1
|
Jiang R, Zhang X, Li N, Mao Y, Chen H, Deng Z, Wang W, Jiang ZX, Xu L, Yang Z. Effective Synthesis of C20-Epi-Isothiocyanato-Salinomycin and its Thiourea Derivatives as Potential Anticancer Agents. Chemistry 2024; 30:e202402483. [PMID: 39316423 DOI: 10.1002/chem.202402483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/30/2024] [Accepted: 09/24/2024] [Indexed: 09/25/2024]
Abstract
Salinomycin, a naturally occurring polyether ionophore antibiotic isolated from Streptomyces albus, has been demonstrated potent cytotoxic activity against a variety of cancer cell lines. In particular, it exhibits selective targeting of cancer stem cells. However, systemic toxicity, drug resistance and low bioavailability of the drug significantly limit its potential applications. In this study, the C20-epi-isothiocyanate of salinomycin was designed and synthesized, and then reacted with amines as a versatile synthon to assemble a series of salinomycin thiourea derivatives, which improved the druggability of salinomycin. The antiproliferative activities of the compounds were evaluated in vitro against A549, HepG2, HeLa, 4T1, and MCF-7 cancer cell lines using the CCK-8 assay. The pharmacological results showed that some salinomycin thiourea derivatives exhibited excellent inhibitory activity against at least one of the tested tumor cells and high selectivity. Further mechanistic studies showed that compound 9 f, containing a 3,5-difluorobenzyl moiety, could directly induce apoptosis, probably by increasing caspase-9 protein expression and cell cycle arrest in G1 phase in a concentration dependent manner.
Collapse
Affiliation(s)
- Rui Jiang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Xin Zhang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Na Li
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yuyin Mao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Huan Chen
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Zhouming Deng
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wentao Wang
- CAS Key Laboratory of Science and Technology on Applied Catalysis, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, 116023, China
| | - Zhong-Xing Jiang
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, National Center for Magnetic Resonance in Wuhan, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences-Wuhan National Laboratory for Optoelectronics, Wuhan, 430071, China
| | - Liying Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zhigang Yang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
2
|
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
|
3
|
Anees M, Mehrotra N, Tiwari S, Kumar D, Kharbanda S, Singh H. Polylactic acid based biodegradable hybrid block copolymeric nanoparticle mediated co-delivery of salinomycin and doxorubicin for cancer therapy. Int J Pharm 2023; 635:122779. [PMID: 36842520 DOI: 10.1016/j.ijpharm.2023.122779] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 02/05/2023] [Accepted: 02/21/2023] [Indexed: 02/28/2023]
Abstract
Existence of cancer stem cells (CSCs) are primarily responsible for chemoresistance, cancer reoccurrence and treatment failure in cancer patients. Eliminating CSCs along with bulk tumor is a necessity to achieve complete cancer inhibition. Salinomycin (SAL) has potential to specifically target and kill CSCs through blocking their multiple pathways simultaneously. SAL has also been reported to improve anti-cancer efficacy of numerous chemo-based drugs when used in combination therapy. However, clinical use of SAL is restricted due to its high off targeted toxicity. Herein, we have developed a PLA based hybrid block copolymer for concomitant delivery of SAL and doxorubicin (DOX) with an aim to reduce their adverse side effects and enhance the therapeutic efficacy of the treatment. Designed PLA based nanoplatform showed high encapsulation and sustained release profile for both the drugs. Cytotoxicity evaluation on cancer cell lines confirmed the synergistic effect of SAL:DOX co-loaded NPs. Additionally, prepared SAL NPs were also found to be highly effective against chemo-resistant cancer cells and CSCs derived from cancer patient. Most importantly, encapsulation of SAL in PLA NPs improved its pharmacokinetics and biodistribution profile. Consequently, undesired toxicity with SAL NPs was significantly reduced which in-turn increased the dose tolerability in mice as compared to free SAL. Treatment of EAC tumor bearing mice with SAL:DOX co-loaded NPs resulted in excellent tumor regression and complete inhibition of cancer reoccurrence. These results conclude that concomitant delivery of SAL and DOX using PLA based block copolymeric nano-carrier have a strong potential for cancer therapy.
Collapse
Affiliation(s)
- Mohd Anees
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Neha Mehrotra
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Sachchidanand Tiwari
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
| | - Dinesh Kumar
- National Institute of Health and Family Welfare (NIHFW), New Delhi 110067, India
| | | | - Harpal Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India; All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
4
|
Uncovering drug repurposing candidates for head and neck cancers: insights from systematic pharmacogenomics data analysis. Sci Rep 2021; 11:23933. [PMID: 34907286 PMCID: PMC8671460 DOI: 10.1038/s41598-021-03418-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 12/02/2021] [Indexed: 12/24/2022] Open
Abstract
Effective treatment options for head and neck squamous cell carcinoma (HNSCC) are currently lacking. We exploited the drug response and genomic data of the 28 HNSCC cell lines, screened with 4,518 compounds, from the PRISM repurposing dataset to uncover repurposing drug candidates for HNSCC. A total of 886 active compounds, comprising of 418 targeted cancer, 404 non-oncology, and 64 chemotherapy compounds were identified for HNSCC. Top classes of mechanism of action amongst targeted cancer compounds included PI3K/AKT/MTOR, EGFR, and HDAC inhibitors. We have shortlisted 36 compounds with enriched killing activities for repurposing in HNSCC. The integrative analysis confirmed that the average expression of EGFR ligands (AREG, EREG, HBEGF, TGFA, and EPGN) is associated with osimertinib sensitivity. Novel putative biomarkers of response including those involved in immune signalling and cell cycle were found to be associated with sensitivity and resistance to MEK inhibitors respectively. We have also developed an RShiny webpage facilitating interactive visualization to fuel further hypothesis generation for drug repurposing in HNSCC. Our study provides a rich reference database of HNSCC drug sensitivity profiles, affording an opportunity to explore potential biomarkers of response in prioritized drug candidates. Our approach could also reveal insights for drug repurposing in other cancers.
Collapse
|
5
|
Spelling Out CICs: A Multi-Organ Examination of the Contributions of Cancer Initiating Cells' Role in Tumor Progression. Stem Cell Rev Rep 2021; 18:228-240. [PMID: 34244971 DOI: 10.1007/s12015-021-10195-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
Tumor invasion and metastasis remain the leading causes of mortality for patients with cancer despite current treatment strategies. In some cancer types, recurrence is considered inevitable due to the lack of effective anti-metastatic therapies. Recent studies across many cancer types demonstrate a close relationship between cancer-initiating cells (CICs) and metastasis, as well as general cancer progression. First, this review describes CICs' contribution to cancer progression. Then we discuss our recent understanding of mechanisms through which CICs promote tumor invasion and metastasis by examining the role of CICs in each stage. Finally, we examine the current understanding of CICs' contribution to therapeutic resistance and recent developments in CIC-targeting drugs. We believe this understanding is key to advancing anti-CIC clinical therapeutics.
Collapse
|
6
|
Tailoring of P-glycoprotein for effective transportation of actives across blood-brain-barrier. J Control Release 2021; 335:398-407. [PMID: 34087246 DOI: 10.1016/j.jconrel.2021.05.046] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/29/2021] [Accepted: 05/31/2021] [Indexed: 12/25/2022]
Abstract
P-Glycoprotein serves as a gatekeeper in the blood-brain-barrier wherein, it shows a vital part in the elimination of xenobiotics, drugs, foreign molecules etc. and guards the central nervous system from infections and external toxic molecules by functioning as an efflux transporter. It plays an essential role in various brain-related conditions like Parkinsonism, Alzheimer's disease, depression, cancer, etc. and terminates the entry of therapeutic agents across blood-brain-barrier which remains a significant challenge serving as major hindrance in pharmacotherapy of disease. The physiological structure and topology of P-glycoprotein and its relation with blood-brain-barrier and central nervous system gives an idea for targeting nanocarriers across the barrier into brain. This review article provides an overview of current understanding of the nanoformulations-based P-gp trafficking strategies like nanocarriers, stem cell therapy, drugs, substrates, polymeric materials, chemical compounds as well as naturally occurring active constituents for improving drug transport in brain across blood-brain-barrier and contributing in effective nanotherapeutic development for treatment of CNS disorders.
Collapse
|
7
|
Mohan CD, Rangappa S, Nayak SC, Jadimurthy R, Wang L, Sethi G, Garg M, Rangappa KS. Bacteria as a treasure house of secondary metabolites with anticancer potential. Semin Cancer Biol 2021; 86:998-1013. [PMID: 33979675 DOI: 10.1016/j.semcancer.2021.05.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/03/2021] [Accepted: 05/03/2021] [Indexed: 12/27/2022]
Abstract
Cancer stands in the frontline among leading killers worldwide and the annual mortality rate is expected to reach 16.4 million by 2040. Humans suffer from about 200 different types of cancers and many of them have a small number of approved therapeutic agents. Moreover, several types of major cancers are diagnosed at advanced stages as a result of which the existing therapies have limited efficacy against them and contribute to a dismal prognosis. Therefore, it is essential to develop novel potent anticancer agents to counteract cancer-driven lethality. Natural sources such as bacteria, plants, fungi, and marine microorganisms have been serving as an inexhaustible source of anticancer agents. Notably, over 13,000 natural compounds endowed with different pharmacological properties have been isolated from different bacterial sources. In the present article, we have discussed about the importance of natural products, with special emphasis on bacterial metabolites for cancer therapy. Subsequently, we have comprehensively discussed the various sources, mechanisms of action, toxicity issues, and off-target effects of clinically used anticancer drugs (such as actinomycin D, bleomycin, carfilzomib, doxorubicin, ixabepilone, mitomycin C, pentostatin, rapalogs, and romidepsin) that have been derived from different bacteria. Furthermore, we have also discussed some of the major secondary metabolites (antimycins, chartreusin, elsamicins, geldanamycin, monensin, plicamycin, prodigiosin, rebeccamycin, salinomycin, and salinosporamide) that are currently in the clinical trials or which have demonstrated potent anticancer activity in preclinical models. Besides, we have elaborated on the application of metagenomics in drug discovery and briefly described about anticancer agents (bryostatin 1 and ET-743) identified through the metagenomics approach.
Collapse
Affiliation(s)
| | - Shobith Rangappa
- Adichunchanagiri Institute for Molecular Medicine, Adichunchanagiri University, BG Nagara, 571448, Nagamangala Taluk, India
| | - S Chandra Nayak
- Department of Studies in Biotechnology, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Ragi Jadimurthy
- Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore, 570006, India
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Uttar Pradesh, Noida, 201313, India
| | | |
Collapse
|
8
|
Lipid nanocapsules co-encapsulating paclitaxel and salinomycin for eradicating breast cancer and cancer stem cells. Colloids Surf B Biointerfaces 2021; 204:111775. [PMID: 33940518 DOI: 10.1016/j.colsurfb.2021.111775] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/22/2021] [Accepted: 04/15/2021] [Indexed: 12/30/2022]
Abstract
Cancer stem cells (CSCs) comprise a diminutive population of the tumor but pose major obstacles in cancer treatment, often their presence being correlated with poor prognosis, therapeutic resistance and relapse. Nanocarriers of combined drugs regimes demonstrate improved pharmacokinetics and decreased systemic toxicity by targeting the bulk tumor cells along with CSCs, holding the key to future successful chemotherapy. Herein, we developed lipid nanocapsules (LNCs) with co-encapsulated paclitaxel (PTX) and salinomycin (SAL) to eliminate breast cancer cells (MCF-7; non-bCSCs) and cancer stem cells (bCSCs) respectively. LNCs loaded with either PTX or SAL alone or in combination were fabricated by the phase inversion temperature (PIT) method. Physicochemical properties such as nano-size (90 ± 5 nm) and spherical morphology of LNCs were confirmed by dynamic light scattering (DLS) and scanning electron microscopy (SEM) respectively. More than 98 % encapsulation efficiency of drug, alone or in combination, and their controlled drug release was obtained. Drug loaded LNCs were efficiently internalized and exhibited cytotoxicity in non-bCSCs and bCSCs, with dual drug loaded LNCs offering superior cytotoxicity and anti-bCSCs property. Drug loaded nanocapsules induced apoptosis in bCSCs, potentiated with the co-delivery of paclitaxel and salinomycin. Synergistic cytotoxic effect on both cells, non-bCSCs and bCSCs and effective reduction of the tumor mammospheres growth by co-encapsulated paclitaxel and salinomycin suggest LNCs to be promising for treatment of breast cancer.
Collapse
|
9
|
Gao G, Liu F, Xu Z, Wan D, Han Y, Kuang Y, Wang Q, Zhi Q. Evidence of nigericin as a potential therapeutic candidate for cancers: A review. Biomed Pharmacother 2021; 137:111262. [PMID: 33508621 DOI: 10.1016/j.biopha.2021.111262] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 02/07/2023] Open
Abstract
Emerging studies have shown that nigericin, an H+, K+ and Pb2+ ionophore, has exhibited a promising anti-cancer activity in various cancers. However, its anti-cancer mechanisms have not been fully elucidated. In this review, the recent progresses on the use of nigericin in human cancers have been summarized. By exchanging H+ and K+ across cell membranes, nigericin shows promising anti-cancer activities in in vitro and in vivo as a single agent or in combination with other anti-cancer drugs through decreasing intracellular pH (pHi). The underlying mechanisms of nigericin also include the inactivation of Wnt/β-catenin signals, blockade of Androgen Receptor (AR) signaling, and activation of Stress-Activated Protein Kinase/c-Jun N-terminal Kinase (SAPK/JNK) signaling pathways. In many cancers, nigericin is proved to specifically target putative Cancer Stem Cells (CSCs), and its synergistic effects on photodynamic therapy are also reported. Other mechanisms of nigericin including influencing the mitochondrial membrane potentials, inducing an increase in drug accumulation and autophagy, controlling insulin accumulation in nuclei, and increasing the cytotoxic activity of liposome-entrapped drugs, are also discussed. Notably, the potential adverse effects such as teratogenic effects, insulin resistance and eryptosis shall not be ignored. Taken together, these reports suggest that treatment of cancer cells with nigericin may offer a novel therapeutic strategy and future potential of translation to clinics.
Collapse
Affiliation(s)
- Guanzhuang Gao
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Fei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Zhihua Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Daiwei Wan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Ye Han
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yuting Kuang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qiang Wang
- Department of General Surgery, Jiangsu Shengze Hospital, Wujiang, Jiangsu, 215228, China.
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
10
|
Maucort C, Di Giorgio A, Azoulay S, Duca M. Differentiation of Cancer Stem Cells by Using Synthetic Small Molecules: Toward New Therapeutic Strategies against Therapy Resistance. ChemMedChem 2020; 16:14-29. [PMID: 32803855 DOI: 10.1002/cmdc.202000251] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 07/19/2020] [Indexed: 12/12/2022]
Abstract
Despite the existing arsenal of anti-cancer drugs, 10 million people die each year worldwide due to cancers; this highlights the need to discover new therapies based on innovative modes of action against these pathologies. Current chemotherapies are based on the use of cytotoxic agents, targeted drugs, monoclonal antibodies or immunotherapies that are able to reduce or stop the proliferation of cancer cells. However, tumor eradication is often hampered by the presence of resistant cells called cancer stem-like cells or cancer stem cells (CSCs). Several strategies have been proposed to specifically target CSCs such as the use of CSC-specific antibodies, small molecules able to target CSC signaling pathways or drugs able to induce CSC differentiation rendering them sensitive to classical chemotherapy. These latter compounds are the focus of the present review, which aims to report recent advances in anticancer-differentiation strategies. This therapeutic approach was shown to be particularly promising for eradicating tumors in which CSCs are the main reason for therapeutic failure. This general view of the chemistry and mechanism of action of compounds inducing the differentiation of CSCs could be particularly useful for a broad range of researchers working in the field of anticancer therapies as the combination of compounds that induce differentiation with classical chemotherapy could represent a successful approach for future therapeutic applications.
Collapse
Affiliation(s)
- Chloé Maucort
- Université Côte d'Azur, CNRS, Institute of Chemistry of Nice (ICN), 28 avenue Valrose, 06108, Nice, France
| | - Audrey Di Giorgio
- Université Côte d'Azur, CNRS, Institute of Chemistry of Nice (ICN), 28 avenue Valrose, 06108, Nice, France
| | - Stéphane Azoulay
- Université Côte d'Azur, CNRS, Institute of Chemistry of Nice (ICN), 28 avenue Valrose, 06108, Nice, France
| | - Maria Duca
- Université Côte d'Azur, CNRS, Institute of Chemistry of Nice (ICN), 28 avenue Valrose, 06108, Nice, France
| |
Collapse
|
11
|
Dinić J, Efferth T, García-Sosa AT, Grahovac J, Padrón JM, Pajeva I, Rizzolio F, Saponara S, Spengler G, Tsakovska I. Repurposing old drugs to fight multidrug resistant cancers. Drug Resist Updat 2020; 52:100713. [PMID: 32615525 DOI: 10.1016/j.drup.2020.100713] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 06/04/2020] [Accepted: 06/06/2020] [Indexed: 02/08/2023]
Abstract
Overcoming multidrug resistance represents a major challenge for cancer treatment. In the search for new chemotherapeutics to treat malignant diseases, drug repurposing gained a tremendous interest during the past years. Repositioning candidates have often emerged through several stages of clinical drug development, and may even be marketed, thus attracting the attention and interest of pharmaceutical companies as well as regulatory agencies. Typically, drug repositioning has been serendipitous, using undesired side effects of small molecule drugs to exploit new disease indications. As bioinformatics gain increasing popularity as an integral component of drug discovery, more rational approaches are needed. Herein, we show some practical examples of in silico approaches such as pharmacophore modelling, as well as pharmacophore- and docking-based virtual screening for a fast and cost-effective repurposing of small molecule drugs against multidrug resistant cancers. We provide a timely and comprehensive overview of compounds with considerable potential to be repositioned for cancer therapeutics. These drugs are from diverse chemotherapeutic classes. We emphasize the scope and limitations of anthelmintics, antibiotics, antifungals, antivirals, antimalarials, antihypertensives, psychopharmaceuticals and antidiabetics that have shown extensive immunomodulatory, antiproliferative, pro-apoptotic, and antimetastatic potential. These drugs, either used alone or in combination with existing anticancer chemotherapeutics, represent strong candidates to prevent or overcome drug resistance. We particularly focus on outcomes and future perspectives of drug repositioning for the treatment of multidrug resistant tumors and discuss current possibilities and limitations of preclinical and clinical investigations.
Collapse
Affiliation(s)
- Jelena Dinić
- Department of Neurobiology, Institute for Biological Research "Siniša Stanković" - National Institute of Republic of Serbia, University of Belgrade, Bulevar Despota Stefana 142, 11060 Belgrade, Serbia
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany
| | | | - Jelena Grahovac
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, Pasterova 14, 11000 Belgrade, Serbia
| | - José M Padrón
- BioLab, Instituto Universitario de Bio-Orgánica Antonio González (IUBO AG), Universidad de La Laguna, Avda. Astrofísico Francisco Sánchez 2, E-38071 La Laguna, Spain.
| | - Ilza Pajeva
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| | - Flavio Rizzolio
- Department of Molecular Sciences and Nanosystems, Ca' Foscari University of Venice, 301724 Venezia-Mestre, Italy; Pathology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Simona Saponara
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Gabriella Spengler
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, H-6720 Szeged, Dóm tér 10, Hungary
| | - Ivanka Tsakovska
- Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Acad. G. Bonchev Str., Bl. 105, 1113 Sofia, Bulgaria
| |
Collapse
|
12
|
Mechanisms of cancer stem cell therapy. Clin Chim Acta 2020; 510:581-592. [PMID: 32791136 DOI: 10.1016/j.cca.2020.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/01/2020] [Accepted: 08/07/2020] [Indexed: 12/12/2022]
Abstract
Cancer stem cells (CSCs) are responsible for carcinogenesis and tumorigenesis and are involved in drug and radiation resistance, metastasis, tumor relapse and initiation. Remarkably, they have other abilities such as inheritance of self-renewal and de-differentiation. Hence, targeting CSCs is considered a potential anti-cancer therapeutic strategy. Recent advances in the identification of biomarkers to recognize CSCs and the development of new techniques to evaluate tumorigenic and carcinogenic roles of CSCs are instrumental to this approach. Elucidation of signaling pathways that regulate CSCs colony progression and drug resistance are critical in establishing effective targeted therapies. CSCs play a central key role in immunomodulation, immune evasion and effector immunity, which alters immune system balancing. These include mTOR, SHH, NOTCH and Wnt/β-catering in cancer progression. In this review article, we discuss the importance of these CSCs pathways in cancer therapy.
Collapse
|
13
|
Nobili S, Lapucci A, Landini I, Coronnello M, Roviello G, Mini E. Role of ATP-binding cassette transporters in cancer initiation and progression. Semin Cancer Biol 2020; 60:72-95. [PMID: 31412294 DOI: 10.1016/j.semcancer.2019.08.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 12/18/2022]
Abstract
ATP Binding Cassette (ABC) transporters, widely studied in cancer for their role in drug resistance, have been more recently also considered for their contribution to cancer cell biology. To date, many data provide evidences for their potential role in all the phases of cancer development from cancer susceptibility, tumor initiation, tumor progression and metastasis. Although many evidences are based on correlative analyses, data describing a direct or indirect role of ABC transporters in cancer biology are increasing. Overall, current available information suggests a relevant molecular effector role of some ABC transporters in cancer invasion and metastasis as reported in experimental tumor models. From a therapeutic point of view, due to the physiological relevant roles that ABC transporters play in the organism, the capability to selectively inhibit the function or the expression of ABC transporters in cancer stem cells or other tumor cells, represents the main challenge for researcher scientists. A detailed and updated description of the current knowledge on the role of ABC transporters in cancer biology is provided.
Collapse
Affiliation(s)
- Stefania Nobili
- Department of Health Sciences, University of Florence, Florence, Italy.
| | - Andrea Lapucci
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Ida Landini
- Department of Health Sciences, University of Florence, Florence, Italy
| | | | | | - Enrico Mini
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
14
|
Overcoming Resistance to Platinum-Based Drugs in Ovarian Cancer by Salinomycin and Its Derivatives-An In Vitro Study. Molecules 2020; 25:molecules25030537. [PMID: 31991882 PMCID: PMC7037477 DOI: 10.3390/molecules25030537] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 01/23/2020] [Accepted: 01/24/2020] [Indexed: 12/16/2022] Open
Abstract
Polyether ionophore salinomycin (SAL) and its semi-synthetic derivatives are recognized as very promising anticancer drug candidates due to their activity against various types of cancer cells, including multidrug-resistant populations. Ovarian cancer is the deadliest among gynecologic malignancies, which is connected with the development of chemoresistant forms of the disease in over 70% of patients after initial treatment regimen. Thus, we decided to examine the anticancer properties of SAL and selected SAL derivatives against a series of drug-sensitive (A2780, SK-OV-3) and derived drug-resistant (A2780 CDDP, SK-OV-3 CDDP) ovarian cancer cell lines. Although SAL analogs showed less promising IC50 values than SAL, they were identified as the antitumor agents that significantly overcome the resistance to platinum-based drugs in ovarian cancer, more potent than unmodified SAL and commonly used anticancer drugs—5-fluorouracil, gemcitabine, and cisplatin. Moreover, when compared with SAL used alone, our experiments proved for the first time increased selectivity of SAL-based dual therapy with 5-fluorouracil or gemcitabine, especially towards A2780 cell line. Looking closer at the results, SAL acted synergistically with 5-fluorouracil towards the drug-resistant A2780 cell line. Our results suggest that combinations of SAL with other antineoplastics may become a new therapeutic option for patients with ovarian cancer.
Collapse
|
15
|
Skeberdytė A, Sarapinienė I, Krasko JA, Barakauskienė A, Žilionytė K, Prokarenkaitė R, Sužiedėlis K, Bukelskienė V, Jarmalaitė S. Salinomycin and dichloroacetate synergistically inhibit Lewis lung carcinoma cell proliferation, tumor growth and metastasis. Biochem Biophys Res Commun 2020; 523:874-879. [PMID: 31955885 DOI: 10.1016/j.bbrc.2019.12.107] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 12/21/2019] [Indexed: 12/27/2022]
Abstract
Drug combination is considered to be the cornerstone of cancer treatment. Simultaneous administration of two or more drugs but at lower doses not only increases cytotoxic effects on tumor cells, but also reduces side effects and possibly overcomes drug resistance. Salinomycin is a well-known cancer stem cell killer, and dichloroacetate is a pyruvate dehydrogenase kinase inhibitor that exclusively targets cells with altered mitochondrial activity, a characteristic being common to most of the cancer cells. In our recent study, we have demonstrated that salinomycin exerted a cytotoxic effect on colorectal carcinoma cells in the 2D and 3D cultures and provided evidence that the mechanism of their synergy was mediated by dichloroacetate-dependent inhibition of the activity of multidrug resistance proteins. In the current work, we confirmed the synergistic cytotoxic properties of salinomycin and dichloroacetate in the 2D and 3D cultures of Lewis lung carcinoma (LLC1) cells. To verify if a synergistic effect of these compounds persisted in vivo, we performed series of experiments using a syngeneic LLC1-C57BL/6 mouse model and demonstrated that combination therapy with salinomycin and DCA increased the survival rate of allografted mice, inhibited metastatic site formation and reduced the populations of cancer stem cells as well as cells that underwent the epithelial-to-mesenchymal transition. Our results demonstrate that a synergistic effect of salinomycin and dichloroacetate exists not only in vitro but also in vivo and suggest their benefits in the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Aistė Skeberdytė
- Institute of Biosciences, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10222, Vilnius, Lithuania.
| | - Ieva Sarapinienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių pr. 15, LT-50162, Kaunas, Lithuania
| | | | | | - Karolina Žilionytė
- National Cancer Institute, Santariškių g. 1, LT-08660, Vilnius, Lithuania
| | | | | | - Virginija Bukelskienė
- Institute of Biochemistry, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10223, Vilnius, Lithuania
| | - Sonata Jarmalaitė
- National Cancer Institute, Santariškių g. 1, LT-08660, Vilnius, Lithuania
| |
Collapse
|
16
|
Antoszczak M, Huczyński A. Salinomycin and its derivatives - A new class of multiple-targeted "magic bullets". Eur J Med Chem 2019; 176:208-227. [PMID: 31103901 DOI: 10.1016/j.ejmech.2019.05.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022]
Abstract
The history of drug development clearly shows the scale of painstaking effort leading to a finished product - a highly biologically active agent that would be at the same time no or little toxic to human organism. Moreover, the aim of modern drug discovery can move from "one-molecule one-target" concept to more promising "one-molecule multiple-targets" one, particularly in the context of effective fight against cancer and other complex diseases. Gratifyingly, natural compounds are excellent source of potential drug leads. One of such promising naturally-occurring drug candidates is a polyether ionophore - salinomycin (SAL). This compound should be identified as multi-target agent for two reasons. Firstly, SAL combines a broad spectrum of bioactivity, including antibacterial, antifungal, antiviral, antiparasitic and anticancer activity, with high selectivity of action, proving its significant therapeutic potential. Secondly, the multimodal mechanism of action of SAL has been shown to be related to its interactions with multiple molecular targets and signalling pathways that are synergistic for achieving a therapeutic anticancer effect. On the other hand, according to the Paul Ehrlich's "magic bullet" concept, invariably inspiring the scientists working on design of novel target-selective molecules, a very interesting direction of research is rational chemical modification of SAL. Importantly, many of SAL derivatives have been found to be more promising as chemotherapeutics than the native structure. This concise review article is focused both on the possible role of SAL and its selected analogues in future antimicrobial and/or cancer therapy, and on the potential use of SAL as a new class of multiple-targeted "magic bullet" because of its multimodal mechanism of action.
Collapse
Affiliation(s)
- Michał Antoszczak
- Department of Bioorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89b, 61‒614, Poznań, Poland
| | - Adam Huczyński
- Department of Bioorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89b, 61‒614, Poznań, Poland.
| |
Collapse
|
17
|
Antoszczak M. A medicinal chemistry perspective on salinomycin as a potent anticancer and anti-CSCs agent. Eur J Med Chem 2019; 164:366-377. [DOI: 10.1016/j.ejmech.2018.12.057] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 01/30/2023]
|
18
|
Borlle L, Dergham A, Wund Z, Zumbo B, Southard T, Hume KR. Salinomycin decreases feline sarcoma and carcinoma cell viability when combined with doxorubicin. BMC Vet Res 2019; 15:36. [PMID: 30678671 PMCID: PMC6346515 DOI: 10.1186/s12917-019-1780-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 01/14/2019] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Cancer is a significant health threat in cats. Chemoresistance is prevalent in solid tumors. The ionophore salinomycin has anti-cancer properties and may work synergistically with chemotherapeutics. The purpose of our study was to determine if salinomycin could decrease cancer cell viability when combined with doxorubicin in feline sarcoma and carcinoma cells. RESULTS We established two new feline injection-site sarcoma cell lines, B4 and C10, and confirmed their tumorigenic potential in athymic nude mice. B4 was more resistant to doxorubicin than C10. Dose-dependent effects were not observed until 92 μM in B4 cells (p = 0.0006) vs. 9.2 μM (p = 0.0004) in C10 cells. Dose-dependent effects of salinomycin were observed at 15 μM in B4 cells (p = 0.025) and at 10 μM in C10 cells (p = 0.020). Doxorubicin plus 5 μM salinomycin decreased viability of B4 cells compared to either agent alone, but only at supra-pharmacological doxorubicin concentrations. However, doxorubicin plus 5 μM salinomycin decreased viability of C10 cells compared to either agent alone at doxorubicin concentrations that can be achieved in vivo (1.84 and 4.6 μM, p < 0.004). In SCCF1 cells, dose-dependent effects of doxorubicin and salinomycin were observed at 9.2 (p = 0.036) and 2.5 (p = 0.0049) μM, respectively. When doxorubicin was combined with either 1, 2.5, or 5 μM of salinomycin in SCCF1 cells, dose-dependent effects of doxorubicin were observed at 9.2 (p = 0.0021), 4.6 (p = 0.0042), and 1.84 (p = 0.0021) μM, respectively. Combination index calculations for doxorubicin plus 2.5 and 5 μM salinomycin in SCCF1 cells were 0.4 and 0.6, respectively. CONCLUSIONS We have developed two new feline sarcoma cell lines that can be used to study chemoresistance. We observed that salinomycin may potentiate (C10 cells) or work synergistically (SCCF1 cells) with doxorubicin in certain feline cancer cells. Further research is indicated to understand the mechanism of action of salinomycin in feline cancer cells as well as potential tolerability and toxicity in normal feline tissues.
Collapse
Affiliation(s)
- Lucia Borlle
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
- Department of Animal Sciences, Cornell University College of Agricultural and Life Sciences, Ithaca, NY 14853 USA
| | - Abdo Dergham
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Zacharie Wund
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Brittany Zumbo
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Teresa Southard
- Department of Biomedical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| | - Kelly R. Hume
- Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY 14853 USA
| |
Collapse
|
19
|
Dichloroacetate and Salinomycin Exert a Synergistic Cytotoxic Effect in Colorectal Cancer Cell Lines. Sci Rep 2018; 8:17744. [PMID: 30531808 PMCID: PMC6288092 DOI: 10.1038/s41598-018-35815-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 11/07/2018] [Indexed: 12/18/2022] Open
Abstract
In the present study, we examined a hypothesis that dichloroacetate, a metabolic inhibitor, might efficiently potentiate the cytotoxic effect of salinomycin, an antibiotic ionophore, on two human colorectal cancer derived cell lines DLD-1 and HCT116. First, we performed a series of dose response experiments in the 2D cell culture by applying mono- and combination therapy and by using the Chou-Talalay method found that salinomycin in combination with dichloroacetate acted synergistically in both cell lines. Secondly, in order to recapitulate the in vivo tumor architecture, we tested various doses of these compounds, alone and in combination, in the 3D multicellular spheroid culture. The effect of combination of dichloracetate and salinomycin on multicellular spheroid size was stronger than the sum of both monotherapies, particularly in HCT116 cells. Further, we demonstrate that the synergistic effect of compounds may be related to the inhibitory effect of dichloroacetate on multidrug resistance proteins, and in contrast, it is not related to dichloroacetate-induced reduction of intracellular pH. Our findings indicate that the combination therapy of salinomycin and dichloroacetate could be an effective option for colorectal cancer treatment and provide the first mechanistic explanation of the synergistic action of these compounds.
Collapse
|
20
|
Kaushik V, Yakisich JS, Kumar A, Azad N, Iyer AKV. Ionophores: Potential Use as Anticancer Drugs and Chemosensitizers. Cancers (Basel) 2018; 10:E360. [PMID: 30262730 PMCID: PMC6211070 DOI: 10.3390/cancers10100360] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/13/2018] [Accepted: 09/21/2018] [Indexed: 01/08/2023] Open
Abstract
Ion homeostasis is extremely important for the survival of both normal as well as neoplastic cells. The altered ion homeostasis found in cancer cells prompted the investigation of several ionophores as potential anticancer agents. Few ionophores, such as Salinomycin, Nigericin and Obatoclax, have demonstrated potent anticancer activities against cancer stem-like cells that are considered highly resistant to chemotherapy and responsible for tumor relapse. The preclinical success of these compounds in in vitro and in vivo models have not been translated into clinical trials. At present, phase I/II clinical trials demonstrated limited benefit of Obatoclax alone or in combination with other anticancer drugs. However, future development in targeted drug delivery may be useful to improve the efficacy of these compounds. Alternatively, these compounds may be used as leading molecules for the development of less toxic derivatives.
Collapse
Affiliation(s)
- Vivek Kaushik
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Juan Sebastian Yakisich
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Anil Kumar
- Great Plains Health, North Platte, NE 69101, USA.
| | - Neelam Azad
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Anand K V Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| |
Collapse
|
21
|
Versini A, Saier L, Sindikubwabo F, Müller S, Cañeque T, Rodriguez R. Chemical biology of salinomycin. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Forouzesh F, Agharezaee N. Review on the molecular signaling pathways involved in controlling cancer stem cells and treatment. THE JOURNAL OF QAZVIN UNIVERSITY OF MEDICAL SCIENCES 2018. [DOI: 10.29252/qums.22.3.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
|
23
|
Moreira H, Szyjka A, Gąsiorowski K. Chemopreventive activity of celastrol in drug-resistant human colon carcinoma cell cultures. Oncotarget 2018; 9:21211-21223. [PMID: 29765532 PMCID: PMC5940375 DOI: 10.18632/oncotarget.25014] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 03/09/2018] [Indexed: 01/06/2023] Open
Abstract
Celastrol (tripterine) a pentacyclic triterpenoid extracted from the roots of Tripterygium wilfordii Hook f., exhibits potent antioxidant and anti-inflammatory activity and also exerts important anti-cancer effects, as induction of apoptosis and lowering the level of drug resistance of several cancers. Increased level of cellular resistance to cytostatic drugs is typical for colorectal cancers, and largely determines the failure of chemotherapy for this tumor. The purpose of our research was to evaluate the chemopreventive effect of celastrol on cultures of colon cancer cells resistant to doxorubicin (LOVO/DX). With the use of flow cytometry we have shown that celastrol reduces the cell size of the SP (side population; subpopulation of cancer cells enriched with cancer stem cells), increases frequency of apoptosis and binds to Pgp protein in cell membranes inhibiting its transport function. The inhibition of the Pgp transport function has been shown to increase the accumulation of rhodamine-123 and standard cytostatic- doxorubicin in LOVO/DX cells. Our results prove that celastrol exhibits significant chemopreventive and chemosensitizing activities on drug resistant colon cancer cells. Celastrol appears to be a good candidate for adjuvant medicine that can improve the effectiveness of standard cytostatic therapy in humans.
Collapse
Affiliation(s)
- Helena Moreira
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Anna Szyjka
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| | - Kazimierz Gąsiorowski
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wrocław, Poland
| |
Collapse
|
24
|
Zhao CY, Cheng R, Yang Z, Tian ZM. Nanotechnology for Cancer Therapy Based on Chemotherapy. Molecules 2018; 23:E826. [PMID: 29617302 PMCID: PMC6017446 DOI: 10.3390/molecules23040826] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 03/27/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023] Open
Abstract
Chemotherapy has been widely applied in clinics. However, the therapeutic potential of chemotherapy against cancer is seriously dissatisfactory due to the nonspecific drug distribution, multidrug resistance (MDR) and the heterogeneity of cancer. Therefore, combinational therapy based on chemotherapy mediated by nanotechnology, has been the trend in clinical research at present, which can result in a remarkably increased therapeutic efficiency with few side effects to normal tissues. Moreover, to achieve the accurate pre-diagnosis and real-time monitoring for tumor, the research of nano-theranostics, which integrates diagnosis with treatment process, is a promising field in cancer treatment. In this review, the recent studies on combinational therapy based on chemotherapy will be systematically discussed. Furthermore, as a current trend in cancer treatment, advance in theranostic nanoparticles based on chemotherapy will be exemplified briefly. Finally, the present challenges and improvement tips will be presented in combination therapy and nano-theranostics.
Collapse
Affiliation(s)
| | | | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Zhong-Min Tian
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, China.
| |
Collapse
|
25
|
Liu F, Li W, Hua S, Han Y, Xu Z, Wan D, Wang Y, Chen W, Kuang Y, Shi J, Zhi Q. Nigericin Exerts Anticancer Effects on Human Colorectal Cancer Cells by Inhibiting Wnt/β-catenin Signaling Pathway. Mol Cancer Ther 2018; 17:952-965. [PMID: 29483216 DOI: 10.1158/1535-7163.mct-17-0906] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 11/20/2017] [Accepted: 02/07/2018] [Indexed: 11/16/2022]
Abstract
Nigericin, an antibiotic derived from Streptomyces hygroscopicus, which works by acting as an H+, K+, and Pb2+ ionophore, has exhibited promising anticancer activity. The main purpose of this study is to investigate its inhibitory effects on Wnt/β-catenin signaling pathway in colorectal cancer cells and clarify the underlying mechanism. We exposed two colorectal cancer lines (SW620 and KM12) to increasing concentrations of nigericin for different time periods and the 50% inhibiting concentration (IC50) values were evaluated. Our data showed that nigericin treatment significantly reduced tumor cell proliferation in dose- and time-dependent manners in colorectal cancer cells. The subsequent experiments in vitro and in vivo implied that nigericin could significantly suppress the tumor growth, migration, and invasion, and induce the apoptosis of colorectal cancer cells. Our results of Western blot and immunofluorescence assay showed that nigericin could suppress the Wnt/β-catenin signaling pathway in colorectal cancer cells with dose-dependent increased expressions of downstream effectors and target proteins. To further elucidate the inhibitory effects of nigericin via a β-catenin-dependent signaling mechanism, we established the stably β-catenin overexpression colorectal cancer cells. Western blot, SuperTOPFlash luciferase reporter, and immunoprecipitation assays all confirmed β-catenin as a critical intermediary and player in Wnt/β-catenin pathway, and nigericin exerted anticancer effects on colorectal cancer cells by directly targeting the β-catenin destruction complex. These results suggested that Wnt/β-catenin signaling might have an essential role in colorectal cancer progression. Nigericin targeting Wnt/β-catenin signaling might provide new insight into the molecular mechanism of nigericin toward cancer cells, and suggest possible clinical application in colorectal cancer. Mol Cancer Ther; 17(5); 952-65. ©2018 AACR.
Collapse
Affiliation(s)
- Fei Liu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wei Li
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shangbo Hua
- Department of General Surgery, Kunshan Hospital of Traditional Chinese Medicine, Kunshan, China
| | - Ye Han
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhihua Xu
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Daiwei Wan
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yilin Wang
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuting Kuang
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jianming Shi
- Department of Oncology, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, China.
| | - Qiaoming Zhi
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
26
|
Identification of DNA-PKcs as a primary resistance factor of salinomycin in osteosarcoma cells. Oncotarget 2018; 7:79417-79427. [PMID: 27765904 PMCID: PMC5346724 DOI: 10.18632/oncotarget.12712] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 01/01/2023] Open
Abstract
Malignant osteosarcoma (OS) is still a deadly disease for many affected patients. The search for the novel anti-OS agent is extremely urgent and important. Our previous study has proposed that salinomycin is a novel anti-OS agent. Here we characterized DNA-dependent protein kinase catalytic subunit (DNA-PKcs) as a primary salinomycin resistance factor in OS cells. DNA-PKcs inhibitors (NU7026, NU7441 and LY294002) or DNA-PKcs shRNA knockdown dramatically potentiated salinomycin-induced death and apoptosis of OS cells (U2OS and MG-63 lines). Further, forced-expression of microRNA-101 (“miR-101”) downregulated DNA-PKcs and augmented salinomycin's cytotoxicity against OS cells. Reversely, over-expression of DNA-PKcs in OS cells inhibited salinomycin's lethality. For the mechanism study, we show that DNA-PKcs is required for salinomycin-induced pro-survival autophagy activation. DNA-PKcs inhibition (by NU7441), shRNA knockdown or miR-101 expression inhibited salinomycin-induced Beclin-1 expression and autophagy induction. Meanwhile, knockdown of Beclin-1 by shRNA significantly sensitized salinomycin-induced OS cell lethality. In vivo, salinomycin administration suppressed U2OS xenograft tumor growth in severe combined immuno-deficient (SCID) mice, and its anti-tumor activity was dramatically potentiated with co-administration of the DNA-PKcs inhibitor NU7026. Together, these results suggest that DNA-PKcs could be a primary resistance factor of salinomycin in OS cells. DNA-PKcs inhibition or silence may thus significantly increase salinomycin's sensitivity in OS cells.
Collapse
|
27
|
Jiang J, Li H, Qaed E, Zhang J, Song Y, Wu R, Bu X, Wang Q, Tang Z. Salinomycin, as an autophagy modulator-- a new avenue to anticancer: a review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:26. [PMID: 29433536 PMCID: PMC5809980 DOI: 10.1186/s13046-018-0680-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
Since Salinomycin (Sal) emerged its ability to target breast cancer stem cells in 2009, numerous experiments have been carried out to test Sal’s anticancer effects. What deserve to be mentioned is that Sal can efficiently induce proliferation inhibition, cell death and metastasis suppression against human cancers from different origins both in vivo and in vitro without causing serious side effects as the conventional chemotherapeutical drugs on the body. There may be novel cell death pathways involving the anticancer effects of Sal except the conventional pathways, such as autophagic pathway. This review is focused on how autophagy involves the effects of Sal, trying to describe clearly and systematically why autophagy plays a vital role in predominant anticancer effects of Sal, including its distinctive characteristic. Based on recent advances, we present evidence that a dual role of Sal involving in autophagy may account for its unique anticancer effects - the preference for cancer cells. Further researches are required to confirm the authenticity of this suppose in order to develop an ideal anticancer drug.
Collapse
Affiliation(s)
- Jiang Jiang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Hailong Li
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Eskandar Qaed
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Jing Zhang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Yushu Song
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Rong Wu
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Xinmiao Bu
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Qinyan Wang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| |
Collapse
|
28
|
Wang T, Narayanaswamy R, Ren H, Torchilin VP. Combination therapy targeting both cancer stem-like cells and bulk tumor cells for improved efficacy of breast cancer treatment. Cancer Biol Ther 2017; 17:698-707. [PMID: 27259361 DOI: 10.1080/15384047.2016.1190488] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Many types of tumors are organized in a hierarchy of heterogeneous cell populations. The cancer stem-like cells (CSCs) hypothesis suggests that tumor development and metastasis are driven by a minority population of cells, which are responsible for tumor initiation, growth and recurrences. The inability to efficiently eliminate CSCs during chemotherapy, together with CSCs being highly tumorigenic and invasive, may result in treatment failure due to cancer relapse and metastases. CSCs are emerging as a promising target for the development of translational cancer therapies. Ideal panacea for cancer would kill all malignant cells, including CSCs and bulk tumor cells. Since both chemotherapy and CSCs-specific therapy are insufficient to cure cancer, we propose combination therapy with CSCs-targeted agents and chemotherapeutics for improved breast cancer treatment. We generated in vitro mammosphere of 2 breast cancer cell lines, and demonstrated ability of mammospheres to grow and enrich cancer cells with stem-like properties, including self-renewal, multilineage differentiation and enrichment of cells expressing breast cancer stem-like cell biomarkers CD44(+)/CD24(-/low). The formation of mammospheres was significantly inhibited by salinomycin, validating its pharmacological role against the cancer stem-like cells. In contrast, paclitaxel showed a minimal effect on the proliferation and growth of breast cancer stem-like cells. While combination therapies of salinomycin with conventional chemotherapy (paclitaxel or lipodox) showed a potential to improve tumor cell killing, different subtypes of breast cancer cells showed different patterns in response to the combination therapies. While optimization of combination therapy is warranted, the design of combination therapy should consider phenotypic attributes of breast cancer types.
Collapse
Affiliation(s)
- Tao Wang
- a Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston , MA , USA
| | - Radhika Narayanaswamy
- a Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston , MA , USA
| | - Huilan Ren
- a Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston , MA , USA
| | - Vladimir P Torchilin
- a Center for Pharmaceutical Biotechnology and Nanomedicine, Northeastern University , Boston , MA , USA.,b Department of Biochemistry , Faculty of Science, King Abdulaziz University , Jeddah , Saudi Arabia
| |
Collapse
|
29
|
Autophagy inhibition enhances antiproliferative effect of salinomycin in pancreatic cancer cells. Pancreatology 2017; 17:990-996. [PMID: 28927939 DOI: 10.1016/j.pan.2017.08.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 08/10/2017] [Accepted: 08/24/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Salinomycin has cytotoxic effects on various types of malignancy and induces autophagy. However, it has not been clarified whether autophagy induced by salinomycin treatment has a protective or cytotoxic role. We investigated whether salinomycin affects autophagy in pancreatic cancer cells and whether autophagy induced by salinomycin treatment has a protective or cytotoxic role in these cells. METHODS We investigated the effect of salinomycin using three pancreatic cancer cell lines. We investigated effect on proliferation and the CD133 positive fraction using flow cytometry. In addition, we monitored the change in autophagic activity after salinomycin treatment using fluorescent immunostaining, western blotting, and flow cytometry. Finally, knockdown of ATG5 or ATG7 by siRNA was used to investigate the impact of autophagy inhibition on sensitivity to salinomycin. RESULTS Salinomycin suppressed the proliferation of pancreatic cancer cells in a concentration dependent manner, and reduced the CD133 positive fraction. Salinomycin enhanced autophagy activity in these cells in a concentration dependent manner. Autophagy inhibition made pancreatic cancer cells more sensitive to salinomycin. CONCLUSIONS Our data provide the first evidence indicating that autophagy induced by salinomycin have a protective role in pancreatic cancer cells. A new therapeutic strategy of combining salinomycin, autophagy inhibitors, and anticancer drugs could hold promise for pancreatic cancer treatment.
Collapse
|
30
|
Manmuan S, Sakunrangsit N, Ketchart W. Salinomycin overcomes acquired tamoxifen resistance through AIB1 and inhibits cancer cell invasion in endocrine resistant breast cancer. Clin Exp Pharmacol Physiol 2017; 44:1042-1052. [PMID: 28656701 DOI: 10.1111/1440-1681.12806] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 06/21/2017] [Accepted: 06/22/2017] [Indexed: 01/09/2023]
Abstract
Salinomycin is a monocarboxylic polyether ionophore isolated from Streptomyces albus. It has been widely used as an antibiotic in veterinary medicine in poultry. A recent study demonstrated that salinomycin selectively inhibits human breast cancer stem cells; one possible mechanism of tamoxifen resistance. Our results show that salinomycin is effective in inhibiting MCF-7/LCC2 and MCF-7/LCC9 cell lines which are well-established endocrine resistant cells and has a synergistic effect in combination with tamoxifen using MTT proliferation assay. The inhibitory effect of salinomycin on the reduction of critical ER co-activator; amplified breast 1 (AIB1) mRNA and protein expression is overcoming tamoxifen resistance. Moreover, salinomycin significantly inhibits cell invasion in Matrigel invasion assay. The effect was mediated at least in part by the decrease of matrix metalopeptidase 9 (MMP-9) which is one critical enzyme facilitated in the cell invasion process. In conclusion, salinomycin should be developed as a novel agent used alone or in combination for endocrine-resistant breast cancer.
Collapse
Affiliation(s)
- Suwisit Manmuan
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nithidol Sakunrangsit
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Wannarasmi Ketchart
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
31
|
Agliano A, Calvo A, Box C. The challenge of targeting cancer stem cells to halt metastasis. Semin Cancer Biol 2017; 44:25-42. [PMID: 28323021 DOI: 10.1016/j.semcancer.2017.03.003] [Citation(s) in RCA: 155] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Revised: 03/10/2017] [Accepted: 03/13/2017] [Indexed: 12/21/2022]
|
32
|
Singh AK, Sharma N, Ghosh M, Park YH, Jeong DK. Emerging importance of dietary phytochemicals in fight against cancer: Role in targeting cancer stem cells. Crit Rev Food Sci Nutr 2017; 57:3449-3463. [DOI: 10.1080/10408398.2015.1129310] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Amit Kumar Singh
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | - Neelesh Sharma
- Division of Veterinary Medicine, Faculty of Veterinary Science and Animal Husbandry, Sher-e-Kashmir University of Agricultural Sciences and Technology, R. S. Pura, Jammu, India
| | - Mrinmoy Ghosh
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| | | | - Dong Kee Jeong
- Laboratory of Animal Genetic Engineering and Stem Cell Biology, Department of Animal Biotechnology, Faculty of Biotechnology, Jeju National University, Jeju, Republic of Korea
| |
Collapse
|
33
|
Dewangan J, Srivastava S, Rath SK. Salinomycin: A new paradigm in cancer therapy. Tumour Biol 2017; 39:1010428317695035. [PMID: 28349817 DOI: 10.1177/1010428317695035] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The primary hurdle in the treatment of cancer is acquisition of resistance by the tumor cells toward multiple drugs and selectively targeting the cancer stem cells. This problem was overcome by the chemotherapeutic property of recently discovered drug salinomycin. Exact mechanism of action of salinomycin is not yet known, but there are multiple pathways by which salinomycin inhibits tumor growth. Salinomycin decreases the expression of adenosine triphosphate-binding cassette transporter in multidrug resistance cells and interferes with Akt signaling pathway, Wnt/β-catenin, Hedgehog, and Notch pathways of cancer progression. Salinomycin selectively targets cancer stem cells. The potential of salinomycin to eliminate both cancer stem cells and therapy-resistant cancer cells may characterize the compound as a novel and an efficient chemotherapeutic drug.
Collapse
Affiliation(s)
- Jayant Dewangan
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sonal Srivastava
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Srikanta Kumar Rath
- Genotoxicity Laboratory, Division of Toxicology, CSIR-Central Drug Research Institute, Lucknow, India
| |
Collapse
|
34
|
Yakisich JS, Azad N, Kaushik V, O’Doherty GA, Iyer AKV. Nigericin decreases the viability of multidrug-resistant cancer cells and lung tumorspheres and potentiates the effects of cardiac glycosides. Tumour Biol 2017; 39:1010428317694310. [DOI: 10.1177/1010428317694310] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Multiple factors including tumor heterogeneity and intrinsic or acquired resistance have been associated with drug resistance in lung cancer. Increased stemness and the plasticity of cancer cells have been identified as important mechanisms of resistance; therefore, treatments targeting cancer cells independent of stemness phenotype would be much more effective in treating lung cancer. In this article, we have characterized the anticancer effects of the antibiotic Nigericin in cells displaying varying degrees of stemness and resistance to anticancer drugs, arising from (1) routine culture conditions, (2) prolonged periods of serum starvation. These cells are highly resistant to conventional anticancer drugs such as Paclitaxel, Hydroxyurea, Colchicine, Obatoclax, Wortmannin, and LY294002, and the multidrug-resistant phenotype of cells growing under prolonged periods of serum starvation is likely the result of extensive rewiring of signaling pathways, and (3) lung tumorspheres that are enriched for cancer stem-like cells. We found that Nigericin potently inhibited the viability of cells growing under routine culture conditions, prolonged periods of serum starvation, and lung tumorspheres. In addition, we found that Nigericin downregulated the expression of key proteins in the Wnt canonical signaling pathway such as LRP6, Wnt5a/b, and β-catenin, but promotes β-catenin translocation into the nucleus. The antitumor effects of Nigericin were potentiated by the Wnt activator HLY78 and by therapeutic levels of the US Food and Drug Administration–approved drug Digitoxin and its novel synthetic analog MonoD. We believe that Nigericin may be used in a co-therapy model in combination with other novel chemotherapeutic agents in order to achieve potent inhibition of cancers that display varying degrees of stemness, potentially leading to sustained anticancer effects.
Collapse
Affiliation(s)
- Juan Sebastian Yakisich
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA, USA
| | - Neelam Azad
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA, USA
| | - Vivek Kaushik
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA, USA
| | | | - Anand Krishnan V Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA, USA
| |
Collapse
|
35
|
Li Y, Atkinson K, Zhang T. Combination of chemotherapy and cancer stem cell targeting agents: Preclinical and clinical studies. Cancer Lett 2017; 396:103-109. [PMID: 28300634 DOI: 10.1016/j.canlet.2017.03.008] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 12/12/2022]
Abstract
The cancer stem cell model claims that the initiation, maintenance, and growth of a tumor are driven by a small population of cancer cells termed cancer stem cells. Cancer stem cells possess a variety of phenotypes associated with therapeutic resistance and often cause recurrence of the diseases. Several strategies have been investigated to target cancer stem cells in a variety of cancers, such as blocking one or more self-renewal signaling pathways, reducing the expression of drug efflux and ATP-binding cassette efflux transporters, modulating epigenetic aberrations, and promoting cancer stem cell differentiation. A number of cell and animal studies strongly support the potential benefits of combining chemotherapeutic drugs with cancer stem cell targeting agents. Clinical trials are still underway to address the pharmacokinetics, safety, and efficacy of combination treatment. This mini-review provides an updated discussion of these preclinical and clinical studies.
Collapse
Affiliation(s)
- Yanyan Li
- College of Science and Humanities, Husson University, 1 College Circle, Bangor, ME, 04401, USA.
| | - Katharine Atkinson
- College of Science and Humanities, Husson University, 1 College Circle, Bangor, ME, 04401, USA
| | - Tao Zhang
- School of Pharmacy, Husson University, 1 College Circle, Bangor, ME, 04401, USA
| |
Collapse
|
36
|
David JM, Dominguez C, Palena C. Pharmacological and immunological targeting of tumor mesenchymalization. Pharmacol Ther 2016; 170:212-225. [PMID: 27916651 DOI: 10.1016/j.pharmthera.2016.11.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Controlling the spread of carcinoma cells to distant organs is the foremost challenge in cancer treatment, as metastatic disease is generally resistant to therapy and is ultimately incurable for the majority of patients. The plasticity of tumor cell phenotype, in which the behaviors and functions of individual tumor cells differ markedly depending upon intrinsic and extrinsic factors, is now known to be a central mechanism in cancer progression. Our expanding knowledge of epithelial and mesenchymal phenotypic states in tumor cells, and the dynamic nature of the transitions between these phenotypes has created new opportunities to intervene to better control the behavior of tumor cells. There are now a variety of innovative pharmacological approaches to preferentially target tumor cells that have acquired mesenchymal features, including cytotoxic agents that directly kill these cells, and inhibitors that block or revert the process of mesenchymalization. Furthermore, novel immunological strategies have been developed to engage the immune system in seeking out and destroying mesenchymalized tumor cells. This review highlights the relevance of phenotypic plasticity in tumor biology, and discusses recently developed pharmacological and immunological means of targeting this phenomenon.
Collapse
Affiliation(s)
- Justin M David
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Charli Dominguez
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States
| | - Claudia Palena
- Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
37
|
Borgström B, Huang X, Hegardt C, Oredsson S, Strand D. Structure-Activity Relationships in Salinomycin: Cytotoxicity and Phenotype Selectivity of Semi-synthetic Derivatives. Chemistry 2016; 23:2077-2083. [PMID: 27740704 DOI: 10.1002/chem.201603621] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Indexed: 01/09/2023]
Abstract
The ionophore salinomycin has attracted attention for its exceptional ability to selectively reduce the proportion of cells with stem-like properties in cancer cell populations of varying origin. Targeting the tumorigenicity of such cells is of interest as they are implicated in recurrence, metastasis, and drug resistance. Structural derivatives of salinomycin are thus sought after, both as tools for probing the molecular mechanism(s) underlying the observed phenotype effects, and for improving selectivity and activity against cancer stem cells. Synthetic strategies for modification of each of the directly accessible functional groups of salinomycin are presented and the resulting library of analogues was investigated to establish structure-activity relationships, both with respect to cytotoxicity and phenotype selectivity in breast cancer cells. 20-O-Acylated derivatives stand out by exhibiting both improved selectivity and activity. Mechanistically, the importance of the ionophore properties of salinomycin is highlighted by a significant loss of activity by modifications directly interfering with either of the two primary ion coordinating motifs in salinomycin, the C11 ketone and the C1 carboxylate.
Collapse
Affiliation(s)
- Björn Borgström
- Centre for Analysis and Synthesis, Lund University, Box 124, 22100, Lund, Sweden
| | - Xiaoli Huang
- Department of Biology, Lund University, Sölvegatan 35/37, 223 62, Lund, Sweden
| | - Cecilia Hegardt
- Division of Oncology and Pathology, Department of Clinical Sciences, Lund University, Medicon Village, 223 81, Lund, Sweden
| | - Stina Oredsson
- Department of Biology, Lund University, Sölvegatan 35/37, 223 62, Lund, Sweden
| | - Daniel Strand
- Centre for Analysis and Synthesis, Lund University, Box 124, 22100, Lund, Sweden
| |
Collapse
|
38
|
Najumudeen AK, Jaiswal A, Lectez B, Oetken-Lindholm C, Guzmán C, Siljamäki E, Posada IMD, Lacey E, Aittokallio T, Abankwa D. Cancer stem cell drugs target K-ras signaling in a stemness context. Oncogene 2016; 35:5248-5262. [PMID: 26973241 PMCID: PMC5057041 DOI: 10.1038/onc.2016.59] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 01/22/2016] [Accepted: 01/25/2016] [Indexed: 01/02/2023]
Abstract
Cancer stem cells (CSCs) are considered to be responsible for treatment relapse and have therefore become a major target in cancer research. Salinomycin is the most established CSC inhibitor. However, its primary mechanistic target is still unclear, impeding the discovery of compounds with similar anti-CSC activity. Here, we show that salinomycin very specifically interferes with the activity of K-ras4B, but not H-ras, by disrupting its nanoscale membrane organization. We found that caveolae negatively regulate the sensitivity to this drug. On the basis of this novel mechanistic insight, we defined a K-ras-associated and stem cell-derived gene expression signature that predicts the drug response of cancer cells to salinomycin. Consistent with therapy resistance of CSC, 8% of tumor samples in the TCGA-database displayed our signature and were associated with a significantly higher mortality. Using our K-ras-specific screening platform, we identified several new candidate CSC drugs. Two of these, ophiobolin A and conglobatin A, possessed a similar or higher potency than salinomycin. Finally, we established that the most potent compound, ophiobolin A, exerts its K-ras4B-specific activity through inactivation of calmodulin. Our data suggest that specific interference with the K-ras4B/calmodulin interaction selectively inhibits CSC.
Collapse
Affiliation(s)
- A K Najumudeen
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| | - A Jaiswal
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - B Lectez
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| | - C Oetken-Lindholm
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| | - C Guzmán
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| | - E Siljamäki
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| | - I M D Posada
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| | - E Lacey
- Microbial Screening Technologies Pty. Ltd., Building C, Smithfield, New South Wales, Australia
| | - T Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - D Abankwa
- Turku Centre for Biotechnology, Åbo Akademi University, Turku, Finland
| |
Collapse
|
39
|
Zhang XF, Gurunathan S. Combination of salinomycin and silver nanoparticles enhances apoptosis and autophagy in human ovarian cancer cells: an effective anticancer therapy. Int J Nanomedicine 2016; 11:3655-75. [PMID: 27536105 PMCID: PMC4977082 DOI: 10.2147/ijn.s111279] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ovarian cancer is one of the most important malignancies, and the origin, detection, and pathogenesis of epithelial ovarian cancer remain elusive. Although many cancer drugs have been developed to dramatically reduce the size of tumors, most cancers eventually relapse, posing a critical problem to overcome. Hence, it is necessary to identify possible alternative therapeutic approaches to reduce the mortality rate of this devastating disease. To identify alternative approaches, we first synthesized silver nanoparticles (AgNPs) using a novel bacterium called Bacillus clausii. The synthesized AgNPs were homogenous and spherical in shape, with an average size of 16–20 nm, which are known to cause cytotoxicity in various types of human cancer cells, whereas salinomycin (Sal) is able to kill cancer stem cells. Therefore, we selected both Sal and AgNPs to study their combined effect on apoptosis and autophagy in ovarian cancer cells. The cells treated with either Sal or AgNPs showed a dose-dependent effect with inhibitory concentration (IC)-50 values of 6.0 µM and 8 µg/mL for Sal and AgNPs, respectively. To determine the combination effect, we measured the IC25 values of both Sal and AgNPs (3.0 µM and 4 µg/mL), which showed a more dramatic inhibitory effect on cell viability and cell morphology than either Sal or AgNPs alone. The combination of Sal and AgNPs had more pronounced effect on cytotoxicity and expression of apoptotic genes and also significantly induced the accumulation of autophagolysosomes, which was associated with mitochondrial dysfunction and loss of cell viability. Our data show a strong synergistic interaction between Sal and AgNPs in tested cancer cells. The combination treatment increased the therapeutic potential and demonstrated the relevant targeted therapy for the treatment of ovarian cancer. Furthermore, we provide, for the first time, a mode of action for Sal and AgNPs in ovarian cancer cells: enhanced apoptosis and autophagy.
Collapse
Affiliation(s)
- Xi-Feng Zhang
- College of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | | |
Collapse
|
40
|
Shah AM, Wani A, Qazi PH, Rehman SU, Mushtaq S, Ali SA, Hussain A, Shah A, Qazi AK, Makhdoomi US, Hamid A, Kumar A. Isolation and characterization of alborixin from Streptomyces scabrisporus: A potent cytotoxic agent against human colon (HCT-116) cancer cells. Chem Biol Interact 2016; 256:198-208. [DOI: 10.1016/j.cbi.2016.06.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/24/2016] [Accepted: 06/30/2016] [Indexed: 10/21/2022]
|
41
|
Kim SH, Choi YJ, Kim KY, Yu SN, Seo YK, Chun SS, Noh KT, Suh JT, Ahn SC. Salinomycin simultaneously induces apoptosis and autophagy through generation of reactive oxygen species in osteosarcoma U2OS cells. Biochem Biophys Res Commun 2016; 473:607-13. [DOI: 10.1016/j.bbrc.2016.03.132] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 03/27/2016] [Indexed: 11/26/2022]
|
42
|
Muntimadugu E, Kumar R, Saladi S, Rafeeqi TA, Khan W. CD44 targeted chemotherapy for co-eradication of breast cancer stem cells and cancer cells using polymeric nanoparticles of salinomycin and paclitaxel. Colloids Surf B Biointerfaces 2016; 143:532-546. [PMID: 27045981 DOI: 10.1016/j.colsurfb.2016.03.075] [Citation(s) in RCA: 138] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/08/2016] [Accepted: 03/25/2016] [Indexed: 12/11/2022]
Abstract
This combinational therapy is mainly aimed for complete eradication of tumor by killing both cancer cells and cancer stem cells. Salinomycin (SLM) was targeted towards cancer stem cells whereas paclitaxel (PTX) was used to kill cancer cells. Drug loaded poly (lactic-co-glycolic acid) nanoparticles were prepared by emulsion solvent diffusion method using cationic stabilizer. Size of the nanoparticles (below 150nm) was determined by dynamic light scattering technique and transmission electron microscopy. In vitro release study confirmed the sustained release pattern of SLM and PTX from nanoparticles more than a month. Cytotoxicity studies on MCF-7 cells revealed the toxicity potential of nanoparticles over drug solutions. Hyaluronic acid (HA) was coated onto the surface of SLM nanoparticles for targeting CD44 receptors over expressed on cancer stem cells and they showed the highest cytotoxicity with minimum IC50 on breast cancer cells. Synergistic cytotoxic effect was also observed with combination of nanoparticles. Cell uptake studies were carried out using FITC loaded nanoparticles. These particles showed improved cellular uptake over FITC solution and HA coating further enhanced the effect by 1.5 folds. CD44 binding efficiency of nanoparticles was studied by staining MDA-MB-231 cells with anti CD44 human antibody and CD44(+) cells were enumerated using flow cytometry. CD44(+) cell count was drastically decreased when treated with HA coated SLM nanoparticles indicating their efficiency towards cancer stem cells. Combination of HA coated SLM nanoparticles and PTX nanoparticles showed the highest cytotoxicity against CD44(+) cells. Hence combinational therapy using conventional chemotherapeutic drug and cancer stem cell inhibitor could be a promising approach in overcoming cancer recurrence due to resistant cell population.
Collapse
Affiliation(s)
- Eameema Muntimadugu
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | - Rajendra Kumar
- UGC Centre of Excellence in Applications of Nanomaterials, Nanoparticles, and Nanocomposites, Panjab University, Chandigarh 160014, India
| | - Shantikumar Saladi
- Department of Pharmaceutical Analysis, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | - Towseef Amin Rafeeqi
- Biochemistry, Cellular and Molecular Biology Laboratories, Central Research Institute of Unani Medicine (CRIUM), Hyderabad 500038, India
| | - Wahid Khan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India.
| |
Collapse
|
43
|
Huang X, Borgström B, Kempengren S, Persson L, Hegardt C, Strand D, Oredsson S. Breast cancer stem cell selectivity of synthetic nanomolar-active salinomycin analogs. BMC Cancer 2016; 16:145. [PMID: 26906175 PMCID: PMC4765157 DOI: 10.1186/s12885-016-2142-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 02/08/2016] [Indexed: 12/28/2022] Open
Abstract
Background Cancer stem cells (CSCs) have been invoked in resistance, recurrence and metastasis of cancer. Consequently, curative cancer treatments may be contingent on CSC selective approaches. Of particular interest in this respect is the ionophore salinomycin, a natural product shown to be 100-fold more active against CSCs than clinically used paclitaxel. We have previously reported that synthetic salinomycin derivatives display increased activity against breast cancer cell lines. Herein we specifically investigate the CSC selectivity of the most active member in each class of C20-O-acylated analogs as well as a C1-methyl ester analog incapable of charge-neutral metal ion transport. Methods JIMT-1 breast cancer cells were treated with three C20-O-acylated analogs, the C1-methyl ester of salinomycin, and salinomycin. The effects of treatment on the CSC-related CD44+/CD24− and the aldehyde dehydrogenase positive (ALDH+) populations were determined using flow cytometry. The survival ability of CSCs after treatment was investigated with a colony formation assay under serum free conditions. The effect of the compounds on cell migration was evaluated using wound-healing and Boyden chamber assays. The expression of vimentin, related to mesenchymal traits and expression of E-cadherin and β-catenin, related to the epithelial traits, were investigated using immunofluorescence microscopy. Results Treatment with each of the three C20-acylated analogs efficiently decreased the putative CSC population as reflected by reduction of the CD44+/CD24− and ALDH+ populations already at a 50 nM concentration. In addition, colony forming efficiency and cell migration were reduced, and the expression of the epithelial markers E-cadherin and β-catenin at the cell surface were increased. In contrast, salinomycin used at the same concentration did not significantly influence the CSC population and the C1-methyl ester was inactive even at a 20 μM concentration. Conclusions Synthetic structural analogs of salinomycin, previously shown to exhibit increased activity against cancer cells, also exhibited improved activity against CSCs across several assays even at nanomolar concentrations where salinomycin was found inactive. The methyl ester analog of salinomycin, incapable of charge-neutral metal ion transport, did not show activity in CSC assays, lending experimental support to ionophoric stress as the molecular initiating event for the CSC effects of salinomycin and related structures. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2142-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xiaoli Huang
- Department of Biology, Lund University, Lund, Sweden.
| | - Björn Borgström
- Department of Chemistry, Center for Analysis and Synthesis, Lund University, Lund, Sweden.
| | | | - Lo Persson
- Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - Cecilia Hegardt
- Division of Oncology and Pathology, Department of Clinical Sciences Lund, Lund University Cancer Center/Medicon Village, Lund, Sweden.
| | - Daniel Strand
- Department of Chemistry, Center for Analysis and Synthesis, Lund University, Lund, Sweden.
| | | |
Collapse
|
44
|
Al Faraj A, Shaik AS, Ratemi E, Halwani R. Combination of drug-conjugated SWCNT nanocarriers for efficient therapy of cancer stem cells in a breast cancer animal model. J Control Release 2016; 225:240-51. [PMID: 26827662 DOI: 10.1016/j.jconrel.2016.01.053] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/24/2016] [Accepted: 01/27/2016] [Indexed: 01/06/2023]
Abstract
Targeting breast cancer and more specifically cancer stem cell (CSC) subpopulation, responsible for tumor growth, resistance and self-renewal, using combination of therapeutic drugs selectively delivered via biocompatible nanocarriers, provides a novel approach for effective therapy. Here, we propose to evaluate the potential therapeutic efficacy of combining Paclitaxel and Salinomycin drugs actively targeted to both breast cancer and CSCs in xenograft murine model after conjugation with biocompatible CD44 antibody conjugated SWCNTs via hydrazone linker allowing pH-responsive release mechanism near the acidic tumor microenvironment. Both in vitro investigations on MDA-MB-231, sorted CSC negative or CSC positive fractions and in vivo evaluations on tumor-bearing mice using noninvasive bioluminescence and magnetic resonance imaging confirmed the enhanced therapeutic effect of the combined therapy compared to treatment with individual drug-conjugated nanocarriers or free drug suspensions. Thus, confirmed the great promise of the developed SWCNTs drug delivery system for effective breast cancer treatment by targeting and eradicating both whole tumor cells and CSCs populations.
Collapse
Affiliation(s)
- Achraf Al Faraj
- King Saud University, College of Applied Medical Sciences, Department of Radiological Sciences, Riyadh, Saudi Arabia.
| | - Asma Sultana Shaik
- King Saud University, Prince Naif Health Research Center, Riyadh, Saudi Arabia
| | - Elaref Ratemi
- Jubail Industrial College, Department of Chemical and Process Engineering Technology, Jubail Industrial City, Saudi Arabia
| | - Rabih Halwani
- King Saud University, College of Medicine, Prince Naif Center for Immunology Research, Department of Pediatrics, Riyadh, Saudi Arabia
| |
Collapse
|
45
|
Zhang Y, Zuo Y, Guan Z, Lu W, Xu Z, Zhang H, Yang Y, Yang M, Zhu H, Chen X. Salinomycin radiosensitizes human nasopharyngeal carcinoma cell line CNE-2 to radiation. Tumour Biol 2016; 37:305-11. [PMID: 26209294 DOI: 10.1007/s13277-015-3730-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023] Open
Abstract
Nasopharyngeal carcinoma (NPC) is primarily treated by chemoradiation. However, how to promote radiation sensitivity in NPC remains a challenge. Salinomycin is potentially useful for the treatment of cancer. This study aimed to explore the radiosensitivity of salinomycin on human nasopharyngeal carcinoma cell line CNE-2. CNE-2 were treated with salinomycin or irradiation, alone or in combination. The cytotoxicity effects of salinomycin were measured using CCK-8 assay. Clonogenic survival assay was used to evaluate the effects of salinomycin on the radiosensitivity of CNE-2. The changes of cell cycle distribution and apoptosis were assayed using flow cytometry. The expression of Caspase3/Bax/Bal-2 was detected by Western blotting. DNA damage was detected via γ-H2AX foci counting. The results showed that salinomycin induced apoptosis and G2/M arrest, increased Bax and cleaved Caspase3, decreased Bcl-2 expression, and increased the formation of γ-H2AX nuclear foci. These data suggest that salinomycin may be a radiosensitizer for NPC radiotherapy.
Collapse
Affiliation(s)
- Yongqin Zhang
- Department of Radiation Oncology, The First People's Hospital of Zhangjiagang, #68 Jiyangxi Road, Zhangjiagang, Jiangsu province, 215600, China
| | - Yun Zuo
- Department of Radiation Oncology, The First People's Hospital of Zhangjiagang, #68 Jiyangxi Road, Zhangjiagang, Jiangsu province, 215600, China.
| | - Zhifeng Guan
- Department of Radiation Oncology, Nantong Tumor Hospital, Affiliated Tumor Hospital of Nantong University, Nantong, 226321, China
| | - Weidong Lu
- Department of Radiation Oncology, The First People's Hospital of Zhangjiagang, #68 Jiyangxi Road, Zhangjiagang, Jiangsu province, 215600, China
| | - Zheng Xu
- Department of Radiation Oncology, The First People's Hospital of Zhangjiagang, #68 Jiyangxi Road, Zhangjiagang, Jiangsu province, 215600, China
| | - Hao Zhang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yan Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Meilin Yang
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hongcheng Zhu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiaochen Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| |
Collapse
|
46
|
de Aberasturi AL, Redrado M, Villalba M, Larzabal L, Pajares MJ, Garcia J, Evans SR, Garcia-Ros D, Bodegas ME, Lopez L, Montuenga L, Calvo A. TMPRSS4 induces cancer stem cell-like properties in lung cancer cells and correlates with ALDH expression in NSCLC patients. Cancer Lett 2015; 370:165-76. [PMID: 26546046 DOI: 10.1016/j.canlet.2015.10.012] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Revised: 10/09/2015] [Accepted: 10/15/2015] [Indexed: 01/04/2023]
Abstract
Metastasis involves a series of changes in cancer cells that promote their escape from the primary tumor and colonization to a new organ. This process is related to the transition from an epithelial to a mesenchymal phenotype (EMT). Recently, some authors have shown that migratory cells with an EMT phenotype share properties of cancer stem cells (CSCs), which allow them to form a new tumor mass. The type II transmembrane serine protease TMPRSS4 is highly expressed in some solid tumors, promotes metastasis and confers EMT features to cancer cells. We hypothesized that TMPRSS4 could also provide CSC properties. Overexpression of TMPRSS4 reduces E-cadherin and induces N-cadherin and vimentin in A549 lung cancer cells, supporting an EMT phenotype. These changes are accompanied by enhanced migration, invasion and tumorigenicity in vivo. TMPRSS4 expression was highly increased in a panel of lung cancer cells cultured as tumorspheres (a typical assay to enrich for CSCs). H358 and H441 cells with knocked-down TMPRSS4 levels were significantly less able to form primary and secondary tumorspheres than control cells. Moreover, they showed a lower proportion of ALDH+ cells (examined by FACS analysis) and lower expression of some CSC markers than controls. A549 cells overexpressing TMPRSS4 conferred the opposite phenotype and were also more sensitive to the CSC-targeted drug salinomycin than control cells, but were more resistant to regular chemotherapeutic drugs (cisplatin, gemcitabine and 5-fluorouracil). Analysis of 70 NSCLC samples from patients revealed a very significant correlation between TMPRSS4 expression and CSC markers ALDH (p = 0.0018) and OCT4 (p = 0.0004), suggesting that TMPRSS4 is associated with a CSC phenotype in patients' tumors. These results show that TMPRSS4, in addition to inducing EMT, can also promote CSC features in lung cancer; therefore, CSC-targeting drugs could be an appropriate treatment for TMPRSS4+ tumors.
Collapse
Affiliation(s)
- Arrate L de Aberasturi
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Miriam Redrado
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Maria Villalba
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Leyre Larzabal
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Maria J Pajares
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Javier Garcia
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - Stephanie R Evans
- Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - David Garcia-Ros
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | - Maria Elena Bodegas
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain
| | | | - Luis Montuenga
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| | - Alfonso Calvo
- Department of Histology and Pathology, School of Medicine, University of Navarra, C/Irunlarrea 1, 31008 Pamplona, Spain; Program of Solid Tumors and Biomarkers, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain.
| |
Collapse
|
47
|
Krishnamurthy S, Ke X, Yang YY. Delivery of therapeutics using nanocarriers for targeting cancer cells and cancer stem cells. Nanomedicine (Lond) 2015; 10:143-60. [PMID: 25597774 DOI: 10.2217/nnm.14.154] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Development of cancer resistance, cancer relapse and metastasis are attributed to the presence of cancer stem cells (CSCs). Eradication of this subpopulation has been shown to increase life expectancy of patients. Since the discovery of CSCs a decade ago, several strategies have been devised to specifically target them but with limited success. Nanocarriers have recently been employed to deliver anti-CSC therapeutics for reducing the population of CSCs at the tumor site with great success. This review discusses the different therapeutic strategies that have been employed using nanocarriers, their advantages, success in targeting CSCs and the challenges that are to be overcome. Exploiting this new modality of cancer treatment in the coming decade may improve outcomes profoundly with promise of effective treatment response and reducing relapse and metastasis.
Collapse
Affiliation(s)
- Sangeetha Krishnamurthy
- Institute of Bioengineering & Nanotechnology, 31 Biopolis Way, The Nanos, Singapore 138669, Singapore
| | | | | |
Collapse
|
48
|
Boesch M, Zeimet AG, Rumpold H, Gastl G, Sopper S, Wolf D. Drug Transporter-Mediated Protection of Cancer Stem Cells From Ionophore Antibiotics. Stem Cells Transl Med 2015; 4:1028-32. [PMID: 26136502 DOI: 10.5966/sctm.2015-0054] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2015] [Accepted: 05/27/2015] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Ionophore antibiotics were reported to selectively kill cancer stem cells and to overcome multidrug resistance, but mechanistic studies of the significance of drug transporters for treatment with these compounds are lacking. We applied chemosensitivity testing of well-characterized human cancer cell lines to elaborate on whether drug transporters are involved in protection from the cytotoxic effects of the ionophore antibiotics salinomycin and nigericin. Our experiments demonstrated that ionophore antibiotics were ineffective against both stem-like ovarian cancer side population cells (expressing either ABCB1 or ABCG2) and K562/Dox-H1 cells, which constitute a genetically defined model system for ABCB1 expression. Considering that cancer stem cells often express high levels of drug transporters, we deduced from our results that ionophore antibiotics are less suited to cancer stem cell-targeted treatment than previously thought. SIGNIFICANCE Ionophore antibiotics such as salinomycin have repeatedly been shown to target cancer stem and progenitor cells from various tumor entities. Meanwhile, cancer stem cell (CSC)-selective toxicity of ionophore antibiotics seems to be a commonly accepted concept that is about to encourage their clinical testing. This study provides data that challenge the concept of targeted elimination of CSC by ionophore antibiotics. Stem-like ovarian cancer side population (SP) cells expressing high levels of ABC drug transporters are shown to largely resist the cytotoxic effects of salinomycin and nigericin. Furthermore, using a small interfering RNA-based knockdown model specific for ABCB1, this study demonstrates that ABC drug transporters are indeed causally involved in mediating protection from ionophore antibiotics. Considering that it is a hallmark of CSCs to exhibit drug resistance conferred by ABC drug transporters, it must be deduced from these results that CSCs may also be protected from ionophore antibiotics by means of drug-transporter mediated efflux.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/antagonists & inhibitors
- ATP-Binding Cassette Transporters/genetics
- ATP-Binding Cassette Transporters/metabolism
- Antineoplastic Agents/metabolism
- Antineoplastic Agents/pharmacology
- Biological Transport
- Cell Line, Tumor
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Ionophores/metabolism
- Ionophores/pharmacology
- K562 Cells
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Nigericin/metabolism
- Nigericin/pharmacology
- Pyrans/metabolism
- Pyrans/pharmacology
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
Collapse
Affiliation(s)
- Maximilian Boesch
- Institute of Immunobiology, St. Gallen, Switzerland; Tyrolean Cancer Research Institute, Innsbruck, Austria; Internal Medicine V and
| | - Alain G Zeimet
- Department of Gynecology and Obstetrics, Medical University Innsbruck, Innsbruck, Austria
| | - Holger Rumpold
- Internal Medicine I, Klinikum der Barmherzigen Schwestern Linz, Linz, Austria
| | | | - Sieghart Sopper
- Tyrolean Cancer Research Institute, Innsbruck, Austria; Internal Medicine V and
| | - Dominik Wolf
- Tyrolean Cancer Research Institute, Innsbruck, Austria; Internal Medicine V and Medical Clinic III, Oncology, Hematology and Rheumatology, University Clinic Bonn, Bonn, Germany
| |
Collapse
|
49
|
SCHERZED AGMAL, HACKENBERG STEPHAN, FROELICH KATRIN, RAK KRISTEN, GINZKEY CHRISTIAN, HAGEN RUDOLF, SCHENDZIELORZ PHILIPP, KLEINSASSER NORBERT. Effects of salinomycin and CGP37157 on head and neck squamous cell carcinoma cell lines in vitro. Mol Med Rep 2015; 12:4455-4461. [DOI: 10.3892/mmr.2015.3981] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 05/13/2015] [Indexed: 11/06/2022] Open
|
50
|
Qin LS, Jia PF, Zhang ZQ, Zhang SM. ROS-p53-cyclophilin-D signaling mediates salinomycin-induced glioma cell necrosis. J Exp Clin Cancer Res 2015; 34:57. [PMID: 26024660 PMCID: PMC4486428 DOI: 10.1186/s13046-015-0174-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 05/16/2015] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The primary glioblastoma multiforme (GBM) is the most malignant form of astrocytic tumor with an average survival of approximately 12-14 months. The search for novel and more efficient chemo-agents against this disease is urgent. Salinomycin induces broad anti-cancer effects; however, its role in GBM and the underlying mechanism are not clear. RESULTS Here we found that salinomycin induced both apoptosis and necrosis in cultured glioma cells, and necrosis played a major role in contributing salinomycin's cytotoxicity. Salinomycin induced p53 translocation to mitochondria, where it formed a complex with cyclophilin-D (CyPD). This complexation was required for mitochondrial permeability transition pore (mPTP) opening and subsequent programmed necrosis. Blockade of Cyp-D by siRNA-mediated depletion or pharmacological inhibitors (cyclosporin A and sanglifehrin A) significantly suppressed salinomycin-induced glioma cell necrosis. Meanwhile, p53 stable knockdown alleviated salinomycin-induced necrosis in glioma cells. Reactive oxygen species (ROS) production was required for salinomycin-induced p53 mitochondrial translocation, mPTP opening and necrosis, and anti-oxidants n-acetylcysteine (NAC) and pyrrolidine dithiocarbamate (PDTC) inhibited p53 translocation, mPTP opening and glioma cell death. CONCLUSIONS Thus, salinomycin mainly induces programmed necrosis in cultured glioma cells.
Collapse
Affiliation(s)
- Li-sen Qin
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No. 188, Shi-zi Street, Suzhou, Jiangsu, People's Republic of China.
- Department of Neurosurgery, the Sixth People's Hospital of Yancheng, Yan-cheng, Jiangsu, People's Republic of China.
| | - Pi-feng Jia
- Department of Neurosurgery, Shanghai Ruijin Hospital North, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China.
| | - Zhi-qing Zhang
- Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases and Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, 215021, China.
| | - Shi-ming Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Soochow University, No. 188, Shi-zi Street, Suzhou, Jiangsu, People's Republic of China.
| |
Collapse
|