1
|
Xiao C, Yin X, Xi R, Yuan C, Ou Y. Molecular Mechanisms of Reversal of Multidrug Resistance in Breast Cancer by Inhibition of P-gp by Cytisine N-Isoflavones Derivatives Explored Through Network Pharmacology, Molecular Docking, and Molecular Dynamics. Int J Mol Sci 2025; 26:3813. [PMID: 40332431 PMCID: PMC12027943 DOI: 10.3390/ijms26083813] [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: 03/04/2025] [Revised: 04/03/2025] [Accepted: 04/11/2025] [Indexed: 05/08/2025] Open
Abstract
The compound CNI1, identified as a novel antitumor agent based on the cytisine N-isoflavones scaffold, and its series of cytisine N-isoflavones derivatives (CNI2, CNI3, and CNI4), were first isolated from bitter bean seeds, a traditional Chinese medicinal source, by our research team. Cellular activity assays combined with virtual screening targeting P-gp revealed that CNI1, along with the three cytisine N-isoflavones derivatives, CNI2, CNI3, and CNI4, exhibited significant multidrug resistance (MDR) reversal activity in breast cancer. Despite this promising outcome, the precise molecular mechanisms and key targets involved in the MDR reversal of these compounds remain to be elucidated. To explore potential mechanisms, targets for CNI1, CNII2, CNI3, and CNI4 (CNI1-4) were predicted using SwissTargetPrediction and Pharmmapper databases, while MDR-related targets in breast cancer were retrieved from OMIM and GeneCards. The overlapping targets were utilized to construct a protein-protein interaction (PPI) network to identify core targets. Additionally, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the DAVID database to identify relevant signaling pathways. Molecular docking simulations were employed to evaluate the binding sites and energies of CNI1-4 with the identified key targets, with the highest binding energy complexes selected for subsequent molecular dynamics simulations. This study identified 81 intersecting multidrug resistance (MDR) targets and 19 core targets in breast cancer. GO and KEGG pathway enrichment analyses revealed that MDR was primarily mediated by genes involved in cellular processes, apoptosis, protein phosphorylation, as well as the MAPK and PI3K-Akt signaling pathways. Molecular docking studies demonstrated that the binding energies of P-gp, AKT1, and SRC to CNI1-4 were all lower than -10 kcal/mol, indicating strong binding affinities. Molecular dynamics simulations further confirmed the stable and favorable binding interactions of CNI1-4 with AKT1 and P-gp. This study provides preliminary insights into the potential targets and molecular mechanisms of cytisine N-isoflavones compounds in reversing MDR in breast cancer, offering crucial data for the pharmacological investigation of CNI1-4 and supporting the development of P-gp inhibitors.
Collapse
Affiliation(s)
- Chuangchuang Xiao
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (C.X.); (R.X.); (C.Y.)
| | - Xiaoying Yin
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (C.X.); (R.X.); (C.Y.)
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China;
- Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Rui Xi
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (C.X.); (R.X.); (C.Y.)
| | - Chunping Yuan
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201620, China; (C.X.); (R.X.); (C.Y.)
- Shanghai Engineering Research Center for Pharmaceutical Intelligent Equipment, Shanghai University of Engineering Science, Shanghai 201620, China
| | - Yangsheng Ou
- Shanghai Frontiers Science Research Center for Druggability of Cardiovascular Noncoding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China;
| |
Collapse
|
2
|
Liu Y, Zhou Y, Chen P. Lung cancer organoids: models for preclinical research and precision medicine. Front Oncol 2023; 13:1293441. [PMID: 37941550 PMCID: PMC10628480 DOI: 10.3389/fonc.2023.1293441] [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: 09/13/2023] [Accepted: 09/27/2023] [Indexed: 11/10/2023] Open
Abstract
Lung cancer is a malignancy with high incidence and mortality rates globally, and it has a 5-year survival rate of only 10%-20%. The significant heterogeneity in clinical presentation, histological features, multi-omics findings, and drug sensitivity among different lung cancer patients necessitate the development of personalized treatment strategies. The current precision medicine for lung cancer, primarily based on pathological and genomic multi-omics testing, fails to meet the needs of patients with clinically refractory lung cancer. Lung cancer organoids (LCOs) are derived from tumor cells within tumor tissues and are generated through three-dimensional tissue culture, enabling them to faithfully recapitulate in vivo tumor characteristics and heterogeneity. The establishment of a series of LCOs biobanks offers promising platforms for efficient screening and identification of novel targets for anti-tumor drug discovery. Moreover, LCOs provide supplementary decision-making factors to enhance the current precision medicine for lung cancer, thereby addressing the limitations associated with pathology-guided approaches in managing refractory lung cancer. This article presents a comprehensive review on the construction methods and potential applications of LCOs in both preclinical and clinical research. It highlights the significance of LCOs in biomarker exploration, drug resistance investigation, target identification, clinical precision drug screening, as well as microfluidic technology-based high-throughput drug screening strategies. Additionally, it discusses the current limitations and future prospects of this field.
Collapse
Affiliation(s)
- Yajing Liu
- School of Pharmacy, Qingdao University, Qingdao, China
- Research and Development Department, NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao, China
| | - Yanbing Zhou
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Pu Chen
- Research and Development Department, NanoPeptide (Qingdao) Biotechnology Ltd., Qingdao, China
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
3
|
In Silico Study: Combination of α-Mangostin and Chitosan Conjugated with Trastuzumab against Human Epidermal Growth Factor Receptor 2. Polymers (Basel) 2022; 14:polym14132747. [PMID: 35808792 PMCID: PMC9268814 DOI: 10.3390/polym14132747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 12/10/2022] Open
Abstract
Breast cancer is a type of cancer with the highest prevalence worldwide. Almost 10–30% of breast cancer cases are diagnosed as positive for HER2 (human epidermal growth factor receptor 2). The currently available treatment methods still exhibit many shortcomings such as a high incidence of side effects and treatment failure due to resistance. This in silico study aims to simulate α-mangostin and chitosan combination conjugated to trastuzumab formulation against HER2 as an effort to improve breast cancer patient therapy. This molecular docking simulation was done through using PatchDock Server. The materials used including the two-dimensional structure of α-mangostin, chitosan, and sodium tripolyphosphate from the PubChem database; trastuzumab FASTA sequence from the DrugBank database; and HER2 structure obtained from a crystal complex with PDB ID: 1N8Z. The results indicated that the particle of α-mangostin and chitosan combinations interacted mostly with the crystallizable fragment (Fc region) of trastuzumab in the conjugation process. The conjugation of trastuzumab to the particle of a combination of α-mangostin and chitosan resulted in the greatest increase in the binding score of the smallest-sized particles (50 Å) with an increase in the score of 3828 and also gave the most similar mode of interaction with trastuzumab. However, the conjugation of trastuzumab eliminated the similarity of the mode of interaction and increased the value of atomic contact energy. Thus, a cominbation of α-mangostin and chitosan conjugated to a trastuzumab formulation was predicted can increase the effectiveness of breast cancer therapy at a relatively small particle size but with the consequence of decreasing atomic contact energy.
Collapse
|
4
|
Neumann-Mufweba A, Kimani S, Khan SF, Chibale K, Prince S. The diaryl-imidazopyridazine anti-plasmodial compound, MMV652103, exhibits anti-breast cancer activity. EXCLI JOURNAL 2022; 21:656-679. [PMID: 35651652 PMCID: PMC9149975 DOI: 10.17179/excli2021-4323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/24/2022] [Indexed: 11/12/2022]
Abstract
Breast cancer is the most common malignancy in women worldwide and it remains a global health burden, in part, due to poor response and tolerance to current therapeutics. Drug repurposing, which seeks to identify new indications for existing and investigational drugs, has become an exciting strategy to address these challenges. Here we describe the anti-breast cancer activity of a diaryl-imidazopyridazine compound, MMV652103, which was previously identified for its anti-plasmodial activity. We demonstrate that MMV652103 potently inhibits the oncogenic PI4KB and PIK3C2G lipid kinases, is selectively cytotoxic to MCF7 and T47D estrogen receptor positive breast cancer cells and inhibits their ability to survive and migrate. The underlying mechanisms involved included the induction of reactive oxygen species and activation of the DNA damage and p38 MAPK stress signaling pathways. This was associated with a G1 cell cycle arrest and an increase in levels of the cyclin-dependent kinase inhibitor p21 and activation of apoptotic and autophagic cell death pathways. Lastly, MMV652103 significantly reduced the weight and metastases of MCF7 induced tumors in an in vivo chick embryo model and displayed a favorable safety profile. These findings position MMV652103 as a promising chemotherapeutic in the treatment of oestrogen receptor positive breast cancers.
Collapse
Affiliation(s)
- Alexis Neumann-Mufweba
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| | - Serah Kimani
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| | - Saif Feroz Khan
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| | - Kelly Chibale
- Department of Chemistry, Faculty of Science, University of Cape Town, Rondebosch, 7701, Cape Town, South Africa
| | - Sharon Prince
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925, Cape Town, South Africa
| |
Collapse
|
5
|
Bhat IA, Kabeer SW, Reza MI, Mir RH, Dar MO. AdipoRon: A Novel Insulin Sensitizer in Various Complications and the Underlying Mechanisms: A Review. Curr Mol Pharmacol 2021; 13:94-107. [PMID: 31642417 DOI: 10.2174/1874467212666191022102800] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 09/26/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AdipoRon is the first synthetic analog of endogenous adiponectin, an adipose tissue-derived hormone. AdipoRon possesses pharmacological properties similar to adiponectin and its ability to bind and activate the adipoR1 and adipoR2 receptors makes it a suitable candidate for the treatment of a multitude of disorders. OBJECTIVE In the present review, an attempt was made to compile and discuss the efficacy of adipoRon against various disorders. RESULTS AdipoRon is a drug that acts not only in metabolic diseases but in other conditions unrelated to energy metabolism. It is well- reported that adipoRon exhibits strong anti-obesity, anti-diabetic, anticancer, anti-depressant, anti-ischemic, anti-hypertrophic properties and also improves conditions like post-traumatic stress disorder, anxiety, and systemic sclerosis. CONCLUSION A lot is known about its effects in experimental systems, but the translation of this knowledge to the clinic requires studies which, for many of the potential target conditions, have yet to be carried out. The beneficial effects of AdipoRon in novel clinical conditions will suggest an underlying pathophysiological role of adiponectin and its receptors in previously unsuspected settings.
Collapse
Affiliation(s)
- Ishfaq Ahmad Bhat
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Shaheen Wasil Kabeer
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Mohammad Irshad Reza
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, S.A.S. Nagar (Mohali), Punjab-160062, India
| | - Reyaz Hassan Mir
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, J&K, India
| | - Muhammad Ovais Dar
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| |
Collapse
|
6
|
Wathoni N, Meylina L, Rusdin A, Mohammed AFA, Tirtamie D, Herdiana Y, Motoyama K, Panatarani C, Joni IM, Lesmana R, Muchtaridi M. The Potential Cytotoxic Activity Enhancement of α-Mangostin in Chitosan-Kappa Carrageenan-Loaded Nanoparticle against MCF-7 Cell Line. Polymers (Basel) 2021; 13:polym13111681. [PMID: 34064093 PMCID: PMC8196802 DOI: 10.3390/polym13111681] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/10/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022] Open
Abstract
α-mangostin (αM), a xanthone derivative compound isolated from the extract of mangosteen pericarp (Garcinia mangostana L), has potential anticancer properties for breast cancer. However, it has poor solubility in water and low selectivity towards cancer cells. The polymeric nanoparticle formulation approach can be used to overcome these problems. In this study, a chitosan biopolymer-based αM polymeric nanoparticle formulation was encapsulated using kappa carrageenan (αM-Ch/Cr) as a novel carrier for breast cancer therapy and evaluated for their physicochemical properties, drug release profile, and in vitro cytotoxicity against breast cancer cells (MCF-7). Polymeric nanoparticles formulated with varying concentrations of kappa carrageenan were successfully prepared by ionic gelation and spray pyrolysis techniques. αM-Ch/Cr nanoparticles formed perfectly round particles with a size of 200–400 nm and entrapment efficiency ≥ 98%. In vitro release studies confirmed that αM-Ch/Cr nanoparticles had a sustained release system profile. Interestingly, the formulation of polymeric nanoparticles significantly (p < 0.05) increased the cytotoxicity of αM against MCF-7 cell with IC50 value of 4.7 μg/mL compared to the non-nanoparticle with IC50 of 8.2 μg/mL. These results indicate that αM-Ch/Cr nanoparticles have the potential to improve the physicochemical properties and cytotoxicity effects of αM compounds as breast cancer therapy agents.
Collapse
Affiliation(s)
- Nasrul Wathoni
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (L.M.); (A.R.); (D.T.); (Y.H.)
- Correspondence: ; Tel.: +62-22-842-888888 (ext. 3510)
| | - Lisna Meylina
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (L.M.); (A.R.); (D.T.); (Y.H.)
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Mulawarman, Samarinda 75119, Indonesia
| | - Agus Rusdin
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (L.M.); (A.R.); (D.T.); (Y.H.)
- Department of Pharmacy, Faculty of Sports and Health, Universitas Negeri Gorontalo, Gorontalo 96128, Indonesia
| | | | - Dorandani Tirtamie
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (L.M.); (A.R.); (D.T.); (Y.H.)
| | - Yedi Herdiana
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia; (L.M.); (A.R.); (D.T.); (Y.H.)
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto 862-0973, Japan;
| | - Camelia Panatarani
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang 45363, Indonesia; (C.P.); (I.M.J.)
- Functional Nano Powder University Center of Excellence, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - I Made Joni
- Department of Physics, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Sumedang 45363, Indonesia; (C.P.); (I.M.J.)
- Functional Nano Powder University Center of Excellence, Universitas Padjadjaran, Sumedang 45363, Indonesia
| | - Ronny Lesmana
- Department of Anatomy, Physiology and Biology Cell, Faculty of Medicine, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang 45363, Indonesia;
| |
Collapse
|
7
|
Sabit H, Abdel-Ghany S, Tombuloglu H, Cevik E, Alqosaibi A, Almulhim F, Al-Muhanaa A. New insights on CRISPR/Cas9-based therapy for breast Cancer. Genes Environ 2021; 43:15. [PMID: 33926574 PMCID: PMC8082964 DOI: 10.1186/s41021-021-00188-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 04/12/2021] [Indexed: 12/26/2022] Open
Abstract
CRISPR/Cas9 has revolutionized genome-editing techniques in various biological fields including human cancer research. Cancer is a multi-step process that encompasses the accumulation of mutations that result in the hallmark of the malignant state. The goal of cancer research is to identify these mutations and correlate them with the underlying tumorigenic process. Using CRISPR/Cas9 tool, specific mutations responsible for cancer initiation and/or progression could be corrected at least in animal models as a first step towards translational applications. In the present article, we review various novel strategies that employed CRISPR/Cas9 to treat breast cancer in both in vitro and in vivo systems.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, P. O. Box: 1982, Dammam, 31441, Saudi Arabia.
| | - Shaimaa Abdel-Ghany
- Department of Environmental Biotechnology, College of Biotechnology, Misr University for Science and Technology, P. O. Box 77, Giza, Egypt
| | - Huseyin Tombuloglu
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, P. O. Box: 1982, Dammam, 31441, Saudi Arabia
| | - Emre Cevik
- Department of Genetics, Institute for Medical Research and Consultations, Imam Abdulrahman Bin Faisal University, P. O. Box: 1982, Dammam, 31441, Saudi Arabia
| | - Amany Alqosaibi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, P. O. 4 Box, Dammam, 1982, Saudi Arabia
| | - Fatma Almulhim
- Breast Imaging Division, KFHU, Imam Abdulrahman Bin Faisal University, P. O. 4 Box, Dammam, 1982, Saudi Arabia
| | - Afnan Al-Muhanaa
- Breast Imaging Division, KFHU, Imam Abdulrahman Bin Faisal University, P. O. 4 Box, Dammam, 1982, Saudi Arabia
| |
Collapse
|
8
|
Lu H, Xie Y, Tran L, Lan J, Yang Y, Murugan NL, Wang R, Wang YJ, Semenza GL. Chemotherapy-induced S100A10 recruits KDM6A to facilitate OCT4-mediated breast cancer stemness. J Clin Invest 2021; 130:4607-4623. [PMID: 32427586 DOI: 10.1172/jci138577] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022] Open
Abstract
Breast cancer stem cells (BCSCs) play a critical role in cancer recurrence and metastasis. Chemotherapy induces BCSC specification through increased expression of pluripotency factors, but how their expression is regulated is not fully understood. Here, we delineate a pathway controlled by hypoxia-inducible factor 1 (HIF-1) that epigenetically activates pluripotency factor gene transcription in response to chemotherapy. Paclitaxel induces HIF-1-dependent expression of S100A10, which forms a complex with ANXA2 that interacts with histone chaperone SPT6 and histone demethylase KDM6A. S100A10, ANXA2, SPT6, and KDM6A are recruited to OCT4 binding sites and KDM6A erases H3K27me3 chromatin marks, facilitating transcription of genes encoding the pluripotency factors NANOG, SOX2, and KLF4, which along with OCT4 are responsible for BCSC specification. Silencing of S100A10, ANXA2, SPT6, or KDM6A expression blocks chemotherapy-induced enrichment of BCSCs, impairs tumor initiation, and increases time to tumor recurrence after chemotherapy is discontinued. Pharmacological inhibition of KDM6A also impairs chemotherapy-induced BCSC enrichment. These results suggest that targeting HIF-1/S100A10-dependent and KDM6A-mediated epigenetic activation of pluripotency factor gene expression in combination with chemotherapy may block BCSC enrichment and improve clinical outcome.
Collapse
Affiliation(s)
- Haiquan Lu
- Vascular Program, Institute for Cell Engineering.,Sidney Kimmel Comprehensive Cancer Center
| | | | - Linh Tran
- Vascular Program, Institute for Cell Engineering
| | - Jie Lan
- Vascular Program, Institute for Cell Engineering
| | - Yongkang Yang
- Vascular Program, Institute for Cell Engineering.,Sidney Kimmel Comprehensive Cancer Center
| | | | - Ru Wang
- Vascular Program, Institute for Cell Engineering
| | | | - Gregg L Semenza
- Vascular Program, Institute for Cell Engineering.,Sidney Kimmel Comprehensive Cancer Center.,Department of Genetic Medicine.,Department of Pediatrics.,Department of Medicine.,Department of Oncology.,Department of Radiation Oncology, and.,Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Gokce B, Akcok I, Cagir A, Pesen-Okvur D. A new drug testing platform based on 3D tri-culture in lab-on-a-chip devices. Eur J Pharm Sci 2020; 155:105542. [PMID: 32927074 DOI: 10.1016/j.ejps.2020.105542] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/10/2020] [Accepted: 09/02/2020] [Indexed: 01/13/2023]
Abstract
Drug discovery has a 90% rate of failure because preclinical platforms for drug testing do not mimic the in vivo conditions. Doxorubicin (DOX) is a commonly used drug to treat breast cancer patients even though it has side effects. Lab-on-a-chip (LOC) devices provide spatial control at the micrometer scale and can thus emulate the cancer microenvironment. Here, using a multidisciplinary approach, a new drug testing platform based on 3D tri-culture in LOC devices was developed. Breast cancer cells alone or with normal mammary epithelial cells and macrophages were cultured in matrigel in LOC devices. The platform was used to test DOX and (R)-4'-methylklavuzon (KLA), which is a new anti-cancer drug candidate. Results showed that DOX and KLA were equally effective on breast cancer cells in 3D monoculture. KLA produced 26% less death for breast cancer cells than DOX in 3D tri-culture. More importantly, DOX was not selective between breast cancer cells and normal mammary epithelial cells in 3D tri- culture whereas KLA caused 56% less cell death than DOX for normal mammary epithelial cells. Results strongly recommend that 3D tri-culture in LOC devices be used for assessment of drug toxicity at the preclinical stage.
Collapse
Affiliation(s)
- Begum Gokce
- Izmir Institute of Technology, Biotechnology and Bioengineering Graduate Program, Turkey
| | - Ismail Akcok
- Izmir Institute of Technology, Department of Chemistry, Turkey
| | - Ali Cagir
- Izmir Institute of Technology, Department of Chemistry, Turkey
| | - Devrim Pesen-Okvur
- Izmir Institute of Technology, Department of Molecular Biology and Genetics, Turkey.
| |
Collapse
|
10
|
Mardianingrum R, Yusuf M, Hariono M, Mohd Gazzali A, Muchtaridi M. α-Mangostin and its derivatives against estrogen receptor alpha. J Biomol Struct Dyn 2020; 40:2621-2634. [PMID: 33155528 DOI: 10.1080/07391102.2020.1841031] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Estrogen receptor alpha (ERα) acts as the transcription factor and the main therapeutic target against breast cancer. One of the compounds that has been shown to act as an ERα is α-mangostin. However, it still has weaknesses due to its low solubility and low potent activity. In this study, α-mangostin was modified by substituting -OH group at C6 using benzoyl derivatives through a step by step in silico study, namely pharmacokinetic prediction (https://preadmet.bmdrc.kr/adme/), pharmacophore modeling (LigandScout 4.1), molecular docking simulation (AutoDock 4.2), molecular dynamics simulation (AMBER 16) and a binding free energy analysis using MM-PBSA method. From the computational studies, three compounds which are derived from α-mangostin (AMB-1 (-9.84 kcal/mol), AMB-2 (-6.80 kcal/mol) and AMB-10 (-12.42 kcal/mol)) have lower binding free energy than α-mangostin (-1.77 kcal/mol), as evidenced by the binding free energy calculation using the MM-PBSA method. They can then be predicted to have potent activities as ERα antagonists.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Richa Mardianingrum
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia.,Department of Pharmacy, Universitas Perjuangan, Tasikmalaya, Indonesia
| | - Muhammad Yusuf
- Department of Chemistry, Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Maywan Hariono
- Faculty of Pharmacy, Universitas Sanata Dharma, Yogyakarta, Indonesia
| | - Amira Mohd Gazzali
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Pulau Pinang, Malaysia
| | - Muchtaridi Muchtaridi
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Jatinangor, Indonesia
| |
Collapse
|
11
|
Ozates NP, Soğutlu F, Lerminoglu F, Demir B, Gunduz C, Shademan B, Avci CB. Effects of rapamycin and AZD3463 combination on apoptosis, autophagy, and cell cycle for resistance control in breast cancer. Life Sci 2020; 264:118643. [PMID: 33141044 DOI: 10.1016/j.lfs.2020.118643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 10/15/2020] [Accepted: 10/20/2020] [Indexed: 01/20/2023]
Abstract
Breast cancer is the most common cancer in women and the leading cause of cancer mortality in women over 40 it's the year. The existence of the PI3K/AKT/mTOR pathway aberrations in more than 70% of breast cancer has caused to become a therapeutic target. AZD3463 is an anti-cancer agent used as a potential inhibitor of ALK/IGF1R. It also induces apoptosis and autophagy of the PI3K/AKT/mTOR pathway in cancer cells. Although the mTOR signaling might be inhibited by rapamycin treatment, signals transmitted from the upstream pathway supports cell survival and proliferation. The WST-1 assay test was performed to evaluate the anti-proliferative effects of rapamycin and AZD3463. Besides, the effects of them on apoptosis, autophagy, cytostatic, and metabolism in MCF7 breast cancer cells were investigated. Also, changes in the expression of apoptotic regulatory genes, cell cycle, and metabolism in the PI3K/AKT/mTOR Pathway were determined by Quantitative RT-PCR. The results showed that rapamycin and AZD3463 treatments significantly reduced survival in MCF7 cells. Also, apoptosis, autophagy, and cell population in the G0/G1 stage in the MCF7 cell category in the treatment group showed an increase compared to the control group. The combination of rapamycin and AZD3463 (AZD-RAPA) was determined as an additive according to isobologram analysis. In the combination of rapamycin with AZD3463, the expression of CDKN1B, PTEN, FOXO3, and APC genes increases, and the expression of PRKCB and PIK3CG genes decreases. Our results showed that the use of AZD-RAPA reduced the resistance of cancer cells to treatment and it leads cancer cells to apoptosis.
Collapse
Affiliation(s)
| | - Fatma Soğutlu
- Department of Medical Biology, Ege University Medicine Faculty, İzmir, Turkey
| | - Ferzan Lerminoglu
- Department of Toxicology, Ege University Pharmacy Faculty, Izmir, Turkey
| | - Busra Demir
- Department of Toxicology, Ege University Pharmacy Faculty, Izmir, Turkey
| | - Cumhur Gunduz
- Department of Medical Biology, Ege University Medicine Faculty, İzmir, Turkey
| | - Behrouz Shademan
- Department of Medical Biology, Ege University Medicine Faculty, İzmir, Turkey
| | - Cigir Biray Avci
- Department of Medical Biology, Ege University Medicine Faculty, İzmir, Turkey.
| |
Collapse
|
12
|
Soopramanien M, Khan NA, Abdalla SAO, Sagathevan K, Siddiqui R. Scorpion and Frog Organ Lysates are Potential Source of Antitumour Activity. Asian Pac J Cancer Prev 2020; 21:3011-3018. [PMID: 33112561 PMCID: PMC7798147 DOI: 10.31557/apjcp.2020.21.10.3011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 10/04/2020] [Indexed: 01/18/2023] Open
Abstract
OBJECTIVES It is noteworthy that several animal species are known to withstand high levels of radiation, and are exposed to heavy metals but rarely been reported to develop cancer. For example, the scorpion has been used as folk medicine in ancient civilizations of Iran and China, while amphibian skin is known to possess medicinal properties. Here, we elucidated the anti-tumour activity of the scorpion (Uropygi) and frog (Lithobates catesbeianus). MATERIALS AND METHODS Animals were procured and their organ lysates and sera were prepared and tested against Michigan Cancer Foundation-7 breast cancer (MCF-7), prostate cancer (PC3), Henrietta Lacks cervical cancer (HeLa), and normal human keratinocyte cells. Exoskeleton, appendages and hepatopancreas were dissected from the scorpion, whereas liver, lungs, heart, oviduct, gastrointestinal tract, gall bladder, kidneys, eggs and sera were collected from frog and organ lysates/sera were prepared. Growth inhibition assays and cytotoxicity assays were performed. RESULTS Appendages, exoskeleton lysates, and hepatopancreas from scorpion exhibited potent growth inhibition, and cytotoxic effects. Furthermore, lungs, liver, gastrointestinal tract, heart, oviduct, kidneys, eggs, and sera from frog displayed growth inhibition and cytotoxic effects. CONCLUSION Organ lysates, sera of scorpion, and amphibians possess anti-tumour activities. This is a worthy area of research as the molecular identity of the active molecule(s) together with their mechanism of action will lead to the rational development of novel anticancer agent(s).
Collapse
Affiliation(s)
| | - Naveed Ahmed Khan
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates.
| | | | - K Sagathevan
- Department of Biological Sciences, Sunway University, Bandar Sunway, Malaysia.
| | - Ruqaiyyah Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates.
| |
Collapse
|
13
|
Samuel SM, Varghese E, Koklesová L, Líšková A, Kubatka P, Büsselberg D. Counteracting Chemoresistance with Metformin in Breast Cancers: Targeting Cancer Stem Cells. Cancers (Basel) 2020; 12:E2482. [PMID: 32883003 PMCID: PMC7565921 DOI: 10.3390/cancers12092482] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/12/2022] Open
Abstract
Despite the leaps and bounds in achieving success in the management and treatment of breast cancers through surgery, chemotherapy, and radiotherapy, breast cancer remains the most frequently occurring cancer in women and the most common cause of cancer-related deaths among women. Systemic therapeutic approaches, such as chemotherapy, although beneficial in treating and curing breast cancer subjects with localized breast tumors, tend to fail in metastatic cases of the disease due to (a) an acquired resistance to the chemotherapeutic drug and (b) the development of intrinsic resistance to therapy. The existence of cancer stem cells (CSCs) plays a crucial role in both acquired and intrinsic chemoresistance. CSCs are less abundant than terminally differentiated cancer cells and confer chemoresistance through a unique altered metabolism and capability to evade the immune response system. Furthermore, CSCs possess active DNA repair systems, transporters that support multidrug resistance (MDR), advanced detoxification processes, and the ability to self-renew and differentiate into tumor progenitor cells, thereby supporting cancer invasion, metastasis, and recurrence/relapse. Hence, current research is focusing on targeting CSCs to overcome resistance and improve the efficacy of the treatment and management of breast cancer. Studies revealed that metformin (1, 1-dimethylbiguanide), a widely used anti-hyperglycemic agent, sensitizes tumor response to various chemotherapeutic drugs. Metformin selectively targets CSCs and improves the hypoxic microenvironment, suppresses the tumor metastasis and inflammation, as well as regulates the metabolic programming, induces apoptosis, and reverses epithelial-mesenchymal transition and MDR. Here, we discuss cancer (breast cancer) and chemoresistance, the molecular mechanisms of chemoresistance in breast cancers, and metformin as a chemo-sensitizing/re-sensitizing agent, with a particular focus on breast CSCs as a critical contributing factor to acquired and intrinsic chemoresistance. The review outlines the prospects and directions for a better understanding and re-purposing of metformin as an anti-cancer/chemo-sensitizing drug in the treatment of breast cancer. It intends to provide a rationale for the use of metformin as a combinatory therapy in a clinical setting.
Collapse
Affiliation(s)
- Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| | - Lenka Koklesová
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Alena Líšková
- Department of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia; (L.K.); (A.L.)
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar;
| |
Collapse
|
14
|
Rymbai E, Sugumar D, Saravanan J, Divakar S. Ropinirole, a potential drug for systematic repositioning based on side effect profile for management and treatment of breast cancer. Med Hypotheses 2020; 144:110156. [PMID: 32763725 DOI: 10.1016/j.mehy.2020.110156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/18/2020] [Accepted: 07/29/2020] [Indexed: 01/11/2023]
Abstract
Drug repositioning offers two main advantages in drug discovery - the process is less tedious and less costly. In the past, many drugs like thalidomide and sildenafil were successfully repositioned but the process was entirely serendipitous. These days drug repositioning is widely accepted as an alternate method of drug discovery and the process is based on building a strong hypothesis guided by systematic computational and experimental methods. One of the methods used in drug repositioning is based on shared side effects by drugs of different pharmacological categories. This method rests on the principle that drugs that share side effects might also share common biological targets and therefore same pharmacological indications. Old drugs can be repositioned for new uses by identifying the shared side effects of existing drugs and by modulating their chemical structure if required. Breast cancer is the most common type of cancer in women and the second leading cause of death worldwide after lung cancer in both men and women. Letrozole, an aromatase inhibitor, is used in the treatment of advanced, recurrent and metastatic breast cancer in post-menopausal women. Identification of drugs that share side effects with letrozole might help us to identify a potential drug for repositioning in the treatment of breast cancer. Ropinirole, a dopaminergic agonist was found to share the maximum number of side effects with letrozole. Studies have proposed that dopaminergic agonists induce apoptosis in breast, colon, ovarian cancer cells and leukemia neuroblastoma. This is consistent with our hypothesis that ropinirole that shares the maximum number of side effects with letrozole might be effective in the management of breast cancer. This hypothesis was further validated by preliminary molecular docking and in-vitro cell-line studies.
Collapse
Affiliation(s)
- Emdormi Rymbai
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - Deepa Sugumar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India
| | - J Saravanan
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India.
| | - S Divakar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, The Nilgiris, Tamil Nadu, India
| |
Collapse
|
15
|
Pistollato F, Bernasconi C, McCarthy J, Campia I, Desaintes C, Wittwehr C, Deceuninck P, Whelan M. Alzheimer's Disease, and Breast and Prostate Cancer Research: Translational Failures and the Importance to Monitor Outputs and Impact of Funded Research. Animals (Basel) 2020; 10:E1194. [PMID: 32674379 PMCID: PMC7401638 DOI: 10.3390/ani10071194] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 12/24/2022] Open
Abstract
Dementia and cancer are becoming increasingly prevalent in Western countries. In the last two decades, research focused on Alzheimer's disease (AD) and cancer, in particular, breast cancer (BC) and prostate cancer (PC), has been substantially funded both in Europe and worldwide. While scientific research outcomes have contributed to increase our understanding of the disease etiopathology, still the prevalence of these chronic degenerative conditions remains very high across the globe. By definition, no model is perfect. In particular, animal models of AD, BC, and PC have been and still are traditionally used in basic/fundamental, translational, and preclinical research to study human disease mechanisms, identify new therapeutic targets, and develop new drugs. However, animals do not adequately model some essential features of human disease; therefore, they are often unable to pave the way to the development of drugs effective in human patients. The rise of new technological tools and models in life science, and the increasing need for multidisciplinary approaches have encouraged many interdisciplinary research initiatives. With considerable funds being invested in biomedical research, it is becoming pivotal to define and apply indicators to monitor the contribution to innovation and impact of funded research. Here, we discuss some of the issues underlying translational failure in AD, BC, and PC research, and describe how indicators could be applied to retrospectively measure outputs and impact of funded biomedical research.
Collapse
Affiliation(s)
- Francesca Pistollato
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Camilla Bernasconi
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Janine McCarthy
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
- Physicians Committee for Responsible Medicine (PCRM), Washington, DC 20016, USA;
| | - Ivana Campia
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Christian Desaintes
- European Commission, Directorate General for Research and Innovation (RTD), 1000 Brussels, Belgium;
| | - Clemens Wittwehr
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Pierre Deceuninck
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| | - Maurice Whelan
- European Commission, Joint Research Centre (JRC), 21027 Ispra, Italy; (C.B.); (I.C.); (C.W.); (P.D.); (M.W.)
| |
Collapse
|
16
|
Shafi S, Khan S, Hoda F, Fayaz F, Singh A, Khan MA, Ali R, Pottoo FH, Tariq S, Najmi AK. Decoding Novel Mechanisms and Emerging Therapeutic Strategies in Breast Cancer Resistance. Curr Drug Metab 2020; 21:199-210. [DOI: 10.2174/1389200221666200303124946] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 12/12/2019] [Accepted: 12/30/2019] [Indexed: 12/24/2022]
Abstract
Breast cancer (BC), an intricate and highly heterogeneous disorder, has presently afflicted 2.09 million females globally. Chemoresistance remains a paramount challenge in the treatment of BC. Owing to its assorted nature, the chemoresistant mechanisms of BC still need intensive research. Accumulating evidence suggests that abnormalities related to the biogenesis of cancer stem cells (CSCs) and microRNAs (miRNAs) are associated with BC progression and chemoresistance. The presently available interventions are inadequate to target chemoresistance, therefore more efficient alternatives are urgently needed to improvise existing therapeutic regimens. A myriad of strategies is being explored, such as immunotherapy, gene therapy, and combination treatment to surmount chemoresistance. Additionally, nanoparticles as chemotherapeutic carriers put forward the options to encapsulate numerous drugs, alone as well as in combination for cancer theranostics. This review summarizes the chemoresistance mechanisms of miRNAs and CSCs as well as the most recently documented therapeutic approaches for the treatment of chemoresistance in BC. By unraveling the underpinning mechanism of BC chemoresistance, researchers could possibly develop more efficient treatment strategies towards BC.
Collapse
Affiliation(s)
- Sadat Shafi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Farazul Hoda
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Faizana Fayaz
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Archu Singh
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ruhi Ali
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Dammam 31441, Saudi Arabia
| | - Sana Tariq
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, MB Road, Pushp Vihar, New Delhi 110017, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| |
Collapse
|
17
|
Wu B, Yuan Y, Han X, Wang Q, Shang H, Liang X, Jing H, Cheng W. Structure of LINC00511-siRNA-conjugated nanobubbles and improvement of cisplatin sensitivity on triple negative breast cancer. FASEB J 2020; 34:9713-9726. [PMID: 32497336 DOI: 10.1096/fj.202000481r] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/14/2020] [Accepted: 05/15/2020] [Indexed: 01/07/2023]
Abstract
The drug resistance of triple negative breast cancer (TNBC) is considered as a major obstacle for the curative effect of chemotherapy. Long intergenic noncoding RNA 00511 (LINC00511) has been considered as a target gene of drug resistance. A novel theranostic agent loaded with LINC00511-siRNA to deliver siRNA was structured, and the responses of drug sensitivity in TNBC were detected. Next-generation high-throughput RNA sequencing (RNA-Seq) was performed to accurately analyze the differential expression of mRNAs and lncRNA targets after LINC00511-siRNA transfection with low-frequency ultrasound (LFUS). The LINC00511-siRNA conjugated nanobubble complexes showed appropriate characterization, with a mean diameter of 516.1 ± 24.7 nm and a zeta potential of -38.05 ± 0.24 mV. The transfection efficiency of nanobubble complexes was approximately 50% with LFUS. By RNA-Seq, the differential expressions of lncRNA transcripts and mRNA transcripts were identified, and then analyzed. The GO and KEGG enrichment analyses revealed the TNBC drug resistance related target genes and pathways. The combination of LFUS irradiation and nanobubble complexes is regarded as an efficient and safe method for siRNA transfection. The TNBC drug resistance occurs as a result of synergistic reactions between a variety of genes and a variety of pathways.
Collapse
Affiliation(s)
- Bolin Wu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China.,Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yanchi Yuan
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xue Han
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qiucheng Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haitao Shang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xitian Liang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hui Jing
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China.,Department of Interventional Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
18
|
Sinha S, Sharma S, Vora J, Shrivastava N. Emerging role of sirtuins in breast cancer metastasis and multidrug resistance: Implication for novel therapeutic strategies targeting sirtuins. Pharmacol Res 2020; 158:104880. [PMID: 32442721 DOI: 10.1016/j.phrs.2020.104880] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
Sirtuins (SIRTs), a class III histone deacetylases (HDACs) that require NAD+ as a cofactor and include SIRT1-7 proteins in mammals. Accumulative evidence has established that every sirtuin possesses exclusive and poised biology, implicating their role in the regulation of multifaceted biological functions leading to breast cancer initiation, progression, and metastasis. This article provides an outline of recent developments in the role of sirtuins in breast cancer metastasis and development of multidrug resistance (MDR). In addition, we have also highlighted the impending prospects of targeting SIRTs to overcome MDR to bring advancement in breast cancer management. Further, this review will focus on strategies for improving the activity and efficacy of existing cancer therapeutics by combining (adjuvant treatment/therapy) them with sirtuin inhibitors/modulators. All available as well as newly discovered synthetic and dietary sirtuin inhibitors, activators/modulators have been extensively reviewed and compiled to provide a rationale for targeting sirtuins. Further, we discuss their potential in developing future therapeutics against sirtuins proposing their use along with conventional chemotherapeutics to overcome the problem of breast cancer metastasis and MDR.
Collapse
Affiliation(s)
- Sonam Sinha
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Sonal Sharma
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India
| | - Jaykant Vora
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Neeta Shrivastava
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India.
| |
Collapse
|
19
|
Zhang X, Gu G, Song L, Wang D, Xu Y, Yang S, Xu B, Cao Z, Liu C, Zhao C, Zong Y, Qin Y, Xu J. ID4 Promotes Breast Cancer Chemotherapy Resistance via CBF1-MRP1 Pathway. J Cancer 2020; 11:3846-3857. [PMID: 32328189 PMCID: PMC7171490 DOI: 10.7150/jca.31988] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/27/2020] [Indexed: 12/21/2022] Open
Abstract
Chemo-resistance is considered a key problem in triple negative breast cancer (TNBC) chemotherapy and as such, an urgent need exists to identify its exact mechanisms. Inhibitor of DNA binding factor 4 (ID4) was reported to play diverse roles in different breast cancer molecular phenotypes. In addition, ID4 was associated with mammary carcinoma drug resistance however its functions and contributions remain insufficiently defined. The expression of ID4 in MCF-7, MCF-7/Adr and MDA-MB-231 breast cancer cell lines and patients' tissues were detected by RT-PCR, western blot and immunohistochemistry. Furthermore, TCGA database was applied to confirm these results. Edu and CCK8 assay were performed to detect the proliferation and drug resistance in breast cancer cell lines. Transwell and scratch migration assay were used to detected metastasis. Western blot, TCGA database, Immunoprecipitation (IP), Chromatin Immunoprecipitation (ChIP) and Luciferase reporter assay were used to investigate the tumor promotion mechanisms of ID4. In this study, we report that the expression levels of ID4 appeared to correlate with breast cancers subtype differentiation biomarkers (including ER, PR) and chemo-resistance related proteins (including MRP1, ABCG2, P-gp). Down-regulation of ID4 in MCF-7/Adr and MDA-MB-231 breast cancer cell lines significantly suppressed cell proliferation and invasion, however enhanced Adriamycin sensitivity. We further demonstrated that the oncogenic and chemo-resistant effects of ID4 could be mediated by binding to CBF1 promoter region though combination with MyoD1, and then the downstream target MRP1 could be activated. We reveal for the first time that ID4 performs its function via a CBF1-MRP1 signaling axis, and this finding provides a novel perspective to find potential therapeutic targets for breast cancer chemotherapy.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Guangyan Gu
- Department of Histology and Embryology, Shandong University Cheeloo College of Medicine, Jinan, 250012, Shandong, China
| | - Lin Song
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Dan Wang
- Department of Science and education, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Science and education, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Yali Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Shuping Yang
- Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Oncology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Bin Xu
- Department of Pathology, Shengli Oil Field Central Hospital, Dongying, Shandong Province, 257034, P.R China
| | - Zhixin Cao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Chunmei Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Chunming Zhao
- Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Ophthalmology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Yuanyuan Zong
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Yejun Qin
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| | - Jiawen Xu
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Shandong, 250021, PR China.,Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, 250021, PR China
| |
Collapse
|
20
|
Poofery J, Sripanidkulchai B, Banjerdpongchai R. Extracts of Bridelia ovata and Croton oblongifolius induce apoptosis in human MDA‑MB‑231 breast cancer cells via oxidative stress and mitochondrial pathways. Int J Oncol 2020; 56:969-985. [PMID: 32319560 DOI: 10.3892/ijo.2020.4973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 12/12/2019] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is the most common type of cancer and is also the second leading cause of cancer‑associated death in women worldwide. Thus, there is an urgent requirement for the development of effective treatments for this disease. Bridelia ovata and Croton oblongifolius are herbs used in Thai traditional medicine that have been used to treat various health problems; B. ovata has traditionally been used as a purgative, an antipyretic, a leukorrhea treatment and as a birth control herb. C. oblongifolius has been used to increase breast milk production, for post‑partum care (where it is used as a hot bath herb), and as a treatment for flat worms and dysmenorrhea. However, there is little research investigating the anticancer properties of these herbs. The present study aimed to investigate the anticancer properties of crude ethyl acetate extracts of B. ovata (BEA) and C. oblongifolius (CEA) in order to explore their underlying mechanisms in breast cancer cell death. The phytoconstituents of the crude extracts of BEA and CEA were studied using gas chromatography‑mass spectrometry (GC‑MS). GC‑MS analysis showed that the primary compound in BEA is friedelan‑3‑one, and kaur‑16‑en‑18‑oic acid in CEA. Cytotoxicity was investigated using an MTT assay, both BEA and CEA showed greater toxicity against MDA‑MB‑231 breast cancer cells compared with their effect on MCF10A normal epithelial mammary cells. BEA and CEA exerted various effects, including inducing apoptotic cell death, reducing mitochondrial transmembrane potential, increasing the levels of intracellular ROS, activating caspases, upregulating pro‑apoptotic and downregulating anti‑apoptotic genes and proteins. BEA and CEA were shown to have anticancer activity against breast cancer cells and induce apoptosis in these cells via a mitochondrial pathway and oxidative stress.
Collapse
Affiliation(s)
- Juthathip Poofery
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Bungorn Sripanidkulchai
- Center for Research and Development of Herbal Health Products, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ratana Banjerdpongchai
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
21
|
Al-Zharani M, Nasr FA, Abutaha N, Alqahtani AS, Noman OM, Mubarak M, Wadaan MA. Apoptotic Induction and Anti-Migratory Effects of Rhazya Stricta Fruit Extracts on a Human Breast Cancer Cell Line. Molecules 2019; 24:molecules24213968. [PMID: 31683960 PMCID: PMC6864471 DOI: 10.3390/molecules24213968] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/22/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022] Open
Abstract
Rhazya stricta is a medicinal plant that is widely used in Saudi folklore medicine for treatment of various diseases. R. stricta fruit powder was sequentially extracted with n-hexane, chloroform, ethyl acetate, and methanol using a Soxhlet extractor. The cytotoxic effects of these fractions on human breast cancer cells (MDA-MB-231 and MCF-7) and non-tumorigenic control cells (MCF-10A) were evaluated via cell viability measurements, microscopy, gene expression, and migration assays. Moreover, the effect of the most promising extract on 7,12-dimethyl-benz[a]anthracene (DMBA)-induced breast cancer was investigated in rats. The promising extract was also subjected to gas chromatography–mass spectrometry. Fruit extracts of R. stricta were significantly cytotoxic toward all tested cell lines, as demonstrated by MTT and LDH assays. Treatment of MDA-MB-231 cells with fruit ethyl acetate fraction (RSF EtOAc) increased expression 11of P53, Bax and activation of caspase 3/7. A cell migration scratch assay demonstrated that extracts at non-cytotoxic concentrations exerted a potent anti-migration activity against the highly invasive MDA-MB-231 cell line. Moreover, RT-PCR results showed that RSF EtOAc significantly downregulated MMP-2 and MMP-9 expression, which play an important role in breast cancer metastasis. Histological studies of breast tissue in experimental animals showed a slight improvement in tissue treated with fruit ethyl acetate extract. GC-MS chromatogram showed thirteen peaks with major constituents were camphor, trichosenic acid and guanidine. Our current study demonstrates that fruit extracts of R. stricta are cytotoxic toward breast cancer cell lines through apoptotic mechanisms.
Collapse
Affiliation(s)
- Mohammed Al-Zharani
- Imam Mohammad Ibn Saud Islamic University (IMSIU), College of Science, Biology Department, Riyadh 11623, Saudi Arabia.
| | - Fahd A Nasr
- Medicinal Aromatic, and Poisonous Plants Research Centre, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Nael Abutaha
- Bioproducts Research Chair, Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Ali S Alqahtani
- Medicinal Aromatic, and Poisonous Plants Research Centre, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Omar M Noman
- Medicinal Aromatic, and Poisonous Plants Research Centre, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Mohammed Mubarak
- Electron Microscope Unit, King Saud University Medical City, Riyadh 11451, Saudi Arabia.
| | - Muhammad A Wadaan
- Bioproducts Research Chair, Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
22
|
Al-malky HS, Al Harthi SE, Osman AMM. Major obstacles to doxorubicin therapy: Cardiotoxicity and drug resistance. J Oncol Pharm Pract 2019; 26:434-444. [DOI: 10.1177/1078155219877931] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BackgroundDoxorubicin is one of the most commonly prescribed and time-tested anticancer drugs. Although being considered as a first line drug in different types of cancers, the two main obstacles to doxorubicin therapy are drug-induced cardiotoxicity and drug resistance.MethodThe study utilizes systemic reviews on publications of previous studies obtained from scholarly journal databases including PubMed, Medline, Ebsco Host, Google Scholar, and Cochrane. The study utilizes secondary information obtained from health organizations using filters and keywords to sustain information relevancy. The study utilizes information retrieved from studies captured in the peer-reviewed journals on “doxorubicin-induced cardiotoxicity” and “doxorubicin resistance.”Discussion and resultsThe exact mechanisms of cardiotoxicity are not known; various hypotheses are studied. Doxorubicin can lead to free radical generation in various ways. The commonly proposed underlying mechanisms promoting doxorubicin resistance are the expression of multidrug resistance proteins as well as other causes.ConclusionIn this review, we have described the major obstacles to doxorubicin therapy, doxorubicin-induced cardiotoxicity as well as the mechanisms of cancer drug resistance and in following the treatment failures.
Collapse
Affiliation(s)
- Hamdan S Al-malky
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sameer E Al Harthi
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
23
|
Al-Malky HS, Osman AMM, Damanhouri ZA, Alkreathy HM, Al Aama JY, Ramadan WS, Al Qahtani AA, Al Mahdi HB. Modulation of doxorubicin-induced expression of the multidrug resistance gene in breast cancer cells by diltiazem and protection against cardiotoxicity in experimental animals. Cancer Cell Int 2019; 19:191. [PMID: 31367189 PMCID: PMC6657176 DOI: 10.1186/s12935-019-0912-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
Background Doxorubicin (DOX) is one of the most important anticancer agents used in treating breast cancer. However, chronic cardiotoxicity and multidrug resistance limit the chemotherapeutic use of DOX. Methods This study aimed to evaluate the capability of calcium channel blocker diltiazem (DIL) to reverse DOX resistance in breast cancer MCF-7 cells and to confer protection against DOX-induced cardiotoxicity in Wistar rats. For this purpose, we explored the effects of DOX on cell cycle phase distribution and expression of ABCB1, FOXO3a, and p53 genes in the presence and absence of DIL (20 μg/ml) and studied the ability of DIL to prevent DOX-induced cardiotoxicity after a single injection of DOX (15 mg/kg) in male Wister rats. Results We found that compared with DOX alone treatment, DIL + DOX treatment down regulated the ABCB1 gene expression by > fourfold but up regulated the FOXO3a and p53 genes expression by 1.5 fold. DIL treatment conferred protection against DOX-induced cardiotoxicity, as indicated by a decrease in the levels of the cardiac enzyme creatine kinase MB and malondialdehyde and an increase in the total antioxidant capacity and glutathione peroxidase levels. These biochemical results were further confirmed by the histopathological investigation of cardiac cells, which showed normal cardiac cells with central vesicular nuclei and prevention of DOX-induced disruption of normal cardiac architecture in the DIL to DOX group. Conclusions Taken together, our results indicate that DIL treatment can reverse the resistance of breast cancer cells to the therapeutic effects of DOX and can protect against DOX-induced cardiotoxicity in rats.
Collapse
Affiliation(s)
- Hamdan S Al-Malky
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Abdel-Moneim M Osman
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,2Pharmacology Unit, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Huda M Alkreathy
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Jumana Y Al Aama
- Department of Genetic Medicine, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,Princess Aljawhara Center of Excellence in Research of Hereditary Disorders, KAU, Jeddah, Saudi Arabia
| | - Wafaa S Ramadan
- Anatomy Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,6Anatomy Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ali A Al Qahtani
- Pharmacology Department, Faculty of Medicine, KAU, Jeddah, Saudi Arabia
| | - Hadiah B Al Mahdi
- Department of Genetic Medicine, Faculty of Medicine, KAU, Jeddah, Saudi Arabia.,Princess Aljawhara Center of Excellence in Research of Hereditary Disorders, KAU, Jeddah, Saudi Arabia
| |
Collapse
|
24
|
Fahad Ullah M. Breast Cancer: Current Perspectives on the Disease Status. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1152:51-64. [PMID: 31456179 DOI: 10.1007/978-3-030-20301-6_4] [Citation(s) in RCA: 338] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most frequently diagnosed cancer in women and ranks second among causes for cancer related death in women. Evidence in literature has shown that the past and ongoing research has an enormous implication in improving the clinical outcome in breast cancer. This has been attributed to the progress made in the realm of screening, diagnosis and therapeutic strategies engaged in breast cancer management. However, poor prognosis in TNBC and drug resistance presents major inhibitions which are also current challenges for containing the disease. Similarly, a focal point of concern is the rising rate of breast cancer incidence and mortality among the population of under developed world. In this chapter, an overview of the current practices for the diagnosis and treatment of breast cancer and associated impediments has been provided.
Collapse
Affiliation(s)
- Mohammad Fahad Ullah
- Prince Fahd Research Chair, Department of Medical Laboratory Technology, Faculty of Applied Medical Science, University of Tabuk, Tabuk-71491, Saudi Arabia.
| |
Collapse
|