rates of cardiovascular diseases. Comprehensive testing for proarrhythmic risks of drugs has been applied in preclinical cardiac safety assessment for over 15 years. However, other mechanisms of cardiac toxicity have not received such attention. Of them, mitochondrial impairment is a common form of cardiotoxicity and is known to account for over half of cardiovascular adverse-event-related black box warnings imposed by the U.S. Food and Drug Administration. Although it has been studied in great depth, mitochondrial toxicity assessment has not yet been incorporated into routine safety tests for cardiotoxicity at the preclinical stage. This review discusses the main characteristics of mitochondria in cardiomyocytes, drug-induced mitochondrial toxicities, and high-throughput screening strategies for cardiomyocytes, as well as their proposed integration into preclinical safety pharmacology. We emphasize the advantages of using adult human primary cardiomyocytes for the evaluation of mitochondrial morphology and function, and the need for a novel cardiac safety testing platform integrating mitochondrial toxicity and proarrhythmic risk assessments in cardiac safety evaluation.
Collapse
|
25
|
Probable Mechanisms of Doxorubicin Antitumor Activity Enhancement by Ginsenoside Rh2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030628. [PMID: 35163891 PMCID: PMC8838402 DOI: 10.3390/molecules27030628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 11/19/2022]
Abstract
Ginsenoside Rh2 increases the efficacy of doxorubicin (DOX) treatment in murine models of solid and ascites Ehrlich’s adenocarcinoma. In a solid tumor model (treatment commencing 7 days after inoculation), DOX + Rh2 co-treatment was significantly more efficacious than DOX alone. If treatment was started 24 h after inoculation, the inhibition of tumor growth of a solid tumor for the DOX + Rh2 co-treatment group was complete. Furthermore, survival in the ascites model was dramatically higher for the DOX + Rh2 co-treatment group than for DOX alone. Mechanisms underlying the combined DOX and Rh2 effects were studied in primary Ehrlich’s adenocarcinoma-derived cells and healthy mice’s splenocytes. Despite the previously established Rh2 pro-oxidant activity, DOX + Rh2 co-treatment revealed no increase in ROS compared to DOX treatment alone. However, DOX + Rh2 treatment was more effective in suppressing Ehrlich adenocarcinoma cell adhesion than either treatment alone. We hypothesize that the benefits of DOX + Rh2 combination treatment are due to the suppression of tumor cell attachment/invasion that might be effective in preventing metastatic spread of tumor cells. Ginsenoside Rh2 was found to be a modest activator in a Neh2-luc reporter assay, suggesting that Rh2 can activate the Nrf2-driven antioxidant program. Rh2-induced direct activation of Nrf2 might provide additional benefits by minimizing DOX toxicity towards non-cancerous cells.
Collapse
|
26
|
Wang J, Yang J, Cao M, Zhao Z, Cao B, Yu S. The potential roles of Nrf2/Keap1 signaling in anticancer drug interactions. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100028. [PMID: 34909662 PMCID: PMC8663926 DOI: 10.1016/j.crphar.2021.100028] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 12/12/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2), together with its suppressive binding partner Kelch-like ECH-associated protein 1 (Keap1), regulates cellular antioxidant response and drug metabolism. The roles of Nrf2/Keap1 signaling in the pathology of many diseases have been extensively investigated, and small molecules targeting Nrf2/Keap1 signaling have been developed to prevent or treat diseases such as multiple sclerosis, chronic kidney disease and cancer. Notably, Nrf2 plays dual roles in cancer development and treatment. Activation of Nrf2/Keap1 signaling in cancer cells has been reported to promote cancer progression and result in therapy resistance. Since cancer patients are often suffering comorbidities of other chronic diseases, anticancer drugs could be co-administrated with other drugs and herbs. Nrf2/Keap1 signaling modulators, especially activators, are common in drugs, herbs and dietary ingredients, even they are developed for other targets. Therefore, drug-drug or herb-drug interactions due to modulation of Nrf2/Keap1 signaling should be considered in cancer therapies. Here we briefly summarize basic biochemistry and physiology functions of Nrf2/Keap1 signaling, Nrf2/Keap1 signaling modulators that cancer patients could be exposed to, and anticancer drugs that are sensitive to Nrf2/Keap1 signaling, aiming to call attention to the potential drug-drug or herb-drug interactions between anticancer drugs and these Nrf2/Keap1 signaling modulators.
Collapse
Affiliation(s)
- Jingya Wang
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, 100191, PR China
| | - Jin Yang
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, 100191, PR China
| | - Mingnan Cao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Zhigang Zhao
- Department of Pharmacy, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100050, China
| | - Baoshan Cao
- Department of Medical Oncology and Radiation Sickness, Peking University Third Hospital, Beijing, 100191, China
| | - Siwang Yu
- State Key Laboratory of Natural and Biomimetic Drugs; Department of Molecular and Cellular Pharmacology, Peking University School of Pharmaceutical Sciences, Beijing, 100191, PR China
| |
Collapse
|
27
|
Colapietro A, Rossetti A, Mancini A, Martellucci S, Ocone G, Pulcini F, Biordi L, Cristiano L, Mattei V, Delle Monache S, Marampon F, Gravina GL, Festuccia C. Multiple Antitumor Molecular Mechanisms Are Activated by a Fully Synthetic and Stabilized Pharmaceutical Product Delivering the Active Compound Sulforaphane (SFX-01) in Preclinical Model of Human Glioblastoma. Pharmaceuticals (Basel) 2021; 14:1082. [PMID: 34832864 PMCID: PMC8626029 DOI: 10.3390/ph14111082] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/23/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Frequent relapses and therapeutic resistance make the management of glioblastoma (GBM, grade IV glioma), extremely difficult. Therefore, it is necessary to develop new pharmacological compounds to be used as a single treatment or in combination with current therapies in order to improve their effectiveness and reduce cytotoxicity for non-tumor cells. SFX-01 is a fully synthetic and stabilized pharmaceutical product containing the α-cyclodextrin that delivers the active compound 1-isothiocyanato-4-methyl-sulfinylbutane (SFN) and maintains biological activities of SFN. In this study, we verified whether SFX-01 was active in GBM preclinical models. Our data demonstrate that SFX-01 reduced cell proliferation and increased cell death in GBM cell lines and patient-derived glioma initiating cells (GICs) with a stem cell phenotype. The antiproliferative effects of SFX-01 were associated with a reduction in the stemness of GICs and reversion of neural-to-mesenchymal trans-differentiation (PMT) closely related to epithelial-to-mesenchymal trans-differentiation (EMT) of epithelial tumors. Commonly, PMT reversion decreases the invasive capacity of tumor cells and increases the sensitivity to pharmacological and instrumental therapies. SFX-01 induced caspase-dependent apoptosis, through both mitochondrion-mediated intrinsic and death-receptor-associated extrinsic pathways. Here, we demonstrate the involvement of reactive oxygen species (ROS) through mediating the reduction in the activity of essential molecular pathways, such as PI3K/Akt/mTOR, ERK, and STAT-3. SFX-01 also reduced the in vivo tumor growth of subcutaneous xenografts and increased the disease-free survival (DFS) and overall survival (OS), when tested in orthotopic intracranial GBM models. These effects were associated with reduced expression of HIF1α which, in turn, down-regulates neo-angiogenesis. So, SFX-01 may have potent anti-glioma effects, regulating important aspects of the biology of this neoplasia, such as hypoxia, stemness, and EMT reversion, which are commonly activated in this neoplasia and are responsible for therapeutic resistance and glioma recurrence. SFX-01 deserves to be considered as an emerging anticancer agent for the treatment of GBM. The possible radio- and chemo sensitization potential of SFX-01 should also be evaluated in further preclinical and clinical studies.
Collapse
Affiliation(s)
- Alessandro Colapietro
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.R.); (A.M.); (G.O.); (G.L.G.)
| | - Alessandra Rossetti
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.R.); (A.M.); (G.O.); (G.L.G.)
| | - Andrea Mancini
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.R.); (A.M.); (G.O.); (G.L.G.)
| | - Stefano Martellucci
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, 02100 Rieti, Italy; (S.M.); (V.M.)
- Laboratory of Vascular Biology and Stem Cells, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.P.); (S.D.M.)
| | - Giuseppe Ocone
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.R.); (A.M.); (G.O.); (G.L.G.)
| | - Fanny Pulcini
- Laboratory of Vascular Biology and Stem Cells, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.P.); (S.D.M.)
| | - Leda Biordi
- Laboratory of Medical Oncology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Loredana Cristiano
- Department of Clinical Medicine, Public Health, Division of Human Anatomy, University of L’Aquila, 67100 L’Aquila, Italy;
| | - Vincenzo Mattei
- Biomedicine and Advanced Technologies Rieti Center, Sabina Universitas, 02100 Rieti, Italy; (S.M.); (V.M.)
| | - Simona Delle Monache
- Laboratory of Vascular Biology and Stem Cells, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (F.P.); (S.D.M.)
| | - Francesco Marampon
- Department of Radiological, Oncological and Pathological Sciences, La Sapienza University of Rome, 00185 Rome, Italy;
| | - Giovanni Luca Gravina
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.R.); (A.M.); (G.O.); (G.L.G.)
- Department of Biotechnological and Applied Clinical Sciences, Division of Radiotherapy, University of L’Aquila, 67100 L’Aquila, Italy
| | - Claudio Festuccia
- Laboratory of Radiobiology, Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy; (A.C.); (A.R.); (A.M.); (G.O.); (G.L.G.)
| |
Collapse
|
28
|
Abstract
Aim: The current investigation is focused on the targeted delivery of doxorubicin through CD44 aptamer-mediated active targeting to the human breast cancer cells. Methods: CD44 aptamer-doxorubicin (Apt-Dox) conjugates were developed by incubating different molar ratios of aptamer and doxorubicin. Cytotoxicity, selective intracellular accumulation and uptake of the Apt-Dox conjugates were analyzed to evaluate the efficacy of Apt-Dox conjugates. Results: Dox was efficiently conjugated with aptamer at 1:2 Apt-Dox molar ratios. Apt-Dox conjugate significantly inhibited the proliferation of CD44-overexpressing breast cancer cells, whereas negligible inhibition of cell proliferation was found in the control cells. Apt-Dox conjugate selectively internalized and accumulated in CD44-overexpressing cells. Conclusion: Apt-Dox conjugate selectively delivers doxorubicin to CD44-expressing cancer cells, thereby inhibiting selective cell proliferation and enhancing the targeted therapy.
Collapse
|
29
|
Haploinsufficiency Interactions between RALBP1 and p53 in ERBB2 and PyVT Models of Mouse Mammary Carcinogenesis. Cancers (Basel) 2021; 13:cancers13133329. [PMID: 34283045 PMCID: PMC8268413 DOI: 10.3390/cancers13133329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/22/2021] [Accepted: 06/25/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary Rlip knockout has been reported to prevent cancer in highly cancer-susceptible mice lacking p53, and Rlip knockdown kills many types of cancer cells. In humans, breast cancer shows diverse characteristics, including HER2-driven subtypes and viral-driven subtypes. HER2 can be targeted; however, escape of the cancer from targeted therapies remains a problem. In this work we evaluated the capacity of Rlip knockout to prevent breast cancer in genetically engineered mouse models of HER2-driven breast cancer (Erbb2 model) and polyomavirus-driven breast cancer (PyVT model). We found that in Erbb2 mice, Rlip knockout significantly delayed oncogenesis and reduced the expression of genes associated with poor prognosis in patients. In PyVT mice, Rlip knockout did not delay oncogenesis or tumor growth, but Rlip knockdown reduced tumor metastasis to the lung. We conclude that Rlip inhibitors may significantly improve survival in HER2-positive patients, but are unlikely to offer benefits to patients with polyomavirus-associated tumors. Abstract We recently reported that loss of one or both alleles of Ralbp1, which encodes the stress-protective protein RLIP76 (Rlip), exerts a strong dominant negative effect on both the inherent cancer susceptibility and the chemically inducible cancer susceptibility of mice lacking one or both alleles of the tumor suppressor p53. In this paper, we examined whether congenital Rlip deficiency could prevent genetically-driven breast cancer in two transgenic mouse models: the MMTV-PyVT model, which expresses the polyomavirus middle T antigen (PyVT) under control of the mouse mammary tumor virus promoter (MMTV) and the MMTV-Erbb2 model which expresses MMTV-driven erythroblastic leukemia viral oncogene homolog 2 (Erbb2, HER2/Neu) and frequently acquires p53 mutations. We found that loss of either one or two Rlip alleles had a suppressive effect on carcinogenesis in Erbb2 over-expressing mice. Interestingly, Rlip deficiency did not affect tumor growth but significantly reduced the lung metastatic burden of breast cancer in the viral PyVT model, which does not depend on either Ras or loss of p53. Furthermore, spontaneous tumors of MMTV-PyVT/Rlip+/+ mice showed no regression following Rlip knockdown. Finally, mice lacking one or both Rlip alleles differentially expressed markers for apoptotic signaling, proliferation, angiogenesis, and cell cycling in PyVT and Erbb2 breast tumors. Our results support the efficacy of Rlip depletion in suppressing p53 inactivated cancers, and our findings may yield novel methods for prevention or treatment of cancer in patients with HER2 mutations or tumor HER2 expression.
Collapse
|
30
|
Hou K, Shen J, Yan J, Zhai C, Zhang J, Pan JA, Zhang Y, Jiang Y, Wang Y, Lin RZ, Cong H, Gao S, Zong WX. Loss of TRIM21 alleviates cardiotoxicity by suppressing ferroptosis induced by the chemotherapeutic agent doxorubicin. EBioMedicine 2021; 69:103456. [PMID: 34233258 PMCID: PMC8261003 DOI: 10.1016/j.ebiom.2021.103456] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 06/09/2021] [Accepted: 06/10/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Doxorubicin, an anthracycline chemotherapeutic agent, is widely used in the treatment of many cancers. However, doxorubicin posts a great risk of adverse cardiovascular events, which are thought to be caused by oxidative stress. We recently reported that the ubiquitin E3 ligase TRIM21 interacts and ubiquitylates p62 and negatively regulates the p62-Keap1-Nrf2 antioxidant pathway. Therefore, we sought to determine the role TRIM21 in cardiotoxicity induced by oxidative damage. METHODS Using TRIM21 knockout mice, we examined the effects of TRIM21 on cardiotoxicity induced by two oxidative damage models: the doxorubicin treatment model and the Left Anterior Descending (LAD) model. We also explored the underlying mechanism by RNA-sequencing of the heart tissues, and by treating the mouse embryonic fibroblasts (MEFs), immortalized rat cardiomyocyte line H9c2, and immortalized human cardiomyocyte line AC16 with doxorubicin. FINDINGS TRIM21 knockout mice are protected from heart failure and fatality in both the doxorubicin and LAD models. Hearts of doxorubicin-treated wild-type mice exhibit deformed mitochondria and elevated level of lipid peroxidation reminiscent of ferroptosis, which is alleviated in TRIM21 knockout hearts. Mechanistically, TRIM21-deficient heart tissues and cultured MEFs and H9c2 cells display enhanced p62 sequestration of Keap1 and are protected from doxorubicin-induced ferroptosis. Reconstitution of wild-type but not the E3 ligase-dead and the p62 binding-deficient TRIM21 mutants impedes the protection from doxorubicin-induced cell death. INTERPRETATION Our study demonstrates that TRIM21 ablation protects doxorubicin-induced cardiotoxicity and illustrates a new function of TRIM21 in ferroptosis, and suggests TRIM21 as a therapeutic target for reducing chemotherapy-related cardiotoxicity. FUNDING NIH (CA129536; DK108989): data collection, analysis. Shanghai Pujiang Program (19PJ1401900): data collection. National Natural Science Foundation (31971161): data collection. Department of Veteran Affairs (BX004083): data collection. Tianjin Science and Technology Plan Project (17ZXMFSY00020): data collection.
Collapse
Affiliation(s)
- Kai Hou
- School of Medicine, Nankai University, Tianjin, China; Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China; Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jianliang Shen
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Junrong Yan
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Chuannan Zhai
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Jingxia Zhang
- Department of Cardiology, Tianjin Chest Hospital, Tianjin, China
| | - Ji-An Pan
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Ye Zhang
- Tianjin Third Central Hospital, Tianjin, China
| | - Yaping Jiang
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Yongbo Wang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Richard Z Lin
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Hongliang Cong
- School of Medicine, Nankai University, Tianjin, China; Department of Cardiology, Tianjin Chest Hospital, Tianjin, China.
| | - Shenglan Gao
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai, China.
| | - Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA.
| |
Collapse
|
31
|
Mangla B, Javed S, Sultan MH, Kumar P, Kohli K, Najmi A, Alhazmi HA, Al Bratty M, Ahsan W. Sulforaphane: A review of its therapeutic potentials, advances in its nanodelivery, recent patents, and clinical trials. Phytother Res 2021; 35:5440-5458. [PMID: 34184327 DOI: 10.1002/ptr.7176] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/27/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022]
Abstract
Traditionally, herbal supplements have shown an exceptional potential of desirability for the prevention of diseases and their treatment. Sulforaphane (SFN), an organosulfur compound belongs to the isothiocyanate (ITC) group and is mainly found naturally in cruciferous vegetables. Several studies have now revealed that SFN possesses broad spectrum of activities and has shown extraordinary potential as antioxidant, antitumor, anti-angiogenic, and anti-inflammatory agent. In addition, SFN is proven to be less toxic, non-oxidizable, and its administration to individuals is well tolerated, making it an effective natural dietary supplement for clinical trials. SFN has shown its ability to be a promising future drug molecule for the management of various diseases mainly due to its potent antioxidant properties. In recent times, several newer drug delivery systems were designed and developed for this potential molecule in order to enhance its bioavailability, stability, and to reduce its side effects. This review focuses to cover numerous data supporting the wide range of pharmacological activities of SFN, its drug-related issues, and approaches to improve its physicochemical and biological properties, including solubility, stability, and bioavailability. Recent patents and the ongoing clinical trials on SFN are also summarized.
Collapse
Affiliation(s)
- Bharti Mangla
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Shamama Javed
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Muhammad Hadi Sultan
- Department of Pharmaceutics, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Pankaj Kumar
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences & Research University (DPSRU), New Delhi, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Asim Najmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia.,Substance Abuse and Toxicology Research Centre, Jazan University, Jazan, Saudi Arabia
| | - Mohammed Al Bratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
32
|
Milczarek M, Pogorzelska A, Wiktorska K. Synergistic Interaction between 5-FU and an Analog of Sulforaphane-2-Oxohexyl Isothiocyanate-In an In Vitro Colon Cancer Model. Molecules 2021; 26:molecules26103019. [PMID: 34069385 PMCID: PMC8158758 DOI: 10.3390/molecules26103019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/14/2021] [Accepted: 05/16/2021] [Indexed: 12/22/2022] Open
Abstract
Combination therapy is based on the beneficial effects of pharmacodynamic interaction (synergistic or additive) between combined drugs or substances. A considerable group of candidates for combined treatments are natural compounds (e.g., isothiocyanates) and their analogs, which are tested in combination with anticancer drugs. We tested the anticancer effect of the combined treatment of isothiocyanate 2-oxohexyl isothiocyanate and 5-fluorouracil in colon and prostate cancer cell lines. The type of interaction was described using the Chou-Talalay method. The cytostatic and cytotoxic activities of the most promising combined treatments were investigated. In conclusion, we showed that combined treatment with 5-fluorouracil and 2-oxohexyl isothiocyanate acted synergistically in colon cancer. This activity is dependent on the cytostatic properties of the tested compounds and leads to the intensification of their individual cytotoxic activity. The apoptotic process is considered to be the main mechanism of cytotoxicity in this combined treatment.
Collapse
|
33
|
Food-Derived Pharmacological Modulators of the Nrf2/ARE Pathway: Their Role in the Treatment of Diseases. Molecules 2021; 26:molecules26041016. [PMID: 33671866 PMCID: PMC7918973 DOI: 10.3390/molecules26041016] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/08/2021] [Accepted: 02/12/2021] [Indexed: 12/28/2022] Open
Abstract
Oxidative stress, which refers to unbalanced accumulation of reactive oxygen species (ROS) levels in cells, has been linked to acute and chronic diseases. Nuclear factor erythroid 2-related factor 2/antioxidant response element (Nrf2/ARE) pathway plays a vital role in regulating cytoprotective genes and enzymes in response to oxidative stress. Therefore, pharmacological regulation of Nrf2/ARE pathway is an effective method to treat several diseases that are mainly characterized by oxidative stress and inflammation. Natural products that counteract oxidative stress by modulating Nrf2 have contributed significantly to disease treatment. In this review, we focus on bioactive compounds derived from food that are Nrf2/ARE pathway regulators and describe the molecular mechanisms for regulating Nrf2 to exert favorable effects in experimental models of diseases.
Collapse
|
34
|
Abstract
Adriamycin (ADR)-induced chronic heart injury (CHI) is a serious complication of chemotherapy. The present study was designed to assess the ability of fasudil, a Rho kinase inhibitor, to prevent ADR-induced CHI. Forty male 6-week-old C57BL6 mice were randomly divided into the following four groups: (1) control group, (2) CHI induced by adriamycin (ADR group), (3) CHI plus low dose fasudil (ADR + L group), and (4) CHI plus high dose fasudil (ADR + H group). Animals from groups 2-4 received ADR (2.5 mg/kg, i.p.) once a week for 8 weeks, and the control group received saline. Meanwhile, the animals in groups 3-4 received 2 mg/kg/day or 10 mg/kg/day fasudil, respectively. After measurement of cardiac functions, blood samples were collected for biochemical assays. The hearts were excised for histological, immunohistochemistry and western blot study, respectively. Adriamycin produced evident cardiac damage revealed by cardiac functions changes: decreased left ventricular fractional shortening (FS), left ventricular ejection fraction (EF), increased left ventricular volume, cardiac injury marker changes (increased creatine kinase, lactate dehydrogenase), antioxidant enzymes activity changes (decreased superoxide dismutase), and lipid peroxidation (elevated malondialdehyde) to the control group. Fasudil treatment notably ameliorated ADR-induced cardiac damage, restored heart function, suppressed cell apoptosis and senescence, ameliorated redox imbalance, and DNA damage. Fasudil has a protective effect on ADR-induced chronic heart injury, which partially attributed to its antioxidant, anti-apoptotic effects of inhibiting the RhoA/Rho kinase (ROCK) signaling pathway.
Collapse
|
35
|
Human Amnion Membrane Proteins Prevent Doxorubicin-Induced Oxidative Stress Injury and Apoptosis in Rat H9c2 Cardiomyocytes. Cardiovasc Toxicol 2021; 20:370-379. [PMID: 32086724 DOI: 10.1007/s12012-020-09564-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Doxorubicin (DOX) is widely used as an effective chemotherapy agent in cancer treatment. Cardiac toxicity in cancer treatment with DOX demand urgent attention and no effective treatment has been established for DOX-induced cardiomyopathy. It has been well documented that human amniotic membrane proteins (AMPs), extracted from amnion membrane (AM), have antioxidant, anti-apoptotic, and cytoprotective properties. Therefore, in this study, we aimed to investigate the protective effects of AMPs against cardiotoxicity induced by DOX in cultured rat cardiomyocyte cells (H9c2). DOX-induced cell injury was evaluated using multi-parametric assay including thiazolyl blue tetrazolium bromide (MTT), the release of lactic dehydrogenase (LDH), intracellular Ca2+ , reactive oxygen species (ROS) levels, cellular antioxidant status, mitochondrial membrane potential (ΔΨm), malondialdehyde (MDA), and NF-κB p65 DNA-binding activity. Moreover, expression profiling of apoptosis-related genes (P53, Bcl-2, and Bax) and Annexin V by flow cytometry were used for cell apoptosis detection. It was shown that AMPs pretreatment inhibited the cell toxicity induced by DOX. AMPs effectively attenuated the increased levels of LDH, Ca2+ , ROS, and MDA and also simultaneously elevated the ΔΨm and antioxidant status such as superoxide dismutase (SOD) and Catalase (CAT) in pretreated H9c2 cardiomyocytes. Besides, the activity of NF-kB p65 was reduced and the p53 and Bax protein levels were inhibited in these myocardial cells subjected to DOX. These findings provide the first evidence that AMPs potently suppressed DOX-induced toxicity in cardiomyocytes through inhibition of oxidative stress and apoptosis. Thus, AMPs can be a potential therapeutic agent against DOX cardiotoxicity.
Collapse
|
36
|
Tavakoli R, Tabeshpour J, Asili J, Shakeri A, Sahebkar A. Cardioprotective Effects of Natural Products via the Nrf2 Signaling Pathway. Curr Vasc Pharmacol 2020; 19:525-541. [PMID: 33155913 DOI: 10.2174/1570161119999201103191242] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 09/23/2020] [Accepted: 10/12/2020] [Indexed: 11/22/2022]
Abstract
Due to its poor regenerative capacity, the heart is specifically vulnerable to xenobiotic- induced cardiotoxicity, myocardial ischaemia/reperfusion injury and other pathologies. Nuclear factor erythroid-2-related factor 2 (Nrf2) is considered as an essential factor in protecting cardiomyocytes against oxidative stress resulting from free radicals and reactive oxygen species. It also serves as a key regulator of antioxidant enzyme expression via the antioxidant response element, a cis-regulatory element, which is found in the promoter region of several genes encoding detoxification enzymes and cytoprotective proteins. It has been reported that a variety of natural products are capable of activating Nrf2 expression, and in this way, increase the antioxidant potential of cardiomyocytes. In the present review, we consider the cardioprotective activities of natural products and their possible therapeutic potential.
Collapse
Affiliation(s)
- Rasool Tavakoli
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Jamshid Tabeshpour
- Faculty of Pharmacy, Damghan Bransh, Islamic Azad University, Damghan, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Shakeri
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
37
|
Bose C, Alves I, Singh P, Palade PT, Carvalho E, Børsheim E, Jun S, Cheema A, Boerma M, Awasthi S, Singh SP. Sulforaphane prevents age-associated cardiac and muscular dysfunction through Nrf2 signaling. Aging Cell 2020; 19:e13261. [PMID: 33067900 PMCID: PMC7681049 DOI: 10.1111/acel.13261] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 08/17/2020] [Accepted: 08/30/2020] [Indexed: 01/06/2023] Open
Abstract
Age-associated mitochondrial dysfunction and oxidative damage are primary causes for multiple health problems including sarcopenia and cardiovascular disease (CVD). Though the role of Nrf2, a transcription factor that regulates cytoprotective gene expression, in myopathy remains poorly defined, it has shown beneficial properties in both sarcopenia and CVD. Sulforaphane (SFN), a natural compound Nrf2-related activator of cytoprotective genes, provides protection in several disease states including CVD and is in various stages of clinical trials, from cancer prevention to reducing insulin resistance. This study aimed to determine whether SFN may prevent age-related loss of function in the heart and skeletal muscle. Cohorts of 2-month-old and 21- to 22-month-old mice were administered regular rodent diet or diet supplemented with SFN for 12 weeks. At the completion of the study, skeletal muscle and heart function, mitochondrial function, and Nrf2 activity were measured. Our studies revealed a significant drop in Nrf2 activity and mitochondrial functions, together with a loss of skeletal muscle and cardiac function in the old control mice compared to the younger age group. In the old mice, SFN restored Nrf2 activity, mitochondrial function, cardiac function, exercise capacity, glucose tolerance, and activation/differentiation of skeletal muscle satellite cells. Our results suggest that the age-associated decline in Nrf2 signaling activity and the associated mitochondrial dysfunction might be implicated in the development of age-related disease processes. Therefore, the restoration of Nrf2 activity and endogenous cytoprotective mechanisms by SFN may be a safe and effective strategy to protect against muscle and heart dysfunction due to aging.
Collapse
Affiliation(s)
- Chhanda Bose
- Division of Hematology & Oncology Department of Internal Medicine Texas Tech University Medical Sciences Center Lubbock TX USA
| | - Ines Alves
- Arkansas Children's Research Institute Little Rock AR USA
- Center for Neuroscience and Cell Biology University of Coimbra Coimbra Portugal
| | - Preeti Singh
- Department of Pharmacology and Toxicology University of Arkansas for Medical Sciences Little Rock AR USA
| | - Philip T. Palade
- Department of Pharmacology and Toxicology University of Arkansas for Medical Sciences Little Rock AR USA
| | - Eugenia Carvalho
- Arkansas Children's Research Institute Little Rock AR USA
- Center for Neuroscience and Cell Biology University of Coimbra Coimbra Portugal
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Elisabet Børsheim
- Arkansas Children's Research Institute Little Rock AR USA
- Department of Geriatrics University of Arkansas for Medical Sciences Little Rock AR USA
- Arkansas Children’s Nutrition Center Department of Pediatrics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Se‐Ran Jun
- Department of Biomedical Informatics University of Arkansas for Medical Sciences Little Rock AR USA
| | - Amrita Cheema
- Departments of Oncology and Biochemistry, Molecular and Cellular Biology Georgetown University Medical Center Washington DC USA
| | - Marjan Boerma
- Division of Radiation Health Department of Pharmaceutical Sciences University of Arkansas for Medical Sciences Little Rock AR USA
| | - Sanjay Awasthi
- Division of Hematology & Oncology Department of Internal Medicine Texas Tech University Medical Sciences Center Lubbock TX USA
| | - Sharda P. Singh
- Division of Hematology & Oncology Department of Internal Medicine Texas Tech University Medical Sciences Center Lubbock TX USA
| |
Collapse
|
38
|
Mangla B, Neupane YR, Singh A, Kumar P, Shafi S, Kohli K. Lipid-nanopotentiated combinatorial delivery of tamoxifen and sulforaphane: ex vivo, in vivo and toxicity studies. Nanomedicine (Lond) 2020; 15:2563-2583. [DOI: 10.2217/nnm-2020-0277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Aim: This study aims to load tamoxifen (TAM) and sulforaphane (SFN) into nanostructured lipid carriers (NLCs) to enhance their oral delivery. Materials & methods: TAM-SFN-NLCs were prepared using Precirol® ATO5 and Transcutol® HP, characterized and evaluated in vitro and ex vivo to assess the drug release profile and intestinal permeability, respectively. In vivo pharmacokinetic and acute toxicity assessment was performed in Wistar rats. Results: Optimized TAM-SFN-NLCs exhibited a particle size of 121.9 ± 6.42 nm and zeta potential of -21.2 ± 2.91 mV. The NLCs enhanced intestinal permeability of TAM and SFN and augmented oral bioavailability of TAM and SFN 5.2-fold and 4.8-fold, respectively. SFN significantly reduced TAM-associated toxicity in vivo. Conclusion: This coencapsulation of a chemotherapeutic agent with a herbal bioactive in NLCs could pave a novel treatment approach against cancer.
Collapse
Affiliation(s)
- Bharti Mangla
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Yub R Neupane
- Department of Pharmacy, National University of Singapore, 117559 Singapore
| | - Archu Singh
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Pankaj Kumar
- Department of Pharmaceutics, Delhi Pharmaceutical Sciences & Research University, New Delhi 110017, India
| | - Sadat Shafi
- Pharmaceutical Medicine, Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
39
|
Liu C, Ma X, Zhuang J, Liu L, Sun C. Cardiotoxicity of doxorubicin-based cancer treatment: What is the protective cognition that phytochemicals provide us? Pharmacol Res 2020; 160:105062. [DOI: 10.1016/j.phrs.2020.105062] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 12/11/2022]
|
40
|
Yarmohammadi F, Rezaee R, Karimi G. Natural compounds against doxorubicin-induced cardiotoxicity: A review on the involvement of Nrf2/ARE signaling pathway. Phytother Res 2020; 35:1163-1175. [PMID: 32985744 DOI: 10.1002/ptr.6882] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/01/2020] [Accepted: 08/28/2020] [Indexed: 12/12/2022]
Abstract
Cardiotoxicity is the main concern for long-term use of the doxorubicin (DOX). Reactive oxygen species (ROS) generation leads to oxidative stress that significantly contributes to the cardiac damage induced by DOX. The nuclear factor erythroid 2-related factor (Nrf2) acts as a protective player against DOX-induced myocardial oxidative stress. Several natural compounds (NCs) with anti-oxidative effects, were examined to suppress DOX cardiotoxicity such as asiatic acid, α-linolenic acid, apigenin, baicalein, β-lapachone, curdione, dioscin, ferulic acid, Ganoderma lucidum polysaccharides, genistein, ginsenoside Rg3, indole-3-carbinol, naringenin-7-O-glucoside, neferine, p-coumaric acid, pristimerin, punicalagin, quercetin, sulforaphane, and tanshinone IIA. The present article, reviews NCs that showed protective effects against DOX-induced cardiac injury through induction of Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.,Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Pharmaceutical Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
41
|
Jaganjac M, Milkovic L, Gegotek A, Cindric M, Zarkovic K, Skrzydlewska E, Zarkovic N. The relevance of pathophysiological alterations in redox signaling of 4-hydroxynonenal for pharmacological therapies of major stress-associated diseases. Free Radic Biol Med 2020; 157:128-153. [PMID: 31756524 DOI: 10.1016/j.freeradbiomed.2019.11.023] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/04/2019] [Accepted: 11/17/2019] [Indexed: 02/07/2023]
Abstract
Modern analytical methods combined with the modern concepts of redox signaling revealed 4-hydroxy-2-nonenal (4-HNE) as particular growth regulating factor involved in redox signaling under physiological and pathophysiological circumstances. In this review current knowledge of the relevance of 4-HNE as "the second messenger of reactive oxygen species" (ROS) in redox signaling of representative major stress-associated diseases is briefly summarized. The findings presented allow for 4-HNE to be considered not only as second messenger of ROS, but also as one of fundamental factors of the stress- and age-associated diseases. While standard, even modern concepts of molecular medicine and respective therapies in majority of these diseases target mostly the disease-specific symptoms. 4-HNE, especially its protein adducts, might appear to be the bioactive markers that would allow better monitoring of specific pathophysiological processes reflecting their complexity. Eventually that could help development of advanced integrative medicine approach for patients and the diseases they suffer from on the personalized basis implementing biomedical remedies that would optimize beneficial effects of ROS and 4-HNE to prevent the onset and progression of the illness, perhaps even providing the real cure.
Collapse
Affiliation(s)
- Morana Jaganjac
- Qatar Analytics & BioResearch Lab, Anti Doping Lab Qatar, Sport City Street, Doha, Qatar
| | - Lidija Milkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia
| | - Agnieszka Gegotek
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Marina Cindric
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Kamelija Zarkovic
- University of Zagreb, School of Medicine, Div. of Pathology, University Hospital Centre Zagreb, Kispaticeva 12, Zagreb, Croatia
| | - Elzbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Bialystok, Mickiewicza 2D, 15-222, Bialystok, Poland
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Div. of Molecular Medicine, Bijenicka 54, Zagreb, Croatia.
| |
Collapse
|
42
|
Mangla B, Alam O, Rub RA, Iqbal M, Singh A, Patel KS, Kohli K. Development and validation of a high throughput bioanalytical UPLC-MS/MS method for simultaneous determination of tamoxifen and sulphoraphane in rat plasma: Application to an oral pharmacokinetic study. J Chromatogr B Analyt Technol Biomed Life Sci 2020; 1152:122260. [PMID: 32679526 DOI: 10.1016/j.jchromb.2020.122260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 11/19/2022]
Abstract
Tamoxifen (TAM) is the choice of a drug approved by the Food and Drug Administration (FDA) for the treatment of estrogen-positive receptor (ER+) breast cancer. Sulphoraphane (SFN), a natural plant antioxidant compound, also acts on estrogen-positive breast cancer receptor. Thus, a combination of TAM with SFN is preferred as it helps to minimize the drug-related toxicity and increases the therapeutic efficacy by providing synergistic anticancer effects of both drugs. In the present study, a new simple, sensitive, precise, and selective UPLC-MS/MS method was developed for the simultaneous quantification of tamoxifen and sulphoraphane using propranolol as an internal standard (IS) in rat plasma. Chromatographic separation was achieved on reverse phase Acquity UPLC BEH C18 column (50 mm × 2.1 mm, i.d., 1.7 μm) with an isocratic mobile phase composed of solvent A (0.1% formic acid in acetonitrile) and B (0.1% formic acid in water) (80:20, v/v) at a flow-rate of 0.4 mL/min. The detection and quantification of analytes was performed on Waters ZsprayTM Xevo TQD using selected-ion monitoring operated under a positive electrospray ionization mode. The transitions were m/z = 372.0 [M+H]+ → 71.92 for tamoxifen, m/z = 177.9 [M+H]+ → 113.9 for sulphoraphane and m/z = 260.3 [M+H]+ → 116.1 for propranolol. The method was linear over the concentration range of 8-500 ng/mL (r2 = 0.9996) for tamoxifen, 30-2000 ng/mL (r2 = 0.9998) for sulphoraphane with insignificant matrix effect and high extraction recovery on spiked quality control (QC) samples. The intra- and inter-batch precisions and accuracy were within the acceptable limits, and both the analytes were found to be stable throughout the short term, long term and freeze thaw stability studies. The validated method was successfully applied for the simultaneous estimation of TAM and SFN in an oral pharmacokinetic study in female Wistar rats. This developed UPLC-MS/MS method could be a valuable tool for future pharmacokinetic interaction, therapeutic drug monitoring and pharmacokinetic characterization of novel formulations.
Collapse
Affiliation(s)
- Bharti Mangla
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ozair Alam
- Medicinal Chemistry and Molecular Modelling Lab, Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Abdur Rub
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia; Bioavailability Unit, Central Laboratory, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Archu Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Kuldeep Singh Patel
- Department of Pharmaceutical Chemistry, Amity Institute of Pharmacy, Amity University, Noida, Utter Pradesh 201303, India
| | - Kanchan Kohli
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
43
|
Khatoon E, Banik K, Harsha C, Sailo BL, Thakur KK, Khwairakpam AD, Vikkurthi R, Devi TB, Gupta SC, Kunnumakkara AB. Phytochemicals in cancer cell chemosensitization: Current knowledge and future perspectives. Semin Cancer Biol 2020; 80:306-339. [DOI: 10.1016/j.semcancer.2020.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
|
44
|
Kuran D, Pogorzelska A, Wiktorska K. Breast Cancer Prevention-Is there a Future for Sulforaphane and Its Analogs? Nutrients 2020; 12:nu12061559. [PMID: 32471217 PMCID: PMC7352481 DOI: 10.3390/nu12061559] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/30/2022] Open
Abstract
Breast cancer is the most prevalent type of cancer among women worldwide. There are several recommended methods of breast cancer prevention, including chemoprevention. There are several approved drugs used to prevent breast cancer occurrence or recurrence and metastasizing. There are also a number of new substances undergoing clinical trials and at the stage of initial study. Studies suggest that dietary factors play a crucial role in breast cancer etiology. Epidemiological studies indicate that in particular vegetables from the Brassicaceae family are a rich source of chemopreventive substances, with sulforaphane (SFN) being one of the most widely studied and characterized. This review discusses potential applicability of SFN in breast cancer chemoprevention. A comprehensive review of the literature on the impact of SFN on molecular signalling pathways in breast cancer and breast untransformed cells is presented. The presented results of in vitro and in vivo studies show that this molecule has a potential to act as a preventive molecule either to prevent disease development or recurrence and metastasizing, and as a compound protecting normal cells against the toxic effects of cytostatics. Finally, the still scanty attempts to develop an improved analog are also presented and discussed.
Collapse
Affiliation(s)
- Dominika Kuran
- Department of Pharmacology, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Anna Pogorzelska
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, 00-725 Warsaw, Poland;
| | - Katarzyna Wiktorska
- Department of Drug Biotechnology and Bioinformatics, National Medicines Institute, 00-725 Warsaw, Poland;
- OncoBoost Ltd., 02-089 Warsaw, Poland
- Correspondence:
| |
Collapse
|
45
|
Calcabrini C, Maffei F, Turrini E, Fimognari C. Sulforaphane Potentiates Anticancer Effects of Doxorubicin and Cisplatin and Mitigates Their Toxic Effects. Front Pharmacol 2020; 11:567. [PMID: 32425794 PMCID: PMC7207042 DOI: 10.3389/fphar.2020.00567] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
The success of cancer therapy is often compromised by the narrow therapeutic index of many anticancer drugs and the occurrence of drug resistance. The association of anticancer therapies with natural compounds is an emerging strategy to improve the pharmaco-toxicological profile of cancer chemotherapy. Sulforaphane, a phytochemical found in cruciferous vegetables, targets multiple pathways involved in cancer development, as recorded in different cancers such as breast, brain, blood, colon, lung, prostate, and so forth. As examples to make the potentialities of the association chemotherapy raise, here we highlight and critically analyze the information available for two associations, each composed by a paradigmatic anticancer drug (cisplatin or doxorubicin) and sulforaphane.
Collapse
Affiliation(s)
- Cinzia Calcabrini
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Francesca Maffei
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Eleonora Turrini
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| | - Carmela Fimognari
- Department for Life Quality Studies, Alma Mater Studiorum-Università di Bologna, Rimini, Italy
| |
Collapse
|
46
|
Liang X, Wang S, Wang L, Ceylan AF, Ren J, Zhang Y. Mitophagy inhibitor liensinine suppresses doxorubicin-induced cardiotoxicity through inhibition of Drp1-mediated maladaptive mitochondrial fission. Pharmacol Res 2020; 157:104846. [PMID: 32339784 DOI: 10.1016/j.phrs.2020.104846] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 03/17/2020] [Accepted: 04/16/2020] [Indexed: 01/04/2023]
Abstract
Doxorubicin (DOX) is one of the most effective antineoplastic drugs. However, its clinical application has been greatly limited due to the development of cardiotoxicity with DOX utilization. A number of theories have been postulated for DOX-induced cardiotoxicity with a pivotal contribution from unchecked (excess) mitophagy and mitochondrial fission. Liensinine (LIEN), a newly identified mitophagy inhibitor, strengthens the antineoplastic efficacy of DOX although its action on hearts remains elusive. This study was designed to examine the effect of LIEN on DOX-induced cardiotoxicity and the underlying mechanisms involved with a focus on mitochondrial dynamics. Our data revealed that LIEN alleviated DOX-induced cardiac dysfunction and apoptosis through inhibition of dynamin-related protein 1 (Drp1)-mediated excess (unchecked) mitochondrial fission. LIEN treatment decreased Drp1 phosphorylation at Ser616 site, inhibited mitochondrial fragmentation, mitophagy (assessed by TOM20 and TIM23), oxidative stress, cytochrome C leakage, cardiomyocyte apoptosis, as well as improved mitochondrial function and cardiomyocyte contractile function in DOX-induced cardiac injury. In DOX-challenged neonatal mouse ventricular myocytes (NMVMs), LIEN-suppressed Drp1 phosphorylation, mitochondrial fragmentation, and apoptosis were blunted by Rab7 overexpression, the effect of which was reversed by the ERK inhibitor U0126. Moreover, activation of ERK or Drp1 abolished the protective effects of LIEN on cardiomyocyte mechanical anomalies. These data shed some lights towards understanding the role of LIEN as a new protective agent against DOX-associated cardiotoxicity without compromising its anti-tumor effects.
Collapse
Affiliation(s)
- Xinyue Liang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Shuyi Wang
- Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Lifeng Wang
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA; Department of Physiology, Basic Medicine College, Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Asli F Ceylan
- Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; Center for Cardiovascular Research and Alternative Medicine, Laramie, WY 82071, USA.
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China.
| |
Collapse
|
47
|
Pore SK, Hahm ER, Kim SH, Singh KB, Nyiranshuti L, Latoche JD, Anderson CJ, Adamik J, Galson DL, Weiss KR, Watters RJ, Lee B, Kumta PN, Singh SV. A Novel Sulforaphane-Regulated Gene Network in Suppression of Breast Cancer-Induced Osteolytic Bone Resorption. Mol Cancer Ther 2020; 19:420-431. [PMID: 31784454 PMCID: PMC7007818 DOI: 10.1158/1535-7163.mct-19-0611] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 10/05/2019] [Accepted: 11/20/2019] [Indexed: 12/15/2022]
Abstract
Bone is the most preferred site for colonization of metastatic breast cancer cells for each subtype of the disease. The standard of therapeutic care for breast cancer patients with bone metastasis includes bisphosphonates (e.g., zoledronic acid), which have poor oral bioavailability, and a humanized antibody (denosumab). However, these therapies are palliative, and a subset of patients still develop new bone lesions and/or experience serious adverse effects. Therefore, a safe and orally bioavailable intervention for therapy of osteolytic bone resorption is still a clinically unmet need. This study demonstrates suppression of breast cancer-induced bone resorption by a small molecule (sulforaphane, SFN) that is safe clinically and orally bioavailable. In vitro osteoclast differentiation was inhibited in a dose-dependent manner upon addition of conditioned media from SFN-treated breast cancer cells representative of different subtypes. Targeted microarrays coupled with interrogation of The Cancer Genome Atlas data set revealed a novel SFN-regulated gene signature involving cross-regulation of runt-related transcription factor 2 (RUNX2) and nuclear factor-κB and their downstream effectors. Both RUNX2 and p65/p50 expression were higher in human breast cancer tissues compared with normal mammary tissues. RUNX2 was recruited at the promotor of NFKB1 Inhibition of osteoclast differentiation by SFN was augmented by doxycycline-inducible stable knockdown of RUNX2. Oral SFN administration significantly increased the percentage of bone volume/total volume of affected bones in the intracardiac MDA-MB-231-Luc model indicating in vivo suppression of osteolytic bone resorption by SFN. These results indicate that SFN is a novel inhibitor of breast cancer-induced osteolytic bone resorption in vitro and in vivo.
Collapse
Affiliation(s)
- Subrata K Pore
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Eun-Ryeong Hahm
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Su-Hyeong Kim
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Krishna B Singh
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Lea Nyiranshuti
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Joseph D Latoche
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Carolyn J Anderson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Radiology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Juraj Adamik
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Deborah L Galson
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Kurt R Weiss
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Rebecca J Watters
- Department of Orthopedic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Boeun Lee
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Prashant N Kumta
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Chemical and Petroleum Engineering and Department of Mechanical Engineering and Materials Science, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Shivendra V Singh
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
48
|
The Nrf-2/HO-1 Signaling Axis: A Ray of Hope in Cardiovascular Diseases. Cardiol Res Pract 2020; 2020:5695723. [PMID: 32411446 PMCID: PMC7204387 DOI: 10.1155/2020/5695723] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 12/28/2019] [Accepted: 01/07/2020] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular disease, which can lead to angina and shortness of breath, remains one of the most serious threats to human health. Owing to its imperceptible symptoms, it is difficult to determine the pathogenesis and treatment methods for cardiovascular disease. Nuclear factor erythropoietin-2-related factor 2/heme oxygenase 1 (Nrf2/HO-1) is a protein found in all cells of the human body. It is activated, transferred to the nucleus, and bound to DNA by antioxidant response elements (AREs). As a regulator of the antioxidant system, it upregulates the expression of HO-1 to reduce oxidative stress. Nrf2/HO-1 also has the ability to modulate calcium levels to prevent ferroptosis, pyroptosis, autophagy, programmed cell necrosis, alkaliptosis, and clockophagy. In view of the importance of Nrf2/HO-1 in the regulation of homeostasis, this review summarizes current research on the relationship between cardiovascular disease and Nrf2/HO-1. Normal cardiovascular diseases, such as viral myocarditis and myocardial ischemia-reperfusion injury, have been treated with Nrf2/HO-1. Rheumatic heart disease, cardiac tumors, arteriosclerosis, arrhythmia, hypertensive heart disease, and myocardial infarction have also been treated during experiments. Research has demonstrated the clinical application of Nrf2/HO-1 in pediatric cardiovascular disease; further clinical trials will help elucidate the potential of the Nrf2/HO-1 signaling axis.
Collapse
|
49
|
Jabbarzadeh Kaboli P, Afzalipour Khoshkbejari M, Mohammadi M, Abiri A, Mokhtarian R, Vazifemand R, Amanollahi S, Yazdi Sani S, Li M, Zhao Y, Wu X, Shen J, Cho CH, Xiao Z. Targets and mechanisms of sulforaphane derivatives obtained from cruciferous plants with special focus on breast cancer - contradictory effects and future perspectives. Biomed Pharmacother 2019; 121:109635. [PMID: 31739165 DOI: 10.1016/j.biopha.2019.109635] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/29/2019] [Accepted: 10/31/2019] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is the most common type of cancer among women. Therefore, discovery of new and effective drugs with fewer side effects is necessary to treat it. Sulforaphane (SFN) is an organosulfur compound obtained from cruciferous plants, such as broccoli and mustard, and it has the potential to treat breast cancer. Hence, it is vital to find out how SFN targets certain genes and cellular pathways in treating breast cancer. In this review, molecular targets and cellular pathways of SFN are described. Studies have shown SFN inhibits cell proliferation, causes apoptosis, stops cell cycle and has anti-oxidant activities. Increasing reactive oxygen species (ROS) produces oxidative stress, activates inflammatory transcription factors, and these result in inflammation leading to cancer. Increasing anti-oxidant potential of cells and discovering new targets to reduce ROS creation reduces oxidative stress and it eventually reduces cancer risks. In short, SFN effectively affects histone deacetylases involved in chromatin remodeling, gene expression, and Nrf2 anti-oxidant signaling. This review points to the potential of SFN to treat breast cancer as well as the importance of other new cruciferous compounds, derived from and isolated from mustard, to target Keap1 and Akt, two key regulators of cellular homeostasis.
Collapse
Affiliation(s)
- Parham Jabbarzadeh Kaboli
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China; Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia.
| | | | - Mahsa Mohammadi
- Department of Chemistry, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Ardavan Abiri
- Department of Medicinal Chemistry, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Roya Mokhtarian
- Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia
| | - Reza Vazifemand
- Laboratory of Virology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang, Selangor, 43400, Malaysia
| | - Shima Amanollahi
- Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia; School of Mathematical, Physical, and Natural Sciences, University of Florence, Firenze, 50134, Italy
| | - Shaghayegh Yazdi Sani
- Drug Discovery Research Group, Parham Academy of Biomedical Sciences, The Heritage B-16-10, Selangor, 43300, Malaysia
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Chi Hin Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, PR China; South Sichuan Institution for Translational Medicine, Luzhou, 646000, Sichuan, PR China.
| |
Collapse
|
50
|
Houghton CA. Sulforaphane: Its "Coming of Age" as a Clinically Relevant Nutraceutical in the Prevention and Treatment of Chronic Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2716870. [PMID: 31737167 PMCID: PMC6815645 DOI: 10.1155/2019/2716870] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/24/2019] [Accepted: 09/06/2019] [Indexed: 12/17/2022]
Abstract
A growing awareness of the mechanisms by which phytochemicals can influence upstream endogenous cellular defence processes has led to intensified research into their potential relevance in the prevention and treatment of disease. Pharmaceutical medicine has historically looked to plants as sources of the starting materials for drug development; however, the focus of nutraceutical medicine is to retain the plant bioactive in as close to its native state as possible. As a consequence, the potency of a nutraceutical concentrate or an extract may be lower than required for significant gene expression. The molecular structure of bioactive phytochemicals to a large extent determines the molecule's bioavailability. Polyphenols are abundant in dietary phytochemicals, and extensive in vitro research has established many of the signalling mechanisms involved in favourably modulating human biochemical pathways. Such pathways are associated with core processes such as redox modulation and immune modulation for infection control and for downregulating the synthesis of inflammatory cytokines. Although the relationship between oxidative stress and chronic disease continues to be affirmed, direct-acting antioxidants such as vitamins A, C, and E, beta-carotene, and others have not yielded the expected preventive or therapeutic responses, even though several large meta-analyses have sought to evaluate the potential benefit of such supplements. Because polyphenols exhibit poor bioavailability, few of their impressive in vitro findings have been replicated in vivo. SFN, an aliphatic isothiocyanate, emerges as a phytochemical with comparatively high bioavailability. A number of clinical trials have demonstrated its ability to produce favourable outcomes in conditions for which there are few satisfactory pharmaceutical solutions, foreshadowing the potential for SFN as a clinically relevant nutraceutical. Although myrosinase-inert broccoli sprout extracts are widely available, there now exist myrosinase-active broccoli sprout supplements that yield sufficient SFN to match the doses used in clinical trials.
Collapse
|