1
|
Alghamdi MA, El Nashar EM, Elalfy M, Al-Zahrani NS, Alshehri MA, El-Nablaway M, Al-Khater KM, Aldahhan RA, El-Hadidy EG, Sleem F, Aljazzar A, Borlak J, Elhadidy M. Differentially expressed and alternately spliced genes as a novel tool for genotoxicity: a computerized study in ATT-myc transgenic mice for the recognition of genotoxic and non-genotoxic chemical. Front Genet 2025; 16:1505379. [PMID: 40225267 PMCID: PMC11986717 DOI: 10.3389/fgene.2025.1505379] [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: 10/02/2024] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Background Transgenic mice and gene expression in analyses were employed to evaluate hazardous chemicals. Methods Mice received weekly doses of NDEA (75 mg/kg) for six weeks and twice-weekly doses of BHT (300 mg/kg) for eight weeks. Gene expression and splicing alterations in the livers of six transgenic mice for each treatment of NDEA and BHT were examined using the MouseExon10ST array. Results Six hybridizations revealed 645 genes with significant expression changes, and 181 genes showed both expression and splicing alterations (p < 0.01). Furthermore, 2021 genes demonstrated significant exon-group interactions, indicating potential alternative splicing. Pathway analysis identified enriched groups in GOMolFn, GOProcess, GOCellLoc, and Pathway classes, with a higher representation of alternatively spliced and expressed genes (p < 0.01). Discussion Among the top expressed genes was TAT, which encodes the mitochondrial enzyme tyrosine aminotransferase, involved in tyrosine metabolism and recognized as a novel tumor suppressor gene linked to hepatocellular carcinoma (HCC). Additionally, HNF-4, a transcription factor, plays a crucial role in TAT expression. Conclusions This method can be used to identify genotoxic compounds in the att-myc model for short-term toxicity.
Collapse
Affiliation(s)
- Mansour A. Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Genomics and Personalized Medicine Unit, The Center for Medical and Health Research, King Khalid University, Abha, Saudi Arabia
| | - Eman M. El Nashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mahmoud Elalfy
- Clinical Science Department, College of Veterinary Medicine, King Faisal University, Al Hofuf, Saudi Arabia
- Forensic and Toxicology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Norah S. Al-Zahrani
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A. Alshehri
- Department of Child Health, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mohammad El-Nablaway
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh, Saudi Arabia
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Khulood M. Al-Khater
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Rashid A. Aldahhan
- Department of Anatomy, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Eman G. El-Hadidy
- Mathematics and Bioinformatics Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Fathy Sleem
- Forensic and Toxicology Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Ahmed Aljazzar
- Pathology Department, College of Veterinary medicine, King Faisal University, Al Hofuf, Saudi Arabia
| | - Jürgen Borlak
- Centre for Pharmacology and Toxicology, Hannover, Germany
| | - Mona Elhadidy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Medical Physiology, Faculty of Medicine, Al-Baha University, Al Baha, Saudi Arabia
| |
Collapse
|
2
|
Almawash S. Oral Bioavailability Enhancement of Anti-Cancer Drugs Through Lipid Polymer Hybrid Nanoparticles. Pharmaceutics 2025; 17:381. [PMID: 40143044 PMCID: PMC11946161 DOI: 10.3390/pharmaceutics17030381] [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: 10/16/2024] [Revised: 02/13/2025] [Accepted: 03/07/2025] [Indexed: 03/28/2025] Open
Abstract
Cancer is considered as the second leading cause of death worldwide. Chemotherapy, radiotherapy, immunotherapy, and targeted drug delivery are the main treatment options for treating cancers. Chemotherapy drugs are either available for oral or parenteral use. Oral chemotherapy, also known as chemotherapy at home, is more likely to improve patient compliance and convenience. Oral anti-cancer drugs have bioavailability issues associated with lower aqueous solubility, first-pass metabolism, poor intestinal permeability and drug absorption, and degradation of the drug throughout its journey in the gastrointestinal tract. A highly developed carrier system known as lipid polymer hybrid nanoparticles (LPHNs) has been introduced. These nanocarriers enhance drug stability, solubility, and absorption, and reduce first-pass metabolism. Consequently, this will have a positive impact on oral bioavailability enhancement. This article provides an in-depth analysis of LPHNs as a novel drug delivery system for anti-cancer agents. It discusses an overview of the limited bioavailability of anti-cancer drugs, their reasons and consequences, LPHNs based anti-cancer drug delivery, conventional and modern preparation methods as well as their drug loading and entrapment efficiencies. In addition, this article also gives an insight into the mechanistic approach to oral bioavailability enhancement, potential applications in anti-cancer drug delivery, limitations, and future prospects of LPHNs in anti-cancer drug delivery.
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
3
|
Cortes-Dericks L, Galetta D. An Overview of Cellular and Molecular Determinants Regulating Chemoresistance in Pleural Mesothelioma. Cancers (Basel) 2025; 17:979. [PMID: 40149313 PMCID: PMC11940806 DOI: 10.3390/cancers17060979] [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: 01/02/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Malignant pleural mesothelioma (PM) is a highly aggressive disease of the lung pleura associated with poor prognosis. Despite advances in improving the clinical management of this malignancy, there is no effective chemotherapy for refractory or relapsing PM. The acquisition of resistance to standard and targeted therapy in this disease is a foremost concern; therefore, a deeper understanding of the complex factors surrounding the emergence of drug resistance is deemed necessary. In this review, we will present broad insights into various cellular and molecular concepts, accounting for the recalcitrance of PM to chemotherapy, including signaling networks regulating drug tolerance, drug resistance-associated proteins, genes, and miRNAs, as well as the critical role of cancer stem cells. Identification of the biological determinants and their associated mechanisms may provide a framework for the development of appropriate treatment.
Collapse
Affiliation(s)
| | - Domenico Galetta
- Division of Thoracic Surgery, San Giovanni Bosco Hospital, 10154 Turin, Italy;
| |
Collapse
|
4
|
Zhang L, Wang Y, Li Y, Chen ZS, Hu C. Advanced materials for cancer treatment and beyond. Front Pharmacol 2025; 16:1557155. [PMID: 40110134 PMCID: PMC11920709 DOI: 10.3389/fphar.2025.1557155] [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: 01/08/2025] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Conservative anti-cancer treatment represented by chemotherapy and surgery lacks tumor-specificity and could hardly resolve the problems associated with multidrug resistance (MDR) in cancers. Novel therapeutic materials in cancer treatment, such as those with anti-MDR or controllable treatment features, represent a significant trend due to their advantages of high and specific efficacy and timely intervention of cancer progress. In addition to their excellent biocompatibility and specificity, they can be utilized in therapies that require ease of operation, provided they are designed with high detection sensitivity. In this review, we summarize a series of recently developed materials that exhibit these advantages, including immune-enhancing and tumor microenvironment (TME)- responsive materials, and those with integrated therapeutic and imaging capabilities. We also introduce advanced modification approaches that can impart essential targeting functionalities to these materials.
Collapse
Affiliation(s)
- Lei Zhang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, China
| | - Yanan Wang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
- College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yangjia Li
- National Engineering Research Center for Sugarcane, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| | - Chaohua Hu
- National Engineering Research Center for Sugarcane, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
5
|
Shirani N, Abdi N, Chehelgerdi M, Yaghoobi H, Chehelgerdi M. Investigating the role of exosomal long non-coding RNAs in drug resistance within female reproductive system cancers. Front Cell Dev Biol 2025; 13:1485422. [PMID: 39925739 PMCID: PMC11802832 DOI: 10.3389/fcell.2025.1485422] [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: 08/23/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Exosomes, as key mediators of intercellular communication, have been increasingly recognized for their role in the oncogenic processes, particularly in facilitating drug resistance. This article delves into the emerging evidence linking exosomal lncRNAs to the modulation of drug resistance mechanisms in cancers such as ovarian, cervical, and endometrial cancer. It synthesizes current research findings on how these lncRNAs influence cancer cell survival, tumor microenvironment, and chemotherapy efficacy. Additionally, the review highlights potential therapeutic strategies targeting exosomal lncRNAs, proposing a new frontier in overcoming drug resistance. By mapping the interface of exosomal lncRNAs and drug resistance, this article aims to provide a comprehensive understanding that could pave the way for innovative treatments and improved patient outcomes in female reproductive system cancers.
Collapse
Affiliation(s)
- Nooshafarin Shirani
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Neda Abdi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Matin Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Hajar Yaghoobi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Novin Genome (NG) Lab, Research and Development Center for Biotechnology, Shahrekord, Iran
- Young Researchers and Elite Club, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| |
Collapse
|
6
|
Kumar P, Kumari I, Prasad R, Ray S, Banerjee A, Prakash A. Elucidating the binding specificity of interactive compounds targeting ATP-binding cassette subfamily G member 2 (ABCG2). Mol Divers 2025:10.1007/s11030-024-11078-2. [PMID: 39786520 DOI: 10.1007/s11030-024-11078-2] [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: 08/22/2024] [Accepted: 12/02/2024] [Indexed: 01/12/2025]
Abstract
The ATP-binding cassette transporter superfamily plays a pivotal role in cellular detoxification and drug efflux. ATP-binding cassette subfamily G member 2 (ABCG2) referred to as the Breast cancer resistance protein has emerged as a key member involved in multidrug resistance displayed by cancer cells. Understanding the molecular basis of substrate and inhibitor recognition, and binding within the transmembrane domain of ABCG2 is crucial for the development of effective therapeutic strategies. Herein, utilizing state-of-the-art molecular docking algorithms and molecular dynamic (MD) simulations, molecular binding of substrates and inhibitors with ABCG2 are defined, distinctly. We performed extensive virtual screening of Drugbank to identify the potential candidates, and MD simulations of docked complexes were carried out in POPC lipid bilayer. Further, the binding affinities of compounds were estimated by free binding energy employing MM-GBSA. To gain deeper insight into the binding affinities and molecular characteristics contributing to inhibitory potential of certain substrates, we included some well-known inhibitors, like Imatinib, Tariquidar, and Ko 143, in our analysis. Docking results show three compounds, Docetaxel > Tariquidar > Tezacaftor having the highest binding affinities (≤ 12.00 kcal/mol) for ABCG2. Remarkably, MM-GBSA results suggest the most stable binding of Tariquidar with ABCG2 as compared to the other inhibitors. Furthermore, our results suggested that Docetaxel, Ozanimod, Pitavastatin, and Tezacaftor have the strongest affinity for the drug-binding site(s) of ABCG2. These results provide valuable insights into the key residues that may govern substrate/inhibitor recognition, shedding light on the molecular determinants influencing substrate specificity, transport kinetics, and ABCG2-mediated drug efflux.
Collapse
Affiliation(s)
- Pawan Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Indu Kumari
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India
- Data Science, Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, India
| | - Rajendra Prasad
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University, Motihari, 845401, India
| | - Atanu Banerjee
- Amity Institute of Biotechnology, Amity University Haryana, Gurugram, India.
| | - Amresh Prakash
- Data Science, Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurugram, India.
| |
Collapse
|
7
|
Bharathiraja P, Baskar S, Prasad NR. Solasodine Downregulates ABCB1 Overexpression in Multidrug Resistant Cancer Cells Via Inhibiting Nrf2/Keap1 Signaling Pathway. J Cell Biochem 2025; 126:e30674. [PMID: 39535293 DOI: 10.1002/jcb.30674] [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: 07/23/2024] [Revised: 10/19/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Multidrug-resistant (MDR) cancer cells maintain redox homeostasis to eliminate oxidative stress-mediated cell death. This study explores the effects of solasodine on regulating P-glycoprotein (P-gp) expression through the Nrf2/Keap1 signaling pathway and oxidative stress-induced sensitization of drug-resistant cancer cells to chemotherapeutics. Initially, the oxidative stress indicators such as intracellular ROS generation, the levels of 8-hydroxy-2-deoxyguanosine (8-OHdG) and gamma-H2AX (γ-H2AX) in the KBChR-8-5 drug-resistant cells were measured. Additionally, the protein expression levels of Nuclear factor erythroid 2-related factor 2 (Nrf-2), Kelch-like ECH-associated protein 1 (Keap1), and ATP Binding Cassette Subfamily B Member 1 (ABCB1)/P-gp were measured at various concentrations of solasodine (1, 5, & 10 µM) through immunofluorescence and western blot analysis. The antioxidant activities in the KBChR-8-5 cells were assessed using established protocols. In this investigation, the treatment with solasodine and doxorubicin combination showed a notable increase in intracellular ROS generation in KBChR-8-5 cells. Furthermore, this combination treatment led to enhanced nuclear condensation, elevated levels of 8-OHdG, and increased γ-H2AX foci formation in the KBChR-8-5 cells. Solasodine treatment effectively inhibited the nuclear translocation of Nrf2 and activation of the ABCB1 gene, consequently preventing overexpression of P-gp in KBChR-8-5 cells. Additionally, the combination therapy increased the lipid peroxidation levels while simultaneously reducing the activities of antioxidant enzymes such as superoxide dismutase (SOD), catalase (CAT), glutathione peroxidase (GPx), and the levels of glutathione (GSH). These results demonstrated that solasodine disrupts redox balance, and overcomes drug resistance by downregulating P-gp via regulating Nrf2/Keap1 signaling pathway in MDR cancer cells.
Collapse
Affiliation(s)
- Pradhapsingh Bharathiraja
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Sugumar Baskar
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - N Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
| |
Collapse
|
8
|
Ghosh S, Sharma A, Kumar RS, Nasare V. Sorcin: mechanisms of action in cancer hallmarks, drug resistance and opportunities in therapeutics. Med Oncol 2024; 42:29. [PMID: 39673665 DOI: 10.1007/s12032-024-02580-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Soluble resistant related calcium binding protein (Sorcin) plays an important role in tumor progression, angiogenesis, metastasis, and multidrug resistance. Differential expression of Sorcin across different cancers significantly correlates with key clinicopathological characteristics and survival outcomes, underscoring its potential as a prognostic marker. Its involvement in drug-resistant cancers further advert Sorcin as a promising therapeutic target. This review summarizes the mechanistic role of Sorcin in cancer, its contribution to drug resistance, clinical relevance, and the current and emerging therapeutic approaches aimed at translating Sorcin-targeted therapies into clinical practice.
Collapse
Affiliation(s)
- Sushmita Ghosh
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, India
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, India
| | - Arpana Sharma
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, India
| | - R Suresh Kumar
- Molecular Biology Division, National Institute of Cancer Prevention and Research, ICMR, Noida, Delhi, India
| | - Vilas Nasare
- Department of Pathology and Cancer Screening, Chittaranjan National Cancer Institute, Kolkata, India.
| |
Collapse
|
9
|
Bharathidasan D, Maity C. Organelle-Specific Smart Supramolecular Materials for Bioimaging and Theranostics Application. Top Curr Chem (Cham) 2024; 383:1. [PMID: 39607460 DOI: 10.1007/s41061-024-00483-8] [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: 12/29/2023] [Accepted: 11/04/2024] [Indexed: 11/29/2024]
Abstract
In cellular environments, certain synthetic molecules can form nanostructures via self-assembly, impacting molecular imaging, and biomedical applications. Control over the formation of these self-assembled nanostructures in subcellular organelle is challenging. By the action of stimuli, either present in the cellular environment or applied externally, in situ generation of molecular precursors can lead to accumulation and supramolecular nanostructure formation, resulting in efficient bioimaging. Here, we summarize smart fluorophore-based ordered nanostructure preparation at specific organelles for efficient bioimaging and therapeutic application towards cancer theranostics. We also present challenges and an outlook regarding intercellular self-assembly for theranostics application. Altogether, smart nanostructured materials with fluorescence read-outs at specific subcellular compartments would be beneficial in synthetic biology and precision therapeutics.
Collapse
Affiliation(s)
- Dineshkumar Bharathidasan
- (Organic)Material Science and Engineering Laboratory, Centre for Nanobiotechnology (CNBT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamilnadu, 632014, India
| | - Chandan Maity
- (Organic)Material Science and Engineering Laboratory, Centre for Nanobiotechnology (CNBT), Vellore Institute of Technology (VIT), Vellore Campus, Vellore, Tamilnadu, 632014, India.
| |
Collapse
|
10
|
Druzhkova I, Potapov A, Ignatova N, Bugrova M, Shchechkin I, Lukina M, Shimolina L, Kolesnikova E, Shirmanova M, Zagaynova E. Cell hiding in colorectal cancer: correlation with response to chemotherapy in vitro and in vivo. Sci Rep 2024; 14:28762. [PMID: 39567584 PMCID: PMC11579335 DOI: 10.1038/s41598-024-79948-1] [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: 08/05/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024] Open
Abstract
Resistance to chemotherapy remains the main challenge for cancer treatment. One of the mechanisms of tumor escape from cytotoxic agents could be the formation of cell-in-cell (CIC) structures, in which the outer cell protects the inner cell from unfavorable environment. Such structures have been found in many tumor types, however, their link to chemosensitivity is elusive. Here, we tested whether the CIC structures can promote resistance of colorectal cancer cells to chemotherapy. To identify CIC structures in cell cultures and in tumor xenografts, both transmission electron microscopy and confocal fluorescence microscopy of live and fixed cells as well as tissue slices and histopathology were used. Cytogenetic analysis was performed to detect chromosome instability associated with the drug resistance. It was found that in the five colorectal cancer cell lines intrinsic chemoresistance positively correlated with the ability of cells to spontaneously form CIC structures. Cultured cells treated with oxaliplatin and Irinotecan and tumor xenografts treated with FOLFOX or FOLFIRI regimens displayed an increased number of CICs after the treatment. The release of the inner cell from CIC structure was observed after removal of the drug. The number of CICs in the cell lines and tumors with acquired resistance to oxaliplatin was higher than in the drug-naive counterparts. The development of chemoresistance was also accompanied by the changes in the cell's ploidy. These preliminary data clearly demonstrate the associations of CIC structures with chemoresistance of colorectal cancer in cultured cells and tumor xenografts and show the prospect of further clinical validation of CICs as a potential prognostic marker for treatment efficiency.
Collapse
Affiliation(s)
- I Druzhkova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia.
| | - A Potapov
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - N Ignatova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - M Bugrova
- Faculty of Histology with Cytology and Embryology, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - I Shchechkin
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - M Lukina
- Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, Moscow, Russian Federation
| | - L Shimolina
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - E Kolesnikova
- Nizhny Novgorod Regional Oncologic Hospital, Nizhny Novgorod, Russia
| | - M Shirmanova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
| | - E Zagaynova
- Research Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, Nizhny Novgorod, Russia
- Federal Research and Clinical Center of Physical-Chemical Medicine of the Federal Medical Biological Agency, Moscow, Russian Federation
| |
Collapse
|
11
|
Iweala EEJ, Amuji DN, Oluwajembola AM, Ugbogu EA. Targeting c-Met in breast cancer: From mechanisms of chemoresistance to novel therapeutic strategies. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 7:100204. [PMID: 39524211 PMCID: PMC11543557 DOI: 10.1016/j.crphar.2024.100204] [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: 05/07/2024] [Revised: 09/28/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Breast cancer presents a significant challenge due to its heterogeneity and propensity for developing chemoresistance, particularly in the triple-negative subtype. c-Mesenchymal epithelial transition factor (c-Met), a receptor tyrosine kinase, presents a promising target for breast cancer therapy due to its involvement in disease progression and poor prognosis. However, the heterogeneous expression of c-Met within breast cancer subtypes and individual tumors complicates targeted therapy. Also, cancer cells can develop resistance to c-Met inhibitors through various mechanisms, including bypass signaling pathways and genetic mutations. The off-target effects of c-Met inhibitors further limit their clinical utility, necessitating the development of more selective agents. To overcome these challenges, personalized treatment approaches and combination therapies are being explored to improve treatment efficacy while minimizing adverse effects. Novel c-Met inhibitors with improved selectivity and reduced off-target toxicity show promise in preclinical studies. Additionally, targeted delivery systems aim to enhance drug localization and reduce systemic toxicity. Future directions involve refining inhibitor design and integrating c-Met inhibition into personalized treatment regimens guided by molecular profiling. This review explores the mechanisms by which c-Met contributes to chemoresistance in breast cancer and current challenges in targeting c-Met for breast cancer therapy. It discusses strategies to optimize treatment outcomes, ultimately improving patient prognosis and reducing mortality rates associated with this devastating disease.
Collapse
Affiliation(s)
- Emeka Eze Joshua Iweala
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Doris Nnenna Amuji
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | - Abimbola Mary Oluwajembola
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota, Nigeria
- Covenant Applied Informatics and Communication Africa Centre of Excellence (CApIC-ACE), Covenant University, Ota, Nigeria
| | | |
Collapse
|
12
|
Kraft FB, Biermann L, Schäker-Hübner L, Hanl M, Hamacher A, Kassack MU, Hansen FK. Hydrazide-Based Class I Selective HDAC Inhibitors Completely Reverse Chemoresistance Synergistically in Platinum-Resistant Solid Cancer Cells. J Med Chem 2024; 67:17796-17819. [PMID: 39356226 DOI: 10.1021/acs.jmedchem.4c01817] [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: 10/03/2024]
Abstract
In this work, we have synthesized a set of peptoid-based histone deacetylase inhibitors (HDACi) with a substituted hydrazide moiety as zinc-binding group. Subsequently, all compounds were evaluated in biochemical HDAC inhibition assays and for their antiproliferative activity against native and cisplatin-resistant cancer cell lines. The hydrazide derivatives with a propyl or butyl substituent (compounds 5 and 6) emerged as the most potent class I HDAC selective inhibitors (HDAC1-3). Further, compounds 5 and 6 outperformed entinostat in cytotoxicity assays and were able to reverse chemoresistance in cisplatin-resistant A2780 (ovarian) and Cal27 (head-neck) cancer cell lines. Moreover, the hydrazide derivatives 5 and 6 showed strong synergism with cisplatin (combination indices <0.2), again outperforming entinostat, and increased DNA damage, p21, and pro-apoptotic BIM expression, leading to caspase-mediated apoptosis and cell death. Thus, compounds 5 and 6 represent promising lead structures for developing new HDACi capable of reversing chemoresistance in cisplatin resistant cancer cells.
Collapse
Affiliation(s)
- Fabian B Kraft
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Lukas Biermann
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Linda Schäker-Hübner
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Maria Hanl
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| | - Alexandra Hamacher
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Matthias U Kassack
- Institute for Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, 40225 Düsseldorf, Germany
| | - Finn K Hansen
- Department of Pharmaceutical and Cell Biological Chemistry, Pharmaceutical Institute, University of Bonn, An der Immenburg 4, 53121 Bonn, Germany
| |
Collapse
|
13
|
Evren AE, Nuha D, Dawbaa S, Karaduman AB, Sağlik BN, Yurttaş L. Novel oxadiazole-thiadiazole derivatives: synthesis, biological evaluation, and in silico studies. J Biomol Struct Dyn 2024; 42:8688-8700. [PMID: 37587853 DOI: 10.1080/07391102.2023.2247087] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/05/2023] [Indexed: 08/18/2023]
Abstract
In the search for new anticancer agents, we synthesized a new series of thiazole derivatives carried on thiadiazole-oxadiazole hybrid. Final compounds (5a-5i) were analyzed via 1H NMR, 13C NMR, and HRMS. The pharmacokinetic profile of the targeted compounds was predicted via in silico calculations. Their anticancer properties were determined using MTT method against MCF7 and A549 cell lines. Compounds 5a, 5b and 5c were found more active against MCF7 cells than A549 cells while they were not cytotoxic on L929 healthy cells. Generally, it can be summarized that acetamide moiety has a pivotal role in anticancer activity. For further studies, their aromatase inhibitory activity was evaluated. After determination all these features, the binding modes of the active compounds and the stability and relation of the ligand-enzyme complex were investigated using molecular docking and molecular dynamics simulation studies, respectively. In vitro and in silico studies suggest two important structure-activity relationship (SAR) points that at least one azole ring is essential, and if there is approximately 8.0 ± 0.5 Å distance between the H-bond rich zone of ligand and the heteroaryl ring system of ligand has a major impact on aromatase inhibitory activity. Compounds with small group substitution on thiazole are found potentially may be used for the treatment of anti-breast cancer orally.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Asaf Evrim Evren
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Pharmacy Services, Vocational School of Health Services, Bilecik Seyh Edebali University, Bilecik, Turkey
| | - Demokrat Nuha
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Faculty of Pharmacy, University for Business and Technology, Prishtina, Kosovo
| | - Sam Dawbaa
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
- Department of Doctor of Pharmacy (PharmD), Faculty of Medical Sciences, Thamar University, Dhamar, Yemen
- Department of Pharmacy, Faculty of Medical Sciences, Al-Hikma University, Dhamar, Yemen
| | - Abdullah Burak Karaduman
- Department of Pharmaceutical Toxicology, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Begüm Nurpelin Sağlik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| | - Leyla Yurttaş
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskişehir, Turkey
| |
Collapse
|
14
|
Malik S, Sikander M, Wahid M, Dhasmana A, Sarwat M, Khan S, Cobos E, Yallapu MM, Jaggi M, Chauhan SC. Deciphering cellular and molecular mechanism of MUC13 mucin involved in cancer cell plasticity and drug resistance. Cancer Metastasis Rev 2024; 43:981-999. [PMID: 38498072 DOI: 10.1007/s10555-024-10177-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024]
Abstract
There has been a surge of interest in recent years in understanding the intricate mechanisms underlying cancer progression and treatment resistance. One molecule that has recently emerged in these mechanisms is MUC13 mucin, a transmembrane glycoprotein. Researchers have begun to unravel the molecular complexity of MUC13 and its impact on cancer biology. Studies have shown that MUC13 overexpression can disrupt normal cellular polarity, leading to the acquisition of malignant traits. Furthermore, MUC13 has been associated with increased cancer plasticity, allowing cells to undergo epithelial-mesenchymal transition (EMT) and metastasize. Notably, MUC13 has also been implicated in the development of chemoresistance, rendering cancer cells less responsive to traditional treatment options. Understanding the precise role of MUC13 in cellular plasticity, and chemoresistance could pave the way for the development of targeted therapies to combat cancer progression and enhance treatment efficacy.
Collapse
Affiliation(s)
- Shabnam Malik
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Mohammed Sikander
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Mohd Wahid
- Unit of Research and Scientific Studies, College of Nursing and Allied Health Sciences, University of Jazan, Jizan, Saudi Arabia
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Uttar Pradesh, Noida, India
| | - Sheema Khan
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Everardo Cobos
- Department of Medicine, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, Biomedical Research Building, University of Texas Rio Grande Valley, 5300 North L Street, McAllen, TX, 78504, USA.
- South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, USA.
| |
Collapse
|
15
|
Vijayakumar S, Dhakshanamoorthy R, Baskaran A, Sabari Krishnan B, Maddaly R. Drug resistance in human cancers - Mechanisms and implications. Life Sci 2024; 352:122907. [PMID: 39004273 DOI: 10.1016/j.lfs.2024.122907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/27/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
Cancers have complex etiology and pose a significant impact from the health care perspective apart from the socio-economic implications. The enormity of challenge posed by cancers can be understood from the fact that clinical trials for cancer therapy has yielded minimum potential promises compared to those obtained for other diseases. Surgery, chemotherapy and radiotherapy continue to be the mainstay therapeutic options for cancers. Among the challenges posed by these options, induced resistance to chemotherapeutic drugs is probably the most significant contributor for poor prognosis and ineffectiveness of the therapy. Drug resistance is a property exhibited by almost all cancer types including carcinomas, leukemias, myelomas, sarcomas and lymphomas. The mechanisms by which drug resistance is induced include the factors within the tumor microenvironment, mutations in the genes responsible for drug metabolism, changes in the surface drug receptors and increased drug efflux. We present here comprehensively the drug resistance in cancers along with their mechanisms. Also, apart from resistance to regularly used chemotherapeutic drugs, we present resistance induction to new generation therapeutic agents such as monoclonal antibodies. Finally, we have discussed the experimental approaches to understand the mechanisms underlying induction of drug resistance and potential ways to mitigate induced drug resistance.
Collapse
Affiliation(s)
- Sudikshaa Vijayakumar
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu 600116, India
| | - Raveena Dhakshanamoorthy
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu 600116, India
| | - Akshaya Baskaran
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu 600116, India
| | - B Sabari Krishnan
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu 600116, India
| | - Ravi Maddaly
- Department of Human Genetics, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Porur, Chennai, Tamil Nadu 600116, India.
| |
Collapse
|
16
|
Wang X, Xue X, Pang M, Yu L, Qian J, Li X, Tian M, Lyu A, Lu C, Liu Y. Epithelial-mesenchymal plasticity in cancer: signaling pathways and therapeutic targets. MedComm (Beijing) 2024; 5:e659. [PMID: 39092293 PMCID: PMC11292400 DOI: 10.1002/mco2.659] [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: 01/27/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 08/04/2024] Open
Abstract
Currently, cancer is still a leading cause of human death globally. Tumor deterioration comprises multiple events including metastasis, therapeutic resistance and immune evasion, all of which are tightly related to the phenotypic plasticity especially epithelial-mesenchymal plasticity (EMP). Tumor cells with EMP are manifest in three states as epithelial-mesenchymal transition (EMT), partial EMT, and mesenchymal-epithelial transition, which orchestrate the phenotypic switch and heterogeneity of tumor cells via transcriptional regulation and a series of signaling pathways, including transforming growth factor-β, Wnt/β-catenin, and Notch. However, due to the complicated nature of EMP, the diverse process of EMP is still not fully understood. In this review, we systematically conclude the biological background, regulating mechanisms of EMP as well as the role of EMP in therapy response. We also summarize a range of small molecule inhibitors, immune-related therapeutic approaches, and combination therapies that have been developed to target EMP for the outstanding role of EMP-driven tumor deterioration. Additionally, we explore the potential technique for EMP-based tumor mechanistic investigation and therapeutic research, which may burst vigorous prospects. Overall, we elucidate the multifaceted aspects of EMP in tumor progression and suggest a promising direction of cancer treatment based on targeting EMP.
Collapse
Affiliation(s)
- Xiangpeng Wang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoxia Xue
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Mingshi Pang
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Liuchunyang Yu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Jinxiu Qian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Xiaoyu Li
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Meng Tian
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| | - Aiping Lyu
- School of Chinese MedicineHong Kong Baptist UniversityKowloonHong KongChina
| | - Cheng Lu
- Institute of Basic Research in Clinical MedicineChina Academy of Chinese Medical SciencesBeijingChina
| | - Yuanyan Liu
- School of Materia MedicaBeijing University of Chinese MedicineBeijingChina
| |
Collapse
|
17
|
Płonka-Czerw J, Żyrek L, Latocha M. Changes in the Sensitivity of MCF-7 and MCF-7/DX Breast Cancer Cells to Cytostatic in the Presence of Metformin. Molecules 2024; 29:3531. [PMID: 39124936 PMCID: PMC11313889 DOI: 10.3390/molecules29153531] [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: 07/01/2024] [Revised: 07/22/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Multidrug resistance is a serious problem in modern medicine and the reason for the failure of various therapies. A particularly important problem is the occurrence of multidrug resistance in cancer therapies which affects many cancer patients. Observations on the effect of metformin-a well-known hypoglycemic drug used in the treatment of type 2 diabetes-on cancer cells indicate the possibility of an interaction of this substance with drugs already used and, as a result, an increase in the sensitivity of cancer cells to cytostatics. The aim of this study was to evaluate the effect of metformin on the occurrence of multidrug resistance of breast cancer cells. The MCF-7-sensitive cell line and the MCF-7/DX cytostatic-resistant cell line were used for this study. WST-1 and LDH assays were used to evaluate the effects of metformin and doxorubicin on cell proliferation and viability. The effect of metformin on increasing the sensitivity of MCF-7 and MCF-7/DX cells to doxorubicin was evaluated in an MDR test. The participation of metformin in increasing the sensitivity of resistant cells to the effect of the cytostatic (doxorubicin) has been demonstrated.
Collapse
Affiliation(s)
- Justyna Płonka-Czerw
- Department of Cell Biology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, Jedności 8, 41-200 Sosnowiec, Poland; (L.Ż.); (M.L.)
| | | | | |
Collapse
|
18
|
Garg P, Malhotra J, Kulkarni P, Horne D, Salgia R, Singhal SS. Emerging Therapeutic Strategies to Overcome Drug Resistance in Cancer Cells. Cancers (Basel) 2024; 16:2478. [PMID: 39001539 PMCID: PMC11240358 DOI: 10.3390/cancers16132478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/01/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024] Open
Abstract
The rise of drug resistance in cancer cells presents a formidable challenge in modern oncology, necessitating the exploration of innovative therapeutic strategies. This review investigates the latest advancements in overcoming drug resistance mechanisms employed by cancer cells, focusing on emerging therapeutic modalities. The intricate molecular insights into drug resistance, including genetic mutations, efflux pumps, altered signaling pathways, and microenvironmental influences, are discussed. Furthermore, the promising avenues offered by targeted therapies, combination treatments, immunotherapies, and precision medicine approaches are highlighted. Specifically, the synergistic effects of combining traditional cytotoxic agents with molecularly targeted inhibitors to circumvent resistance pathways are examined. Additionally, the evolving landscape of immunotherapeutic interventions, including immune checkpoint inhibitors and adoptive cell therapies, is explored in terms of bolstering anti-tumor immune responses and overcoming immune evasion mechanisms. Moreover, the significance of biomarker-driven strategies for predicting and monitoring treatment responses is underscored, thereby optimizing therapeutic outcomes. For insights into the future direction of cancer treatment paradigms, the current review focused on prevailing drug resistance challenges and improving patient outcomes, through an integrative analysis of these emerging therapeutic strategies.
Collapse
Affiliation(s)
- Pankaj Garg
- Department of Chemistry, GLA University, Mathura 281406, India
| | - Jyoti Malhotra
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Prakash Kulkarni
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - David Horne
- Molecular Medicine, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Ravi Salgia
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| | - Sharad S. Singhal
- Departments of Medical Oncology & Therapeutics Research, Beckman Research Institute of City of Hope, Comprehensive Cancer Center, National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
19
|
McKenzie M, Lian GY, Pennel KA, Quinn JA, Jamieson NB, Edwards J. NFκB signalling in colorectal cancer: Examining the central dogma of IKKα and IKKβ signalling. Heliyon 2024; 10:e32904. [PMID: 38975078 PMCID: PMC11226910 DOI: 10.1016/j.heliyon.2024.e32904] [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: 02/09/2024] [Revised: 05/20/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
The NFκB pathway, known as the central regulator of inflammation, has a well-established role in colorectal cancer (CRC) initiation, progression, and therapy resistance. Due to the pathway's overarching roles in CRC, there have been efforts to characterise NFκB family members and target the pathway for therapeutic intervention. Initial research illustrated that the canonical NFκB pathway, driven by central kinase IKKβ, was a promising target for drug intervention. However, dose limiting toxicities and specificity concerns have resulted in failure of IKKβ inhibitors in clinical trials. The field has turned to look at targeting the less dominant kinase, IKKα, which along with NFκB inducing kinase (NIK), drives the lesser researched non-canonical NFκB pathway. However prognostic studies of the non-canonical pathway have produced conflicting results. There is emerging evidence that IKKα is involved in other signalling pathways, which lie outside of canonical and non-canonical NFκB signalling. Evidence suggests that some of these alternative pathways involve a truncated form of IKKα, and this may drive poor cancer-specific survival in CRC. This review aims to explore the multiple components of NFκB signalling, highlighting that NIK may be the central kinase for non-canonical NFκB signalling, and that IKKα is involved in novel pathways which promote CRC.
Collapse
Affiliation(s)
- Molly McKenzie
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Guang-Yu Lian
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Kathryn A.F. Pennel
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Jean A. Quinn
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Nigel B. Jamieson
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| | - Joanne Edwards
- School of Cancer Sciences, Wolfson Wohl Cancer Research Centre, University of Glasgow, Glasgow, G61 1BD, UK
| |
Collapse
|
20
|
Zhong C, Jiang WJ, Yao Y, Li Z, Li Y, Wang S, Wang X, Zhu W, Wu S, Wang J, Fan S, Ma S, Liu Y, Zhang H, Zhao W, Zhao L, Feng Y, Li Z, Guo R, Yu L, Pei F, Hu J, Feng X, Yang Z, Yang Z, Yang X, Hou Y, Zhang D, Xu D, Sheng R, Li Y, Liu L, Wu HJ, Huang J, Fei T. CRISPR screens reveal convergent targeting strategies against evolutionarily distinct chemoresistance in cancer. Nat Commun 2024; 15:5502. [PMID: 38951519 PMCID: PMC11217446 DOI: 10.1038/s41467-024-49673-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
Resistance to chemotherapy has been a major hurdle that limits therapeutic benefits for many types of cancer. Here we systematically identify genetic drivers underlying chemoresistance by performing 30 genome-scale CRISPR knockout screens for seven chemotherapeutic agents in multiple cancer cells. Chemoresistance genes vary between conditions primarily due to distinct genetic background and mechanism of action of drugs, manifesting heterogeneous and multiplexed routes towards chemoresistance. By focusing on oxaliplatin and irinotecan resistance in colorectal cancer, we unravel that evolutionarily distinct chemoresistance can share consensus vulnerabilities identified by 26 second-round CRISPR screens with druggable gene library. We further pinpoint PLK4 as a therapeutic target to overcome oxaliplatin resistance in various models via genetic ablation or pharmacological inhibition, highlighting a single-agent strategy to antagonize evolutionarily distinct chemoresistance. Our study not only provides resources and insights into the molecular basis of chemoresistance, but also proposes potential biomarkers and therapeutic strategies against such resistance.
Collapse
Affiliation(s)
- Chunge Zhong
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
- Foshan Graduate School of Innovation, Northeastern University, Foshan, 528311, China
| | - Wen-Jie Jiang
- Peking University Third Hospital, Beijing, 100191, China
| | - Yingjia Yao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Zexu Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - You Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shengnan Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Xiaofeng Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Wenjuan Zhu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Siqi Wu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Jing Wang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Shuangshuang Fan
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Shixin Ma
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Yeshu Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Han Zhang
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Wenchang Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Lu Zhao
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Yi Feng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Zihan Li
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China
| | - Ruifang Guo
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Li Yu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Fengyun Pei
- Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jun Hu
- Clinical Research Center, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Xingzhi Feng
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Zihuan Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Institute of Gastroenterology, Guangzhou, China
| | - Zhengjia Yang
- Department of Cardiothoracic Surgery, Jinqiu Hospital of Liaoning Province, Shenyang, China
| | - Xueying Yang
- Department of Thoracic Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Yue Hou
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Danni Zhang
- Key Laboratory for Anisotropy and Texture of Materials (Ministry of Education), School of Materials Science and Engineering, Northeastern University, Shenyang, China
| | - Dake Xu
- Key Laboratory for Anisotropy and Texture of Materials (Ministry of Education), School of Materials Science and Engineering, Northeastern University, Shenyang, China
| | - Ren Sheng
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Yihao Li
- BeiGene Institute, BeiGene (Shanghai) Research & Development Co., Ltd, 200131, Shanghai, China
| | - Lijun Liu
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China
| | - Hua-Jun Wu
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, 100142, China.
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China.
- Center for Precision Medicine Multi-Omics Research, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Jun Huang
- Department of Colorectal Surgery, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Clinical Research Center, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Institute of Gastroenterology, Guangzhou, China.
| | - Teng Fei
- Key Laboratory of Bioresource Research and Development of Liaoning Province, College of Life and Health Sciences, Northeastern University, Shenyang, 110819, China.
- National Frontiers Science Center for Industrial Intelligence and Systems Optimization, Northeastern University, Shenyang, 110819, China.
- Key Laboratory of Data Analytics and Optimization for Smart Industry (Northeastern University), Ministry of Education, Shenyang, 110819, China.
- Foshan Graduate School of Innovation, Northeastern University, Foshan, 528311, China.
| |
Collapse
|
21
|
Kalnins T, Vitkovska V, Kazak M, Zelencova-Gopejenko D, Ozola M, Narvaiss N, Makrecka-Kuka M, Domračeva I, Kinens A, Gukalova B, Konrad N, Aav R, Bonato F, Lucena-Agell D, Díaz JF, Liepinsh E, Suna E. Development of Potent Microtubule Targeting Agent by Structural Simplification of Natural Diazonamide. J Med Chem 2024; 67:9227-9259. [PMID: 38833507 DOI: 10.1021/acs.jmedchem.4c00388] [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: 06/06/2024]
Abstract
The marine metabolite diazonamide A exerts low nanomolar cytotoxicity against a range of tumor cell lines; however, its highly complex molecular architecture undermines the therapeutic potential of the natural product. We demonstrate that truncation of heteroaromatic macrocycle in natural diazonamide A, combined with the replacement of the challenging-to-synthesize tetracyclic hemiaminal subunit by oxindole moiety leads to considerably less complex analogues with improved drug-like properties and nanomolar antiproliferative potency. The structurally simplified macrocycles are accessible in 12 steps from readily available indolin-2-one and tert-leucine with excellent diastereoselectivity (99:1 dr) in the key macrocyclization step. The most potent macrocycle acts as a tubulin assembly inhibitor and exerts similar effects on A2058 cell cycle progression and induction of apoptosis as does marketed microtubule-targeting agent vinorelbine.
Collapse
Affiliation(s)
- Toms Kalnins
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Viktorija Vitkovska
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Mihail Kazak
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | | | - Melita Ozola
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Nauris Narvaiss
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | | | - Ilona Domračeva
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Artis Kinens
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Baiba Gukalova
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Nele Konrad
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn, Harju Maakon 12618, Estonia
| | - Riina Aav
- Department of Chemistry and Biotechnology, Tallinn University of Technology, Akadeemia tee 15, Tallinn, Harju Maakon 12618, Estonia
| | - Francesca Bonato
- Unidad BICS, Centro de Investigaciones Biologicas Margarita Salas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, Madrid 28040, Spain
| | - Daniel Lucena-Agell
- Unidad BICS, Centro de Investigaciones Biologicas Margarita Salas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, Madrid 28040, Spain
| | - J Fernando Díaz
- Unidad BICS, Centro de Investigaciones Biologicas Margarita Salas, Consejo Superior de Investigaciones Cientificas, Ramiro de Maeztu 9, Madrid 28040, Spain
| | - Edgars Liepinsh
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| | - Edgars Suna
- Latvian Institute of Organic Synthesis, Aizkraukles 21, Riga LV-1006, Latvia
| |
Collapse
|
22
|
Fiorini S, Rubini E, Perugini M, Altieri F, Chichiarelli S, Meschiari G, Arrighetti G, Vijgen J, Natali PG, Minacori M, Eufemi M. STAT3 Pathways Contribute to β-HCH Interference with Anticancer Tyrosine Kinase Inhibitors. Int J Mol Sci 2024; 25:6181. [PMID: 38892372 PMCID: PMC11173063 DOI: 10.3390/ijms25116181] [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: 04/30/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024] Open
Abstract
Organochlorine pesticides (OCPs) are a class of environmentally persistent and bioaccumulative pollutants. Among these, β-hexachlorocyclohexane (β-HCH) is a byproduct of lindane synthesis, one of the most worldwide widespread pesticides. β-HCH cellular mechanisms inducing chemical carcinogenesis correspond to many of those inducing chemoresistance, in particular, by the activation of signal transducer and activator of transcription 3 (STAT3) signaling pathways. For this purpose, four cell lines, representative of breast, lung, prostate, and hepatocellular cancers, were treated with β-HCH, specific tyrosine kinase inhibitors (TKIs), and a STAT3 inhibitor. All cell samples were analyzed by a viability assay, immunoblotting analysis, a wound-healing assay, and a colony formation assay. The results show that β-HCH reduces the efficacy of TKIs. The STAT3 protein, in this context, plays a central role. In fact, by inhibiting its activity, the efficacy of the anticancer drug is restored. Furthermore, this manuscript aimed to draw the attention of the scientific and socio-healthcare community to the issue of prolonged exposure to contaminants and their impact on drug efficacy.
Collapse
Affiliation(s)
- Sara Fiorini
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (S.F.); (F.A.); (S.C.); (G.M.); (M.M.); (M.E.)
| | - Elisabetta Rubini
- Institute of Molecular Biology and Pathology, CNR National Research Council, Via degli Apuli, 4, 00185 Rome, Italy;
| | - Monia Perugini
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Campus “Aurelio Saliceti”, Via R. Balzarini 1, 64100 Teramo, Italy;
| | - Fabio Altieri
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (S.F.); (F.A.); (S.C.); (G.M.); (M.M.); (M.E.)
| | - Silvia Chichiarelli
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (S.F.); (F.A.); (S.C.); (G.M.); (M.M.); (M.E.)
| | - Giorgia Meschiari
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (S.F.); (F.A.); (S.C.); (G.M.); (M.M.); (M.E.)
| | - Giulia Arrighetti
- Department of Cultures, Politics and Society, University of Turin, Via Verdi, 8, 10124 Turin, Italy;
| | - John Vijgen
- International HCH and Pesticides Association (IHPA), Elmevej 14, 2840 Holte, Denmark;
| | - Pier Giorgio Natali
- Collegium Ramazzini, Castello di Bentivoglio, Via Saliceto, 3, 40010 Bologna, Italy
- Lega Italiana per la Lotta contro i Tumori (LILT), Associazione Metropolitana di Roma, Via Nomentana, 303, 00162 Rome, Italy
| | - Marco Minacori
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (S.F.); (F.A.); (S.C.); (G.M.); (M.M.); (M.E.)
- Department of Bioscience and Agro-Food and Environmental Technology, University of Teramo, Campus “Aurelio Saliceti”, Via R. Balzarini 1, 64100 Teramo, Italy;
| | - Margherita Eufemi
- Department of Biochemical Science “A. Rossi Fanelli”, Faculty of Pharmacy and Medicine, Sapienza University of Rome, P.le Aldo Moro 5, 00185 Rome, Italy; (S.F.); (F.A.); (S.C.); (G.M.); (M.M.); (M.E.)
| |
Collapse
|
23
|
Lagzian A, Askari M, Haeri MS, Sheikhi N, Banihashemi S, Nabi-Afjadi M, Malekzadegan Y. Increased V-ATPase activity can lead to chemo-resistance in oral squamous cell carcinoma via autophagy induction: new insights. Med Oncol 2024; 41:108. [PMID: 38592406 DOI: 10.1007/s12032-024-02313-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/23/2024] [Indexed: 04/10/2024]
Abstract
Oral squamous cell carcinoma (OSCC) is a cancer type with a high rate of recurrence and a poor prognosis. Tumor chemo-resistance remains an issue for OSCC patients despite the availability of multimodal therapy options, which causes an increase in tumor invasiveness. Vacuolar ATPase (V-ATPase), appears to be one of the most significant molecules implicated in MDR in tumors like OSCC. It is primarily responsible for controlling the acidity in the solid tumors' microenvironment, which interferes with the absorption of chemotherapeutic medications. However, the exact cellular and molecular mechanisms V-ATPase plays in OSCC chemo-resistance have not been understood. Uncovering these mechanisms can contribute to combating OSCC chemo-resistance and poor prognosis. Hence, in this review, we suggest that one of these underlying mechanisms is autophagy induced by V-ATPase which can potentially contribute to OSCC chemo-resistance. Finally, specialized autophagy and V-ATPase inhibitors may be beneficial as an approach to reduce drug resistance to anticancer therapies in addition to serving as coadjuvants in antitumor treatments. Also, V-ATPase could be a prognostic factor for OSCC patients. However, in the future, more investigations are required to demonstrate these suggestions and hypotheses.
Collapse
Affiliation(s)
- Ahmadreza Lagzian
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Marziye Askari
- Department of Immunology, School of Medicine, Hamedan University of Medical Sciences, Hamedan, Iran
| | - Melika Sadat Haeri
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nastaran Sheikhi
- Biotechnology Department, Biological Sciences Faculty, Alzahra University, Tehran, Iran
| | - Sara Banihashemi
- Department of Bioscience, School of Science and Technology, Nottingham Trend University, Nottingham, UK
| | - Mohsen Nabi-Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Yalda Malekzadegan
- Department of Microbiology, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
24
|
Wang Q, Huang CH, Wibowo FS, Amin R, Shen J, Li F, Babu RJ. Elesclomol-Copper Nanoparticles Overcome Multidrug Resistance in Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:13509-13524. [PMID: 38466024 DOI: 10.1021/acsami.3c17792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Elesclomol (ES), a copper-binding ionophore, forms an ES-Cu complex with copper ions (Cu(II)). ES-Cu has been proven to induce mitochondrial oxidative stress and copper-dependent cell death (cuprotosis). However, ES-Cu is poorly water-soluble, and its delivery to various cancer cells is a challenge. Herein, we designed a d-α-tocopherol polyethylene glycol 1000 succinate/chondroitin sulfate-cholic acid (TPGS/CS-CA)-based micellar nanoparticle for delivering the ES-Cu complex to various cancer cell lines to demonstrate its efficacy as an anticancer agent. The ES-Cu nanoparticles exerted high encapsulation efficiency and excellent serum stability. The anticancer efficacy of ES-Cu nanoparticles was evaluated in various drug-sensitive cell lines (DU145, PC3, and A549) and drug-resistant cell lines (DU145TXR, PC3TXR, and A549TXR). The results showed that ES-Cu nanoparticles exerted potent anticancer activities in both drug-sensitive and drug-resistant cell lines. The Western blotting, reverse transcription quantitative polymerase chain reaction (RT-qPCR), and molecular docking results suggested that ES-Cu is not a substrate for P glycoprotein (P-gp), which is an efflux transporter potentially causing multidrug resistance (MDR) in cancer cells. ES-Cu nanoparticles could bypass P-gp without compromising their activity, indicating that they may overcome MDR in cancer cells and provide a novel therapeutic strategy. Additionally, the extracellular matrix of ES-Cu nanoparticles-pretreated drug-resistant cells could polarize Raw 264.7 macrophages into the M1 phenotype. Therefore, our TPGS/CS-CA-based ES-Cu nanoparticles provide an effective method of delivering the ES-Cu complex, a promising strategy to overcome MDR in cancer therapy with potential immune response stimulation.
Collapse
Affiliation(s)
- Qi Wang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Chung-Hui Huang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Fajar S Wibowo
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Rajesh Amin
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Jianzhong Shen
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| | - Feng Li
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
- National Institute on Drug Abuse, 11601 Landsdown Street, North Bethesda, Maryland 20852, United States
| | - R Jayachandra Babu
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
25
|
Cao X, Gong Y. Recent developments of hydroxamic acid hybrids as potential anti-breast cancer agents. Future Med Chem 2024; 16:469-492. [PMID: 38293775 DOI: 10.4155/fmc-2023-0284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 12/20/2023] [Indexed: 02/01/2024] Open
Abstract
Histone deacetylase inhibitors not only possess favorable effects on modulating tumor microenvironment and host immune cells but also can reactivate the genes silenced due to deacetylation and chromatin condensation. Hydroxamic acid hybrids as promising histone deacetylase inhibitors have the potential to address drug resistance and reduce severe side effects associated with a single drug molecule due to their capacity to simultaneously modulate multiple targets in cancer cells. Accordingly, rational design of hydroxamic acid hybrids may provide valuable therapeutic interventions for the treatment of breast cancer. This review aimed to provide insights into the in vitro and in vivo anti-breast cancer therapeutic potential of hydroxamic acid hybrids, together with their mechanisms of action and structure-activity relationships, covering articles published from 2020 to the present.
Collapse
Affiliation(s)
- Xinran Cao
- School of Pharmacy, University College London (UCL), London, WC1E 6BT, UK
| | - Yufeng Gong
- The Second Affiliated Hospital of Mudanjiang Medical University, Mudanjiang, 157011, China
| |
Collapse
|
26
|
Lakra DS, Bharathiraja P, Dhanalakshmi T, Prasad NR. Andrographolide reverts multidrug resistance in KBCh R 8-5 cells through AKT signaling pathway. Cell Biochem Funct 2024; 42:e3948. [PMID: 38379216 DOI: 10.1002/cbf.3948] [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: 11/17/2023] [Revised: 01/22/2024] [Accepted: 01/25/2024] [Indexed: 02/22/2024]
Abstract
Multidrug resistance (MDR) is a major obstacle in cancer chemotherapy. P-glycoprotein (P-gp) one of the ATP-binding cassette (ABC) transporters plays an important role in MDR. In this study, we examined the sensitizing property of andrographolide (Andro) to reverse MDR in the drug-resistant KBChR 8-5 cells. Andro exhibited increased cytotoxicity in a concentration-dependent manner in the P-gp overexpressing KBChR 8-5 cells. Furthermore, Andro showed synergistic interactions with PTX and DOX in this drug-resistant cells. Andro co-administration enhanced PTX- and DOX-induced cytotoxicity and reduced cell proliferation in the MDR cancer cells. Moreover, reactive oxygen species (ROS) were elevated with a decrease in the mitochondrial membrane potential (MMP) during Andro and chemotherapeutic drugs combination treatment in the drug-resistant cells. Furthermore, Andro and PTX-induced cell cycle arrest was observed in the drug-resistant cell. We also noticed that the expression of ABCB1 and AKT were downregulated during Andro (4 µM) treatment. Furthermore, Andro treatment enhanced the expression of caspase 3 and caspase 9 in the combinational groups that support the enhanced apoptotic cell death in drug-resistant cancer cells. Therefore, the results reveal that Andro plays a role in the reversal of P-gp-mediated MDR in KBChR 8-5 cells which might be due to regulating ABCB1/AKT signaling pathway.
Collapse
Affiliation(s)
- Deepa S Lakra
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - Pradhapsingh Bharathiraja
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
| | - T Dhanalakshmi
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
- Department of Biochemistry, Dharmapuram Gnanambigai Government Arts College for Women, Mayiladuthurai, Tamil Nadu, India
| | - N Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalainagar, Tamil Nadu, India
| |
Collapse
|
27
|
Dhanyamraju PK. Drug resistance mechanisms in cancers: Execution of pro-survival strategies. J Biomed Res 2024; 38:95-121. [PMID: 38413011 PMCID: PMC11001593 DOI: 10.7555/jbr.37.20230248] [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: 10/13/2023] [Revised: 11/21/2023] [Accepted: 12/07/2023] [Indexed: 02/29/2024] Open
Abstract
One of the quintessential challenges in cancer treatment is drug resistance. Several mechanisms of drug resistance have been described to date, and new modes of drug resistance continue to be discovered. The phenomenon of cancer drug resistance is now widespread, with approximately 90% of cancer-related deaths associated with drug resistance. Despite significant advances in the drug discovery process, the emergence of innate and acquired mechanisms of drug resistance has impeded the progress in cancer therapy. Therefore, understanding the mechanisms of drug resistance and the various pathways involved is integral to treatment modalities. In the present review, I discuss the different mechanisms of drug resistance in cancer cells, including DNA damage repair, epithelial to mesenchymal transition, inhibition of cell death, alteration of drug targets, inactivation of drugs, deregulation of cellular energetics, immune evasion, tumor-promoting inflammation, genome instability, and other contributing epigenetic factors. Furthermore, I highlight available treatment options and conclude with future directions.
Collapse
Affiliation(s)
- Pavan Kumar Dhanyamraju
- Fels Cancer Institute of Personalized Medicine, Lewis-Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
28
|
Sharma NK, Bahot A, Sekar G, Bansode M, Khunteta K, Sonar PV, Hebale A, Salokhe V, Sinha BK. Understanding Cancer's Defense against Topoisomerase-Active Drugs: A Comprehensive Review. Cancers (Basel) 2024; 16:680. [PMID: 38398072 PMCID: PMC10886629 DOI: 10.3390/cancers16040680] [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: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, the emergence of cancer drug resistance has been one of the crucial tumor hallmarks that are supported by the level of genetic heterogeneity and complexities at cellular levels. Oxidative stress, immune evasion, metabolic reprogramming, overexpression of ABC transporters, and stemness are among the several key contributing molecular and cellular response mechanisms. Topo-active drugs, e.g., doxorubicin and topotecan, are clinically active and are utilized extensively against a wide variety of human tumors and often result in the development of resistance and failure to therapy. Thus, there is an urgent need for an incremental and comprehensive understanding of mechanisms of cancer drug resistance specifically in the context of topo-active drugs. This review delves into the intricate mechanistic aspects of these intracellular and extracellular topo-active drug resistance mechanisms and explores the use of potential combinatorial approaches by utilizing various topo-active drugs and inhibitors of pathways involved in drug resistance. We believe that this review will help guide basic scientists, pre-clinicians, clinicians, and policymakers toward holistic and interdisciplinary strategies that transcend resistance, renewing optimism in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Anjali Bahot
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Gopinath Sekar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Mahima Bansode
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Kratika Khunteta
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Priyanka Vijay Sonar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Ameya Hebale
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Vaishnavi Salokhe
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Birandra Kumar Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
29
|
Avgoustakis K, Angelopoulou A. Biomaterial-Based Responsive Nanomedicines for Targeting Solid Tumor Microenvironments. Pharmaceutics 2024; 16:179. [PMID: 38399240 PMCID: PMC10892652 DOI: 10.3390/pharmaceutics16020179] [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: 12/12/2023] [Revised: 01/16/2024] [Accepted: 01/23/2024] [Indexed: 02/25/2024] Open
Abstract
Solid tumors are composed of a highly complex and heterogenic microenvironment, with increasing metabolic status. This environment plays a crucial role in the clinical therapeutic outcome of conventional treatments and innovative antitumor nanomedicines. Scientists have devoted great efforts to conquering the challenges of the tumor microenvironment (TME), in respect of effective drug accumulation and activity at the tumor site. The main focus is to overcome the obstacles of abnormal vasculature, dense stroma, extracellular matrix, hypoxia, and pH gradient acidosis. In this endeavor, nanomedicines that are targeting distinct features of TME have flourished; these aim to increase site specificity and achieve deep tumor penetration. Recently, research efforts have focused on the immune reprograming of TME in order to promote suppression of cancer stem cells and prevention of metastasis. Thereby, several nanomedicine therapeutics which have shown promise in preclinical studies have entered clinical trials or are already in clinical practice. Various novel strategies were employed in preclinical studies and clinical trials. Among them, nanomedicines based on biomaterials show great promise in improving the therapeutic efficacy, reducing side effects, and promoting synergistic activity for TME responsive targeting. In this review, we focused on the targeting mechanisms of nanomedicines in response to the microenvironment of solid tumors. We describe responsive nanomedicines which take advantage of biomaterials' properties to exploit the features of TME or overcome the obstacles posed by TME. The development of such systems has significantly advanced the application of biomaterials in combinational therapies and in immunotherapies for improved anticancer effectiveness.
Collapse
Affiliation(s)
- Konstantinos Avgoustakis
- Department of Pharmacy, School of Health Sciences, University of Patras, 26504 Patras, Greece;
- Clinical Studies Unit, Biomedical Research Foundation Academy of Athens (BRFAA), 4 Soranou Ephessiou Street, 11527 Athens, Greece
| | - Athina Angelopoulou
- Department of Chemical Engineering, Polytechnic School, University of Patras, 26504 Patras, Greece
| |
Collapse
|
30
|
Saleh RO, Al-Ouqaili MTS, Ali E, Alhajlah S, Kareem AH, Shakir MN, Alasheqi MQ, Mustafa YF, Alawadi A, Alsaalamy A. lncRNA-microRNA axis in cancer drug resistance: particular focus on signaling pathways. Med Oncol 2024; 41:52. [PMID: 38195957 DOI: 10.1007/s12032-023-02263-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 11/20/2023] [Indexed: 01/11/2024]
Abstract
Cancer drug resistance remains a formidable challenge in modern oncology, necessitating innovative therapeutic strategies. The convergence of intricate regulatory networks involving long non-coding RNAs, microRNAs, and pivotal signaling pathways has emerged as a crucial determinant of drug resistance. This review underscores the multifaceted roles of lncRNAs and miRNAs in orchestrating gene expression and cellular processes, mainly focusing on their interactions with specific signaling pathways. Dysregulation of these networks leads to the acquisition of drug resistance, dampening the efficacy of conventional treatments. The review highlights the potential therapeutic avenues unlocked by targeting these non-coding RNAs. Developing specific inhibitors or mimics for lncRNAs and miRNAs, alone or in combination with conventional chemotherapy, emerges as a promising strategy. In addition, epigenetic modulators, immunotherapies, and personalized medicine present exciting prospects in tackling drug resistance. While substantial progress has been made, challenges, including target validation and safety assessment, remain. The review emphasizes the need for continued research to overcome these hurdles and underscores the transformative potential of lncRNA-miRNA interplay in revolutionizing cancer therapy.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq.
| | - Mushtak T S Al-Ouqaili
- Department of Microbiology, College of Medicine, University of Anbar, Ramadi, Anbar, Iraq
| | - Eyhab Ali
- College of Chemistry, Al-Zahraa University for Women, Karbala, Iraq
| | - Sharif Alhajlah
- Department of Medical Laboratories, College of Applied Medical Sciences, Shaqra University, 11961, Shaqra, Saudi Arabia.
| | | | - Maha Noori Shakir
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Ahmed Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| |
Collapse
|
31
|
Laiolo J, Graikioti DG, Barbieri CL, Joray MB, Antoniou AI, Vera DMA, Athanassopoulos CM, Carpinella MC. Novel betulin derivatives as multidrug reversal agents targeting P-glycoprotein. Sci Rep 2024; 14:70. [PMID: 38167542 PMCID: PMC10762177 DOI: 10.1038/s41598-023-49939-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Chemotherapy is a powerful means of cancer treatment but its efficacy is compromised by the emergence of multidrug resistance (MDR), mainly linked to the efflux transporter ABCB1/P-glycoprotein (P-gp). Based on the chemical structure of betulin, identified in our previous work as an effective modulator of the P-gp function, a series of analogs were designed, synthesized and evaluated as a source of novel inhibitors. Compounds 6g and 6i inhibited rhodamine 123 efflux in the P-gp overexpressed leukemia cells, K562/Dox, at concentrations of 0.19 µM and 0.39 µM, respectively, and increased the intracellular accumulation of doxorubicin at the submicromolar concentration of 0.098 µM. Compounds 6g and 6i were able to restore the sensitivity of K562/Dox to Dox at 0.024 µM and 0.19 µM, respectively. Structure-activity relationship analysis and molecular modeling revealed important information about the structural features conferring activity. All the active compounds fitted in a specific region involving mainly transmembrane helices (TMH) 4-6 from one homologous half and TMH 7 and 12 from the other, also showing close contacts with TMH 6 and 12. Compounds that bound preferentially to another region were inactive, regardless of their free energy of binding. It should be noted that compounds 6g and 6i were devoid of toxic effects against peripheral blood mononuclear normal cells and erythrocytes. The data obtained indicates that both compounds might be proposed as scaffolds for obtaining promising P-gp inhibitors for overcoming MDR.
Collapse
Affiliation(s)
- Jerónimo Laiolo
- Fine Chemical and Natural Products Laboratory, IRNASUS CONICET-UCC, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Dafni G Graikioti
- Synthetic Organic Chemistry Laboratory, Department of Chemistry, University of Patras, 26504, Patras, Greece
| | - Cecilia L Barbieri
- Department of Chemistry and Biochemistry, College of Exact and Natural Sciences, Universidad Nacional de Mar del Plata - QUIAMM - INBIOTEC CONICET, Mar del Plata, Argentina
| | - Mariana B Joray
- Fine Chemical and Natural Products Laboratory, IRNASUS CONICET-UCC and CIDIE CONICET-UCC, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Antonia I Antoniou
- Synthetic Organic Chemistry Laboratory, Department of Chemistry, University of Patras, 26504, Patras, Greece
| | - D Mariano A Vera
- Department of Chemistry and Biochemistry, College of Exact and Natural Sciences, Universidad Nacional de Mar del Plata - QUIAMM - INBIOTEC CONICET, Mar del Plata, Argentina.
| | | | - María C Carpinella
- Fine Chemical and Natural Products Laboratory, IRNASUS CONICET-UCC and CIDIE CONICET-UCC, Universidad Católica de Córdoba, Córdoba, Argentina.
| |
Collapse
|
32
|
Kumari P, Dang S. Evaluation of Enhanced Cytotoxicity Effect of Repurposed Drug Simvastatin/Thymoquinone Combination against Breast Cancer Cell Line. Cardiovasc Hematol Agents Med Chem 2024; 22:348-366. [PMID: 37907488 DOI: 10.2174/0118715257259037231012182741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/02/2023] [Accepted: 09/15/2023] [Indexed: 11/02/2023]
Abstract
INTRODUCTION Repurposing of drugs for their anticancer potential is gaining a lot of importance in drug discovery. AIMS The present study aims to explore the potential of Simvastatin (SIM), a drug used in the treatment of high cholesterol, and Thymoquinone (Nigella Sativa) (THY) for its anti-cancer activity on breast cancer cell lines. Thymoquinone is reported to have many potential medicinal properties exhibiting antioxidant, antiinflammatory, anti-cancer, activities like inhibition of tissue growth and division. METHODS In this analysis, we explored the inhibitory effects of the combination of Simvastatin ad Thymoquinone on two breast cancer cell lines viz MCF-7 and MDA-MB-231 cells. The combined effect of Simvastatin and Thymoquinone on Cell viability, Colony formation, Cell migration, and orientation of more programmed cell death in vitro was studied. Cell cycle arrest in the G2/M phase was concomitant with the combined effect of SIM and THY persuading apoptosis and generating reactive oxygen species (ROS). RESULTS The cell cycle arrest with combined treatment was observed that only 1.8% and 1.1% cells gated in G2/M phase in MCF-7 & MDA-MB-231 cell. An increased apoptosis was observed when cells were treated in combination which was about 76.20% and 58.15% respectively for MCF-7 and MDA-MB-231 cells. CONCLUSION It was concluded that the combined effect of simvastatin and thymoquinone stimulates apoptosis in breast cancer cells.
Collapse
Affiliation(s)
- Pallavi Kumari
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Noida, Sector 62, U.P., 201309, India
| | - Shweta Dang
- Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Noida, Sector 62, U.P., 201309, India
| |
Collapse
|
33
|
Wilczak M, Surman M, Przybyło M. The Role of Intracellular and Extracellular Vesicles in the Development of Therapy Resistance in Cancer. Curr Pharm Des 2024; 30:2765-2784. [PMID: 39113303 DOI: 10.2174/0113816128326325240723051625] [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: 04/26/2024] [Accepted: 06/19/2024] [Indexed: 10/22/2024]
Abstract
Cancer is the second leading cause of global mortality and claims approximately 10 million lives annually. Despite advances in treatments such as surgery, chemotherapy, and immunotherapy, resistance to these methods has emerged. Multidrug resistance (MDR), where cancer cells resist diverse treatments, undermines therapy effectiveness, escalating mortality rates. MDR mechanisms include, among others, drug inactivation, reduced drug uptake, enhanced DNA repair, and activation of survival pathways such as autophagy. Moreover, MDR mechanisms can confer resistance to other therapies like radiotherapy. Understanding these mechanisms is crucial for improving treatment efficacy and identifying new therapeutic targets. Extracellular vesicles (EVs) have gathered attention for their role in cancer progression, including MDR development through protein transfer and genetic reprogramming. Autophagy, a process balancing cellular resources, also influences MDR. The intersection of EVs and autophagy further complicates the understanding of MDR. Both extracellular (exosomes, microvesicles) and intracellular (autophagic) vesicles contribute to therapy resistance by regulating the tumor microenvironment, facilitating cell communication, and modulating drug processing. While much is known about these pathways, there is still a need to explore their potential for predicting treatment responses and understanding tumor heterogeneity.
Collapse
Affiliation(s)
- Magdalena Wilczak
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Krakow, Poland
| | - Magdalena Surman
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| | - Małgorzata Przybyło
- Department of Glycoconjugate Biochemistry, Faculty of Biology, Institute of Zoology and Biomedical Research, Jagiellonian University, Krakow, Poland
| |
Collapse
|
34
|
Lima EN, Lamichhane S, KC P, Ferreira ES, Koul S, Koul HK. Tetrandrine for Targeting Therapy Resistance in Cancer. Curr Top Med Chem 2024; 24:1035-1049. [PMID: 38445699 PMCID: PMC11259026 DOI: 10.2174/0115680266282360240222062032] [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: 09/14/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 03/07/2024]
Abstract
During the last five decades, there has been tremendous development in our understanding of cancer biology and the development of new and novel therapeutics to target cancer. However, despite these advances, cancer remains the second leading cause of death across the globe. Most cancer deaths are attributed to the development of resistance to current therapies. There is an urgent and unmet need to address cancer therapy resistance. Tetrandrine, a bis-benzyl iso-quinoline, has shown a promising role as an anti-cancer agent. Recent work from our laboratory and others suggests that tetrandrine and its derivatives could be an excellent adjuvant to the current arsenal of anti-cancer drugs. Herein, we provide an overview of resistance mechanisms to current therapeutics and review the existing literature on the anti-cancer effects of tetrandrine and its potential use for overcoming therapy resistance in cancer.
Collapse
Affiliation(s)
- Ellen Nogueira Lima
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Southeast Louisiana Veterans Health Care System, New Orleans – LA
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Santosh Lamichhane
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Southeast Louisiana Veterans Health Care System, New Orleans – LA
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Pramod KC
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Elisa Silva Ferreira
- Brazilian Nanotechnology National Laboratory (LNNano) Brazilian Center for Research in Energy and Materials (CNPEM) Campinas, SP, Brazil
| | - Sweaty Koul
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Department of Urology, LSUHSC-New Orleans
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Hari K Koul
- Department of Interdisciplinary Oncology, LSUHSC-New Orleans
- Department of Biochemistry & Molecular Biology, LSUHSC-New Orleans
- Department of Urology, LSUHSC-New Orleans
- Southeast Louisiana Veterans Health Care System, New Orleans – LA
- LSU-LCMC Cancer Center, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
35
|
George DC, Bertrand FE, Sigounas G. Notch-3 affects chemoresistance in colorectal cancer via DNA base excision repair enzymes. Adv Biol Regul 2024; 91:101013. [PMID: 38290285 DOI: 10.1016/j.jbior.2024.101013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/21/2023] [Accepted: 01/08/2024] [Indexed: 02/01/2024]
Abstract
Colon cancer is the second leading cause of cancer death. With over 153,000 new CRC cases predicted, it is the third most commonly diagnosed cancer. Early detection can lead to curative surgical intervention, but recurrent and late metastatic disease is frequently treated with chemotherapeutic options based on induction of DNA damage. Understanding mechanism(s) that regulate DNA damage repair within colon tumor cells is essential to developing effective therapeutic strategies. The Notch signaling pathway is known to participate in normal colon development and we have recently described a pathway by which Notch-1, Notch-3 and Smad may regulated EMT and stem-like properties in colon tumor cells, promoting tumorigenesis. Little is known about how Notch may regulate drug resistance. In this study, we used shRNA to generate colon tumor cells with loss of Notch-3 expression. These cells exhibited reduced expression of the base-excision repair proteins PARP1 and APE1, along with increased sensitivity to ara-c and cisplatin. These data point to a pathway in which Notch-3 signaling can regulate DNA repair within colon tumor cells and suggests that targeting Notch-3 may be an effective approach to rendering colon tumors sensitive to chemotherapeutic drugs.
Collapse
Affiliation(s)
- Dennis C George
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Fred E Bertrand
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - George Sigounas
- Department of Internal Medicine, Division of Hematology/Oncology, Brody School of Medicine, East Carolina University, Greenville, NC, USA.
| |
Collapse
|
36
|
Salmani-Javan E, Farhoudi Sefidan Jadid M, Zarghami N. Recent advances in molecular targeted therapy of lung cancer: Possible application in translation medicine. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2024; 27:122-133. [PMID: 38234663 PMCID: PMC10790298 DOI: 10.22038/ijbms.2023.72407.15749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/23/2023] [Indexed: 01/19/2024]
Abstract
Lung cancer is one of the leading causes of death among all cancer deaths. This cancer is classified into two different histological subtypes: non-small cell lung cancer (NSCLC), which is the most common subtype, and small cell lung cancer (SCLC), which is the most aggressive subtype. Understanding the molecular characteristics of lung cancer has expanded our knowledge of the cellular origins and molecular pathways affected by each of these subtypes and has contributed to the development of new therapies. Traditional treatments for lung cancer include surgery, chemotherapy, and radiotherapy. Advances in understanding the nature and specificity of lung cancer have led to the development of immunotherapy, which is the newest and most specialized treatment in the treatment of lung cancer. Each of these treatments has advantages and disadvantages and causes side effects. Today, combination therapy for lung cancer reduces side effects and increases the speed of recovery. Despite the significant progress that has been made in the treatment of lung cancer in the last decade, further research into new drugs and combination therapies is needed to extend the clinical benefits and improve outcomes in lung cancer. In this review article, we discussed common lung cancer treatments and their combinations from the most advanced to the newest.
Collapse
Affiliation(s)
- Elnaz Salmani-Javan
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Farhoudi Sefidan Jadid
- Department of Clinical Biochemistry and Laboratory Medicine, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Aydin University, Istanbul, Turkey
| |
Collapse
|
37
|
Rezakhani L, Salmani S, Eliyasi Dashtaki M, Ghasemi S. Resveratrol: Targeting Cancer Stem Cells and ncRNAs to Overcome Cancer Drug Resistance. Curr Mol Med 2024; 24:951-961. [PMID: 37592772 DOI: 10.2174/1566524023666230817102114] [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: 02/18/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 08/19/2023]
Abstract
A major challenge in treating cancer is the development of drug resistance, which can result in treatment failure and tumor recurrence. Targeting cancer stem cells (CSCs) and non-coding RNAs (ncRNAs) with a polyphenolic substance called resveratrol has the ability to combat this problem by lowering cancer resistance to drugs and opening up new therapeutic options. Resveratrol alters the expression of genes related to self-renewal, modulating important signaling pathways involved in cancer initiation and CSC control. Additionally, resveratrol affects non-coding RNAs (ncRNAs), including Micro-RNAs (miRNAs) and long non-coding RNAs (lncRNAs which are essential for stemness, drug resistance, and other cancer-related activities. Numerous studies have shown that resveratrol has the potential to be an effective anticancer drug when used in combination therapy, but issues with absorption and pharmacokinetics still need to be resolved before it can be used in clinical applications. Reducing chemotherapy resistance by better understanding the intricate mechanisms by which resveratrol affects cancer cells and CSCs, as well as its impact on ncRNA expression, could eventually contribute to more effective cancer treatments. To completely understand these pathways and optimize the utilization of resveratrol in combination treatments, additional study is necessary.
Collapse
Affiliation(s)
- Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sima Salmani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Masoumeh Eliyasi Dashtaki
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sorayya Ghasemi
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
38
|
Basu SM, Chauhan M, Giri J. pH-Responsive Polypropylene Sulfide Magnetic Nanocarrier-Mediated Chemo-Hyperthermia Kills Breast Cancer Stem Cells by Long-Term Reversal of Multidrug Resistance and Chemotherapy Resensitization. ACS APPLIED MATERIALS & INTERFACES 2023; 15:58151-58165. [PMID: 38063494 DOI: 10.1021/acsami.3c12303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
Cancer stem cells (CSCs) present a formidable challenge in cancer treatment due to their inherent resistance to chemotherapy, primarily driven by the overexpression of ABC transporters and multidrug resistance (MDR). Despite extensive research on pharmacological small-molecule inhibitors, effectively managing MDR and improving chemotherapeutic outcomes remain elusive. On the other hand, magnetic hyperthermia (MHT) holds great promise as a cancer therapeutic, but there is limited research on its potential to reverse MDR in breast CSCs and effectively eliminate CSCs through combined chemo-hyperthermia. To address these gaps, we developed tumor microenvironment-sensitive, drug-loaded poly(propylene sulfide) (PPS)-coated magnetic nanoparticles (PPS-MnFe). These nanoparticles were employed to investigate hyperthermia sensitivity and MDR reversion in breast CSCs, comparing their performance to that of small-molecule inhibitors. Additionally, we explored the efficacy of combined chemo-hyperthermia in killing CSCs. CSC-enriched breast cancer cells were subjected to low-dose MHT at 42 °C for 30 min and then treated with the chemical MDR inhibitor salinomycin (SAL). The effectiveness of each treatment in inhibiting MDR was assessed by measuring the efflux of the MDR substrate, rhodamine 123 (R123) dye. Notably, MHT induced a prolonged reversal of MDR activity compared with SAL treatment alone. After successfully inhibiting MDR, the breast CSCs were exposed to chemotherapy using paclitaxel to trigger synergistic cell death. The combination of MHT and chemotherapy demonstrated remarkable reductions in stemness properties, MDR reversal, and the effective eradication of breast CSCs in this innovative dual-modality approach.
Collapse
Affiliation(s)
- Suparna Mercy Basu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana 502285, India
| | - Meenakshi Chauhan
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana 502285, India
| | - Jyotsnendu Giri
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Telangana 502285, India
| |
Collapse
|
39
|
Gupta U, Maity D, Sharma VK. Recent advances of polymeric nanoplatforms for cancer treatment: smart delivery systems (SDS), nanotheranostics and multidrug resistance (MDR) inhibition. Biomed Mater 2023; 19:012003. [PMID: 37944188 DOI: 10.1088/1748-605x/ad0b23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/09/2023] [Indexed: 11/12/2023]
Abstract
Nanotheranostics is a promising field that combines the benefits of diagnostic and treatment into a single nano-platform that not only administers treatment but also allows for real-time monitoring of therapeutic response, decreasing the possibility of under/over-drug dosing. Furthermore, developing smart delivery systems (SDSs) for cancer theranostics that can take advantage of various tumour microenvironment (TME) conditions (such as deformed tumour vasculature, various over-expressed receptor proteins, reduced pH, oxidative stress, and resulting elevated glutathione levels) can aid in achieving improved pharmacokinetics, higher tumour accumulation, enhanced antitumour efficacy, and/or decreased side effects and multidrug resistance (MDR) inhibition. Polymeric nanoparticles (PNPs) are being widely investigated in this regard due to their unique features such as small size, passive/active targeting possibility, better pharmaceutical kinetics and biological distribution, decreased adverse reactions of the established drugs, inherent inhibitory properties to MDR efflux pump proteins, as well as the feasibility of delivering numerous therapeutic substances in just one design. Hence in this review, we have primarily discussed PNPs based targeted and/or controlled SDSs in which we have elaborated upon different TME mediated nanotheranostic platforms (NTPs) including active/passive/magnetic targeting platforms along with pH/ROS/redox-responsive platforms. Besides, we have elucidated different imaging guided cancer therapeutic platforms based on four major cancer imaging techniques i.e., fluorescence/photo-acoustic/radionuclide/magnetic resonance imaging, Furthermore, we have deliberated some of the most recently developed PNPs based multimodal NTPs (by combining two or more imaging or therapy techniques on a single nanoplatform) in cancer theranostics. Moreover, we have provided a brief update on PNPs based NTP which are recently developed to overcome MDR for effective cancer treatment. Additionally, we have briefly discussed about the tissue biodistribution/tumour targeting efficiency of these nanoplatforms along with recent preclinical/clinical studies. Finally, we have elaborated on various limitations associated with PNPs based nanoplatforms.
Collapse
Affiliation(s)
- Urvashi Gupta
- Department of Bioengineering, Imperial College London, London SW7 2BX, United Kingdom
| | - Dipak Maity
- School of Health Sciences & Technology, University of Petroleum and Energy Studies, Dehradun, Uttarakhand 248007, India
| | - Virender K Sharma
- Program for the Environment and Sustainability, Department of Environmental and Occupational Health, School of Public Health, Texas A&M University, 1266 TAMU, College Station, TX 77843, United States of America
| |
Collapse
|
40
|
Aleksandrova Y, Munkuev A, Mozhaitsev E, Suslov E, Volcho K, Salakhutdinov N, Neganova M. Hydroxamic Acids Containing a Bicyclic Pinane Backbone as Epigenetic and Metabolic Regulators: Synergizing Agents to Overcome Cisplatin Resistance. Cancers (Basel) 2023; 15:4985. [PMID: 37894352 PMCID: PMC10605847 DOI: 10.3390/cancers15204985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/10/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Multidrug resistance is the dominant obstacle to effective chemotherapy for malignant neoplasms. It is well known that neoplastic cells use a wide range of adaptive mechanisms to form and maintain resistance against antitumor agents, which makes it urgent to identify promising therapies to solve this problem. Hydroxamic acids are biologically active compounds and in recent years have been actively considered to be potentially promising drugs of various pharmacological applications. In this paper, we synthesized a number of hydroxamic acids containing a p-substituted cinnamic acid core and bearing bicyclic pinane fragments, including derivatives of (-)-myrtenol, (+)-myrtenol and (-)-nopol, as a Cap-group. Among the synthesized compounds, the most promising hydroxamic acid was identified, containing a fragment of (-)-nopol in the Cap group 18c. This compound synergizes with cisplatin to increase its anticancer effect and overcomes cisplatin resistance, which may be associated with the inhibition of histone deacetylase 1 and glycolytic function. Taken together, our results demonstrate that the use of hydroxamic acids with a bicyclic pinane backbone can be considered to be an effective approach to the eradication of tumor cells and overcoming drug resistance in the treatment of malignant neoplasms.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, 142432 Chernogolovka, Russia;
| | - Aldar Munkuev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia; (A.M.); (E.M.); (E.S.); (K.V.); (N.S.)
| | - Evgenii Mozhaitsev
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia; (A.M.); (E.M.); (E.S.); (K.V.); (N.S.)
| | - Evgeniy Suslov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia; (A.M.); (E.M.); (E.S.); (K.V.); (N.S.)
| | - Konstantin Volcho
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia; (A.M.); (E.M.); (E.S.); (K.V.); (N.S.)
| | - Nariman Salakhutdinov
- Department of Medicinal Chemistry, N. N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch, Russian Academy of Sciences, Lavrentiev Ave., 9, 630090 Novosibirsk, Russia; (A.M.); (E.M.); (E.S.); (K.V.); (N.S.)
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, Severnij Pr. 1, 142432 Chernogolovka, Russia;
| |
Collapse
|
41
|
Seydi H, Nouri K, Rezaei N, Tamimi A, Hassan M, Mirzaei H, Vosough M. Autophagy orchestrates resistance in hepatocellular carcinoma cells. Biomed Pharmacother 2023; 161:114487. [PMID: 36963361 DOI: 10.1016/j.biopha.2023.114487] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/26/2023] Open
Abstract
Treatment resistance is one of the major barriers for therapeutic strategies in hepatocellular carcinoma (HCC). Many studies have indicated that chemotherapy and radiotherapy induce autophagy machinery (cell protective autophagy) in HCC cells. In addition, many experiments report a remarkable crosstalk between treatment resistance and autophagy pathways. Thus, autophagy could be one of the key factors enabling tumor cells to hinder induced cell death after medical interventions. Therefore, extensive research on the molecular pathways involved in resistance induction and autophagy have been conducted to achieve the desired therapeutic response. The key molecular pathways related to the therapy resistance are TGF-β, MAPK, NRF2, NF-κB, and non-coding RNAs. In addition, EMT, drug transports, apoptosis evasion, DNA repair, cancer stem cells, and hypoxia could have considerable impact on the hepatoma cell's response to therapies. These mechanisms protect tumor cells against various treatments and many studies have shown that each of them is connected to the molecular pathways of autophagy induction in HCC. Hence, autophagy inhibition may be an effective strategy to improve therapeutic outcome in HCC patients. In this review, we further highlight how autophagy leads to poor response during treatment through a complex molecular network and how it enhances resistance in primary liver cancer. We propose that combinational regimens of approved HCC therapeutic protocols plus autophagy inhibitors may overcome drug resistance in HCC therapy.
Collapse
Affiliation(s)
- Homeyra Seydi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran
| | - Kosar Nouri
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran
| | - Niloufar Rezaei
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran; Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran, Islamic Republic of Iran
| | - Atena Tamimi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Islamic Republic of Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
42
|
Nunes M, Duarte D, Vale N, Ricardo S. The Antineoplastic Effect of Carboplatin Is Potentiated by Combination with Pitavastatin or Metformin in a Chemoresistant High-Grade Serous Carcinoma Cell Line. Int J Mol Sci 2022; 24:ijms24010097. [PMID: 36613537 PMCID: PMC9820586 DOI: 10.3390/ijms24010097] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
The combination of Carboplatin with Paclitaxel is the mainstay treatment for high-grade serous carcinoma; however, many patients with advanced disease undergo relapse due to chemoresistance. Drug repurposing coupled with a combination of two or more compounds with independent mechanisms of action has the potential to increase the success rate of the antineoplastic treatment. The purpose of this study was to explore whether the combination of Carboplatin with repurposed drugs led to a therapeutic benefit. Hence, we assessed the cytotoxic effects of Carboplatin alone and in combination with several repurposed drugs (Pitavastatin, Metformin, Ivermectin, Itraconazole and Alendronate) in two tumoral models, i.e., Carboplatin (OVCAR8) and Carboplatin-Paclitaxel (OVCAR8 PTX R P) chemoresistant cell lines and in a non-tumoral (HOSE6.3) cell line. Cellular viability was measured using the Presto Blue assay, and the synergistic interactions were evaluated using the Chou-Talalay, Bliss Independence and Highest Single Agent reference models. Combining Carboplatin with Pitavastatin or Metformin displayed the highest cytotoxic effect and the strongest synergism among all combinations for OVCAR8 PTX R P cells, resulting in a chemotherapeutic effect superior to Carboplatin as a single agent. Concerning HOSE6.3 cells, combining Carboplatin with almost all the repurposed drugs demonstrated a safe pharmacological profile. Overall, we propose that Pitavastatin or Metformin could act synergistically in combination with Carboplatin for the management of high-grade serous carcinoma patients with a Carboplatin plus Paclitaxel resistance profile.
Collapse
Affiliation(s)
- Mariana Nunes
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, 4050-313 Porto, Portugal
| | - Diana Duarte
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal
- Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- Department of Community Medicine, Health Information and Decision (MEDCIDS), Faculty of Medicine, University of Porto, 4200-450 Porto, Portugal
| | - Sara Ricardo
- Differentiation and Cancer Group, Institute for Research and Innovation in Health (i3S) of the University of Porto, 4200-135 Porto, Portugal
- Toxicology Research Unit (TOXRUN), University Institute of Health Sciences, Polytechnic and University Cooperative (CESPU), 4585-116 Gandra, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
- Correspondence:
| |
Collapse
|
43
|
Mishra SK, Dhadve AC, Mal A, Reddy BPK, Hole A, Chilakapati MK, Ray P, Srivastava R, De A. Photothermal therapy (PTT) is an effective treatment measure against solid tumors which fails to respond conventional chemo/radiation therapies in clinic. BIOMATERIALS ADVANCES 2022; 143:213153. [PMID: 36343390 DOI: 10.1016/j.bioadv.2022.213153] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 10/06/2022] [Accepted: 10/09/2022] [Indexed: 06/16/2023]
Abstract
Photothermal therapy (PTT) has emerged as a fast, precisive, and cost-effective anticancer therapy protocol. Here we applied our previously designed nanomaterial (Tocophotoxil) for prospective PTT application to manage radiation- and chemo-resistant cancers in a preclinical model. A PTT dose vs. efficacy relationship was established for radioresistant breast (ZR-75-1 50Gy, 4T1 20Gy) and chemo-resistant ovarian (A2780LR) cancer cells and tumors in mice models. Compared to the sensitive cases, resistant cells treated with PTT for a shorter duration show higher endurance. However, preclinical tumor xenografts treated with optimal PTT dose show 2-3 fold higher longevity (P ≤ 0.05) of treated mice monitored by non-invasive imaging methods. Elevated ERK and AKT activation in radioresistant or only AKT activation in chemo-resistant cells were contributory to higher cell survival in sub-optimal PTT dose. A comprehensive single-cell Raman map of PTT treated ZR-75-1 cell reveals broad-spectrum macromolecular deformities, including protein damage features. Marked induction of pJNK, unfolded protein response (UPR) pathway, increased reactive oxygen species (ROS), and lipid peroxidation in PTT-treated cells disrupted the intracellular homeostasis. Analyzing cellular ultrastructure, the coexistence of swollen endoplasmic reticulum, and autophagic bodies after PTT indicate possible coordination between UPR and autophagy pathways. Therefore, this comprehensive study provides new evidence on the potential impact of PTT as a standalone therapy for ablation of failed conventional therapy-resistant cancers in vivo, the success of which is intricately linked to the PTT dose optimization. The study, for the first time, also illustrates that under PTT treatment, concerted action of novel molecular switches such as JNK activation and UPR activation plays a vital role in triggering autophagy and cancer cell death.
Collapse
Affiliation(s)
- Sumit K Mishra
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India
| | - Ajit C Dhadve
- Imaging Cell Signaling and Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India
| | - Arijit Mal
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India
| | - B Pradeep K Reddy
- NanoBios Lab, Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India
| | - Arti Hole
- Chilakapati Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India
| | - Murali Krishna Chilakapati
- Chilakapati Laboratory, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India.
| | - Pritha Ray
- Imaging Cell Signaling and Therapeutics Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India.
| | - Rohit Srivastava
- NanoBios Lab, Department of Bioscience and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai, India.
| | - Abhijit De
- Molecular Functional Imaging Lab, Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Kharghar, Navi Mumbai, India; Department of Life Sciences, Homi Bhaba National Institute, Mumbai, India.
| |
Collapse
|
44
|
Ding P, Gao Y, Wang J, Xiang H, Zhang C, Wang L, Ji G, Wu T. Progress and challenges of multidrug resistance proteins in diseases. Am J Cancer Res 2022; 12:4483-4501. [PMID: 36381332 PMCID: PMC9641395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 10/09/2022] [Indexed: 06/16/2023] Open
Abstract
Chemotherapy remains the first choice for patients with advanced cancers when other treatments are ineffective. Multidrug resistance (MDR) is an unavoidable factor that negatively affects the effectiveness of cancer chemotherapy drugs. Researchers are trying to reduce MDR, improve the effectiveness of chemotherapeutic drugs, and alleviate patient suffering to positively contribute to disease treatment. MDR also occurs in inflammation and genetic disorders, which increases the difficulty of clinically beneficial treatments. The ATP-binding cassette (ABC) is an active transporter that plays an important role in the barrier and secretory functions of many normal cells. As the C subfamily in the ABC family, multidrug resistance proteins (MRPs/ABCCs) export a variety of antitumour drugs and are expressed in a variety of cancers. The present review summarises the role of MRPs in cancer and other diseases and recent research progress of MRP inhibitors to better examine the mechanism and function of MRPs, and establish a good relationship with clinical treatment.
Collapse
Affiliation(s)
- Peilun Ding
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Ying Gao
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Junmin Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Hongjiao Xiang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Caiyun Zhang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| | - Lei Wang
- Department of Hepatology, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Guang Ji
- Institute of Digestive Disease, Longhua Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200032, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese MedicineShanghai 201203, China
| |
Collapse
|
45
|
Gupta S, Verma V, Dwarakanath BS. Emerging concepts in cancer therapy: Mechanisms of resistance. Cancer Rep (Hoboken) 2022; 5:e1715. [PMID: 36083019 PMCID: PMC9780417 DOI: 10.1002/cnr2.1715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/30/2022] [Indexed: 01/25/2023] Open
Affiliation(s)
- Seema Gupta
- The Loop Immuno‐Oncology Laboratory, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
| | - Vivek Verma
- The Loop Immuno‐Oncology Laboratory, Lombardi Comprehensive Cancer CenterGeorgetown University Medical CenterWashingtonDistrict of ColumbiaUSA
- The Hormel Institute, Masonic Cancer CenterUniversity of MinnesotaAustinMinnesotaUSA
| | - Bilikere S. Dwarakanath
- Central Research FacilitySri Ramachandra Institute of Higher Education and Research InstituteChennaiIndia
| |
Collapse
|
46
|
Abdelaal MR, Ibrahim E, Elnagar MR, Soror SH, Haffez H. Augmented Therapeutic Potential of EC-Synthetic Retinoids in Caco-2 Cancer Cells Using an In Vitro Approach. Int J Mol Sci 2022; 23:ijms23169442. [PMID: 36012706 PMCID: PMC9409216 DOI: 10.3390/ijms23169442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 07/29/2022] [Accepted: 08/05/2022] [Indexed: 11/16/2022] Open
Abstract
Colorectal cancer therapies have produced promising clinical responses, but tumor cells rapidly develop resistance to these drugs. It has been previously shown that EC19 and EC23, two EC-synthetic retinoids, have single-agent preclinical anticancer activity in colorectal carcinoma. Here, isobologram analysis revealed that they have synergistic cytotoxicity with retinoic acid receptor (RAR) isoform-selective agonistic retinoids such as AC261066 (RARβ2-selective agonist) and CD437 (RARγ-selective agonist) in Caco-2 cells. This synergism was confirmed by calculating the combination index (lower than 1) and the dose reduction index (higher than 1). Flow cytometry of combinatorial IC50 (the concentration causing 50% cell death) confirmed the cell cycle arrest at the SubG0-G1 phase with potentiated apoptotic and necrotic effects. The reported synergistic anticancer activity can be attributed to their ability to reduce the expression of ATP-binding cassette (ABC) transporters including P-glycoprotein (P-gp1), breast cancer resistance protein (BCRP) and multi-drug resistance-associated protein-1 (MRP1) and Heat Shock Protein 70 (Hsp70). This adds up to the apoptosis-promoting activity of EC19 and EC23, as shown by the increased Caspase-3/7 activities and DNA fragmentation leading to DNA double-strand breaks. This study sheds the light on the possible use of EC-synthetic retinoids in the rescue of multi-drug resistance in colorectal cancer using Caco-2 as a model and suggests new promising combinations between different synthetic retinoids. The current in vitro results pave the way for future studies on these compounds as possible cures for colorectal carcinoma.
Collapse
Affiliation(s)
- Mohamed R. Abdelaal
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Esraa Ibrahim
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Mohamed R. Elnagar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Al-Azhar University, Cairo 11823, Egypt
| | - Sameh H. Soror
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
| | - Hesham Haffez
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Helwan University, Cairo 11795, Egypt
- Center of Scientific Excellence “Helwan Structural Biology Research, (HSBR)”, Helwan University, Cairo 11795, Egypt
- Correspondence: ; Tel.: +20-1094-970-173
| |
Collapse
|
47
|
Curcio M, Vittorio O, Bell JL, Iemma F, Nicoletta FP, Cirillo G. Hyaluronic Acid within Self-Assembling Nanoparticles: Endless Possibilities for Targeted Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:nano12162851. [PMID: 36014715 PMCID: PMC9413373 DOI: 10.3390/nano12162851] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/03/2022] [Accepted: 08/16/2022] [Indexed: 05/27/2023]
Abstract
Self-assembling nanoparticles (SANPs) based on hyaluronic acid (HA) represent unique tools in cancer therapy because they combine the HA targeting activity towards cancer cells with the advantageous features of the self-assembling nanosystems, i.e., chemical versatility and ease of preparation and scalability. This review describes the key outcomes arising from the combination of HA and SANPs, focusing on nanomaterials where HA and/or HA-derivatives are inserted within the self-assembling nanostructure. We elucidate the different HA derivatization strategies proposed for this scope, as well as the preparation methods used for the fabrication of the delivery device. After showing the biological results in the employed in vivo and in vitro models, we discussed the pros and cons of each nanosystem, opening a discussion on which approach represents the most promising strategy for further investigation and effective therapeutic protocol development.
Collapse
Affiliation(s)
- Manuela Curcio
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jessica Lilian Bell
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sidney, NSW 2052, Australia
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Francesca Iemma
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Fiore Pasquale Nicoletta
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy
| |
Collapse
|
48
|
Islas JF, Quiroz-Reyes AG, Delgado-Gonzalez P, Franco-Villarreal H, Delgado-Gallegos JL, Garza-Treviño EN, Gonzalez-Villarreal CA. Cancer Stem Cells in Tumor Microenvironment of Adenocarcinoma of the Stomach, Colon, and Rectum. Cancers (Basel) 2022; 14:3948. [PMID: 36010940 PMCID: PMC9405851 DOI: 10.3390/cancers14163948] [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: 06/30/2022] [Revised: 07/29/2022] [Accepted: 08/09/2022] [Indexed: 11/17/2022] Open
Abstract
Gastrointestinal adenocarcinomas are one of the world's deadliest cancers. Cancer stem cells and the tissue microenvironment are highly regulated by cell and molecular mechanisms. Cancer stem cells are essential for maintenance and progression and are associated with resistance to conventional treatments. This article reviews the current knowledge of the role of the microenvironment during the primary establishment of gastrointestinal adenocarcinomas in the stomach, colon, and rectum and its relationship with cancer stem cells. We also describe novel developments in cancer therapeutics, such as targeted therapy, and discuss the advantages and disadvantages of different treatments for improving gastrointestinal cancer prognosis.
Collapse
Affiliation(s)
- Jose Francisco Islas
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Adriana G. Quiroz-Reyes
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Paulina Delgado-Gonzalez
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | | | - Juan Luis Delgado-Gallegos
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | - Elsa N. Garza-Treviño
- Biochemistry and Molecular Medicine Department, School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey 64460, Mexico
| | | |
Collapse
|
49
|
Find new channel for overcoming chemoresistance in cancers: Role of stem cells-derived exosomal microRNAs. Int J Biol Macromol 2022; 219:530-537. [PMID: 35948201 DOI: 10.1016/j.ijbiomac.2022.07.253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 12/16/2022]
|
50
|
Karthika C, Sureshkumar R, Zehravi M, Akter R, Ali F, Ramproshad S, Mondal B, Tagde P, Ahmed Z, Khan FS, Rahman MH, Cavalu S. Multidrug Resistance of Cancer Cells and the Vital Role of P-Glycoprotein. Life (Basel) 2022; 12:897. [PMID: 35743927 PMCID: PMC9227591 DOI: 10.3390/life12060897] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/08/2022] [Accepted: 06/13/2022] [Indexed: 12/12/2022] Open
Abstract
P-glycoprotein (P-gp) is a major factor in the multidrug resistance phenotype in cancer cells. P-gp is a protein that regulates the ATP-dependent efflux of a wide range of anticancer medicines and confers resistance. Due to its wide specificity, several attempts have been made to block the action of P-gp to restore the efficacy of anticancer drugs. The major goal has been to create molecules that either compete with anticancer medicines for transport or function as a direct P-gp inhibitor. Despite significant in vitro success, there are presently no drugs available in the clinic that can "block" P-gp-mediated resistance. Toxicity, unfavourable pharmacological interactions, and a variety of pharmacokinetic difficulties might all be the reason for the failure. On the other hand, P-gp has a significant effect in the body. It protects the vital organs from the entry of foreign bodies and other toxic chemicals. Hence, the inhibitors of P-gp should not hinder its action in the normal cells. To develop an effective inhibitor of P-gp, thorough background knowledge is needed in this field. The main aim of this review article was to set forth the merits and demerits of the action of P-gp on cancer cells as well as on normal cells. The influence of P-gp on cancer drug delivery and the contribution of P-gp to activating drug resistance were also mentioned.
Collapse
Affiliation(s)
- Chenmala Karthika
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Raman Sureshkumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty 643001, Tamil Nadu, India;
| | - Mehrukh Zehravi
- Department of Clinical Pharmacy Girls Section, Prince Sattam Bin Abdul Aziz University Alkharj, Alkharj 11942, Saudi Arabia;
| | - Rokeya Akter
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Faraat Ali
- Department of Licensing and Enforcement, Laboratory Services, Botswana Medicines Regulatory Authority (BoMRA), Gaborone 999106, Botswana;
| | - Sarker Ramproshad
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Banani Mondal
- Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj 1400, Bangladesh; (S.R.); (B.M.)
| | - Priti Tagde
- Amity Institute of Pharmacy, Amity University, Noida 201303, Uttar Pradesh, India;
| | - Zubair Ahmed
- Unit of Bee Research and Honey Production, Faculty of Science, King Khalid University, Abha 61413, Saudi Arabia;
- Research Center for Advanced Materials Science (RCAMS), King Khalid University, Abha 61413, Saudi Arabia
- Mahala Campus, Community College, King Khalid University, Abha 61413, Saudi Arabia
| | - Farhat S. Khan
- Biology Department, Faculty of Sciences and Arts, King Khalid University, Dhahran Al Janoub, Abha 61413, Saudi Arabia;
| | - Md. Habibur Rahman
- Department of Global Medical Science, Wonju College of Medicine, Yonsei University, Wonju 26426, Gangwon-do, Korea;
| | - Simona Cavalu
- Faculty of Medicine and Pharmacy, University of Oradea, P-ta 1 Decembrie 10, 410087 Oradea, Romania
| |
Collapse
|