101
|
Schein CH. Repurposing approved drugs on the pathway to novel therapies. Med Res Rev 2020; 40:586-605. [PMID: 31432544 PMCID: PMC7018532 DOI: 10.1002/med.21627] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 12/22/2022]
Abstract
The time and cost of developing new drugs have led many groups to limit their search for therapeutics to compounds that have previously been approved for human use. Many "repurposed" drugs, such as derivatives of thalidomide, antibiotics, and antivirals have had clinical success in treatment areas well beyond their original approved use. These include applications in treating antibiotic-resistant organisms, viruses, cancers and to prevent burn scarring. The major theoretical justification for reusing approved drugs is that they have known modes of action and controllable side effects. Coadministering antibiotics with inhibitors of bacterial toxins or enzymes that mediate multidrug resistance can greatly enhance their activity. Drugs that control host cell pathways, including inflammation, tumor necrosis factor, interferons, and autophagy, can reduce the "cytokine storm" response to injury, control infection, and aid in cancer therapy. An active compound, even if previously approved for human use, will be a poor clinical candidate if it lacks specificity for the new target, has poor solubility or can cause serious side effects. Synergistic combinations can reduce the dosages of the individual components to lower reactivity. Preclinical analysis should take into account that severely ill patients with comorbidities will be more sensitive to side effects than healthy trial subjects. Once an active, approved drug has been identified, collaboration with medicinal chemists can aid in finding derivatives with better physicochemical properties, specificity, and efficacy, to provide novel therapies for cancers, emerging and rare diseases.
Collapse
Affiliation(s)
- Catherine H Schein
- Department of Biochemistry and Molecular Biology, Institute for Human Infection and Immunity (IHII), University of Texas Medical Branch at Galveston, Galveston, Texas
| |
Collapse
|
102
|
Carlisle BG, Doussau A, Kimmelman J. Patient burden and clinical advances associated with postapproval monotherapy cancer drug trials: a retrospective cohort study. BMJ Open 2020; 10:e034306. [PMID: 32071183 PMCID: PMC7044865 DOI: 10.1136/bmjopen-2019-034306] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVES After regulatory approval, drug companies, public funding agencies and academic researchers often pursue trials aimed at extending the uses of a new drug by testing it in new non-approved indications. Patient burden and clinical impact of such research are not well understood. DESIGN AND SETTING We conducted a retrospective cohort study of postapproval clinical trials launched within 5 years after the drug's first approval, testing anticancer drugs in monotherapy in indications that were first pursued after a drug's first Food and Drug Administration (FDA) license, for all 12 anticancer drugs approved between 2005 and 2007. FDA, Medline and Embase search date 2019 February 12. PRIMARY AND SECONDARY OUTCOME MEASURES Our primary objective was to measure burden and clinical impact for patients enrolling in these trials. Each trial was sorted into a 'trajectory' defined by the drug and cancer indication. The risk was operationalised by proportions of grade 3-4 severe adverse events and deaths. The clinical impact was measured by estimating the proportion of patients participating in trajectories that resulted in FDA approval, uptake into National Comprehensive Cancer Network (NCCN) clinical practice guidelines or advancement to randomised controlled trials within 8 years. RESULTS Our search captured 104 published trials exploring monotherapy, including 69 unique trajectories. In total, trials in our sample enrolled 4699 patients. Grade 3-4 adverse events were experienced by 19.6% of patients; grade 5 events were experienced by 2.8% of patients. None of the trajectories launched after initial drug approval received FDA approval. Five trajectories were recommended by the NCCN within 8 years of the first trial within that trajectory. Eleven trajectories were advanced to randomised controlled testing. CONCLUSIONS The challenges associated with unlocking new applications for drugs that first received approval from 2005 to 2007 were similar to those for developing new drugs altogether. Our findings can help inform priority setting in research and provide a basis for calibrating expectations when considering enrolment in label-extending trials.
Collapse
Affiliation(s)
| | | | - Jonathan Kimmelman
- Biomedical Ethics Unit / SSOM, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
103
|
Rushworth LK, Hewit K, Munnings-Tomes S, Somani S, James D, Shanks E, Dufès C, Straube A, Patel R, Leung HY. Repurposing screen identifies mebendazole as a clinical candidate to synergise with docetaxel for prostate cancer treatment. Br J Cancer 2020; 122:517-527. [PMID: 31844184 PMCID: PMC7028732 DOI: 10.1038/s41416-019-0681-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 11/05/2019] [Accepted: 11/19/2019] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Docetaxel chemotherapy in prostate cancer has a modest impact on survival. To date, efforts to develop combination therapies have not translated into new treatments. We sought to develop a novel therapeutic strategy to tackle chemoresistant prostate cancer by enhancing the efficacy of docetaxel. METHODS We performed a drug-repurposing screen by using murine-derived prostate cancer cell lines driven by clinically relevant genotypes. Cells were treated with docetaxel alone, or in combination with drugs (n = 857) from repurposing libraries, with cytotoxicity quantified using High Content Imaging Analysis. RESULTS Mebendazole (an anthelmintic drug that inhibits microtubule assembly) was selected as the lead drug and shown to potently synergise docetaxel-mediated cell killing in vitro and in vivo. Dual targeting of the microtubule structure was associated with increased G2/M mitotic block and enhanced cell death. Strikingly, following combined docetaxel and mebendazole treatment, no cells divided correctly, forming multipolar spindles that resulted in aneuploid daughter cells. Liposomes entrapping docetaxel and mebendazole suppressed in vivo prostate tumour growth and extended progression-free survival. CONCLUSIONS Docetaxel and mebendazole target distinct aspects of the microtubule dynamics, leading to increased apoptosis and reduced tumour growth. Our data support a new concept of combined mebendazole/docetaxel treatment that warrants further clinical evaluation.
Collapse
Affiliation(s)
- Linda K Rushworth
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Kay Hewit
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Sophie Munnings-Tomes
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, CV4 7AL, UK
| | - Sukrut Somani
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Daniel James
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Emma Shanks
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Christine Dufès
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Anne Straube
- Centre for Mechanochemical Cell Biology, University of Warwick, Coventry, CV4 7AL, UK
| | - Rachana Patel
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK
| | - Hing Y Leung
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Bearsden, Glasgow, G61 1QH, UK.
- CRUK Beatson Institute, Bearsden, Glasgow, G61 1BD, UK.
| |
Collapse
|
104
|
Rámirez H, Rodrigues JR, Mijares MR, De Sanctis JB, Charris JE. Synthesis and biological activity of 2-[2-(7-chloroquinolin-4-ylthio)-4-methylthiazol-5-yl]-N-phenylacetamide derivatives as antimalarial and cytotoxic agents. JOURNAL OF CHEMICAL RESEARCH 2020. [DOI: 10.1177/1747519819899073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
A novel series of 2-[2-(7-chloroquinolin-4-ylthio)-4-methylthiazol-5-yl]- N-phenylacetamide derivatives is synthesized via substitution with 2-mercapto-4-methyl-5-thiazoleacetic acid at position 4 of 4,7-dichloroquinoline to obtain an intermediate acetic acid derivative. The chemical behavior of these reactants was investigated using different reaction conditions to optimize the nucleophilic substitution at position 4. The final compounds are prepared using a modified version of the Steglich esterification reaction between the acetic acid intermediate 3 and different anilines. The structures are confirmed by infrared, 1H, 13C, distortionless enhancement by polarization transfer 135°, Correlated Spectroscopy, heteronuclear correlation spectroscopy and (Long range HETCOR using three BIRD pulses) FLOCK-NMR spectral studies, and by elemental analysis. The synthesized compounds are tested in vitro and in vivo for their potential antimalarial and anticancer activities, with derivative 11 being the most promising candidate.
Collapse
Affiliation(s)
- Hegira Rámirez
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, Caracas, Venezuela
- Faculty of Medicine, Universidad de las Américas, Quito, Ecuador
| | - Juan R Rodrigues
- Laboratory of Pharmacology and Toxicology, Department of Cell Biology, Simón Bolívar University, Caracas, Venezuela
| | - Michael R Mijares
- Biotechnology Unit, Faculty of Pharmacy, Central University of Venezuela, Caracas, Venezuela
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, Caracas, Venezuela
| | - Juan B De Sanctis
- Institute of Immunology, Faculty of Medicine, Central University of Venezuela, Caracas, Venezuela
- Institute of Molecular and Translational Medicine, Faculty of Medicine, Palacky University Olomouc, Olomouc, Czech Republic
| | - Jaime E Charris
- Organic Synthesis Laboratory, Faculty of Pharmacy, Central University of Venezuela, Caracas, Venezuela
| |
Collapse
|
105
|
Benavides-Serrato A, Saunders JT, Holmes B, Nishimura RN, Lichtenstein A, Gera J. Repurposing Potential of Riluzole as an ITAF Inhibitor in mTOR Therapy Resistant Glioblastoma. Int J Mol Sci 2020; 21:ijms21010344. [PMID: 31948038 PMCID: PMC6981868 DOI: 10.3390/ijms21010344] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 12/27/2019] [Accepted: 12/31/2019] [Indexed: 12/17/2022] Open
Abstract
Internal ribosome entry site (IRES)-mediated protein synthesis has been demonstrated to play an important role in resistance to mechanistic target of rapamycin (mTOR) targeted therapies. Previously, we have demonstrated that the IRES trans-acting factor (ITAF), hnRNP A1 is required to promote IRES activity and small molecule inhibitors which bind specifically to this ITAF and curtail IRES activity, leading to mTOR inhibitor sensitivity. Here we report the identification of riluzole (Rilutek®), an FDA-approved drug for amyotrophic lateral sclerosis (ALS), via an in silico docking analysis of FDA-approved compounds, as an inhibitor of hnRNP A1. In a riluzole-bead coupled binding assay and in surface plasmon resonance imaging analyses, riluzole was found to directly bind to hnRNP A1 and inhibited IRES activity via effects on ITAF/RNA-binding. Riluzole also demonstrated synergistic anti-glioblastoma (GBM) affects with mTOR inhibitors in vitro and in GBM xenografts in mice. These data suggest that repurposing riluzole, used in conjunction with mTOR inhibitors, may serve as an effective therapeutic option in glioblastoma.
Collapse
Affiliation(s)
- Angelica Benavides-Serrato
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA 91343, USA
| | - Jacquelyn T. Saunders
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA 91343, USA
| | - Brent Holmes
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA 91343, USA
| | - Robert N. Nishimura
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA 91343, USA
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Alan Lichtenstein
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA 91343, USA
- Jonnson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Joseph Gera
- Department of Research & Development, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA 91343, USA
- Jonnson Comprehensive Cancer Center, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- Molecular Biology Institute, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +00-1-818-895-9416
| |
Collapse
|
106
|
Yang W, Veroniaina H, Qi X, Chen P, Li F, Ke PC. Soft and Condensed Nanoparticles and Nanoformulations for Cancer Drug Delivery and Repurpose. ADVANCED THERAPEUTICS 2020; 3:1900102. [PMID: 34291146 PMCID: PMC8291088 DOI: 10.1002/adtp.201900102] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Indexed: 12/24/2022]
Abstract
Drug repurpose or reposition is recently recognized as a high-performance strategy for developing therapeutic agents for cancer treatment. This approach can significantly reduce the risk of failure, shorten R&D time, and minimize cost and regulatory obstacles. On the other hand, nanotechnology-based delivery systems are extensively investigated in cancer therapy due to their remarkable ability to overcome drug delivery challenges, enhance tumor specific targeting, and reduce toxic side effects. With increasing knowledge accumulated over the past decades, nanoparticle formulation and delivery have opened up a new avenue for repurposing drugs and demonstrated promising results in advanced cancer therapy. In this review, recent developments in nano-delivery and formulation systems based on soft (i.e., DNA nanocages, nanogels, and dendrimers) and condensed (i.e., noble metal nanoparticles and metal-organic frameworks) nanomaterials, as well as their theranostic applications in drug repurpose against cancer are summarized.
Collapse
Affiliation(s)
- Wen Yang
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | | | - Xiaole Qi
- Key Laboratory of Modern Chinese Medicines, China Pharmaceutical University, Nanjing 210009, China; ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| | - Pengyu Chen
- Materials Research and Education Center, Auburn University, Auburn, AL 36849, USA
| | - Feng Li
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn AL 36849, USA
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade Parkville, VIC 3052, Australia
| |
Collapse
|
107
|
Agarwal G, Carcache PJB, Addo EM, Kinghorn AD. Current status and contemporary approaches to the discovery of antitumor agents from higher plants. Biotechnol Adv 2020; 38:107337. [PMID: 30633954 PMCID: PMC6614024 DOI: 10.1016/j.biotechadv.2019.01.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 01/03/2019] [Accepted: 01/07/2019] [Indexed: 12/13/2022]
Abstract
Higher plant constituents have afforded clinically available anticancer drugs. These include both chemically unmodified small molecules and their synthetic derivatives currently used or those in clinical trials as antineoplastic agents, and an updated summary is provided. In addition, botanical dietary supplements, exemplified by mangosteen and noni constituents, are also covered as potential cancer chemotherapeutic agents. Approaches to metabolite purification, rapid dereplication, and biological evaluation including analytical hyphenated techniques, molecular networking, and advanced cellular and animal models are discussed. Further, enhanced and targeted drug delivery systems for phytochemicals, including micelles, nanoparticles and antibody drug conjugates (ADCs) are described herein.
Collapse
Affiliation(s)
- Garima Agarwal
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Peter J Blanco Carcache
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - Ermias Mekuria Addo
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States
| | - A Douglas Kinghorn
- Division of Medicinal Chemistry and Pharmacognosy, College of Pharmacy, The Ohio State University, Columbus, OH 43210, United States.
| |
Collapse
|
108
|
Olgen S, Kotra LP. Drug Repurposing in the Development of Anticancer Agents. Curr Med Chem 2019; 26:5410-5427. [PMID: 30009698 DOI: 10.2174/0929867325666180713155702] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/14/2018] [Accepted: 06/28/2018] [Indexed: 01/07/2023]
Abstract
BACKGROUND Research into repositioning known drugs to treat cancer other than the originally intended disease continues to grow and develop, encouraged in part, by several recent success stories. Many of the studies in this article are geared towards repurposing generic drugs because additional clinical trials are relatively easy to perform and the drug safety profiles have previously been established. OBJECTIVE This review provides an overview of anticancer drug development strategies which is one of the important areas of drug restructuring. METHODS Repurposed drugs for cancer treatments are classified by their pharmacological effects. The successes and failures of important repurposed drugs as anticancer agents are evaluated in this review. RESULTS AND CONCLUSION Drugs could have many off-target effects, and can be intelligently repurposed if the off-target effects can be employed for therapeutic purposes. In cancer, due to the heterogeneity of the disease, often targets are quite diverse, hence a number of already known drugs that interfere with these targets could be deployed or repurposed with appropriate research and development.
Collapse
Affiliation(s)
- Sureyya Olgen
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Biruni University, Istanbul, Turkey
| | - Lakshmi P Kotra
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, M5S 3M2, Canada.,Center for Molecular Design and Preformulations, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, M5G 1L7 Canada.,Multi-Organ Transplant Program, Toronto General Hospital, Toronto, Ontario, M5G 1L7 Canada
| |
Collapse
|
109
|
Ye N, Xu Q, Li W, Wang P, Zhou J. Recent Advances in Developing K-Ras Plasma Membrane Localization Inhibitors. Curr Top Med Chem 2019; 19:2114-2127. [PMID: 31475899 DOI: 10.2174/1568026619666190902145116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/02/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022]
Abstract
The Ras proteins play an important role in cell growth, differentiation, proliferation and survival by regulating diverse signaling pathways. Oncogenic mutant K-Ras is the most frequently mutated class of Ras superfamily that is highly prevalent in many human cancers. Despite intensive efforts to combat various K-Ras-mutant-driven cancers, no effective K-Ras-specific inhibitors have yet been approved for clinical use to date. Since K-Ras proteins must be associated to the plasma membrane for their function, targeting K-Ras plasma membrane localization represents a logical and potentially tractable therapeutic approach. Here, we summarize the recent advances in the development of K-Ras plasma membrane localization inhibitors including natural product-based inhibitors achieved from high throughput screening, fragment-based drug design, virtual screening, and drug repurposing as well as hit-to-lead optimizations.
Collapse
Affiliation(s)
- Na Ye
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.,Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China.,Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Qingfeng Xu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Wanwan Li
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu 215123, China
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| |
Collapse
|
110
|
Vargas V, Leopold C, Castillo-Riquelme M, Darrow JJ. Expanding Coverage of Oncology Drugs in an Aging, Upper-Middle-Income Country: Analyses of Public and Private Expenditures in Chile. J Glob Oncol 2019; 5:1-17. [PMID: 31860377 PMCID: PMC6939746 DOI: 10.1200/jgo.19.00223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/29/2019] [Indexed: 11/24/2022] Open
Abstract
PURPOSE The population of Chile has aged, and in 2017, cancer became the leading cause of death. Since 2005, a national health program has expanded coverage of drugs for 13 types of cancer and related palliative care. We describe the trends in public and private oncology drug expenditures in Chile and consider how increasing expenditures might be addressed. METHODS We analyzed total quarterly drug expenditures for 131 oncology drugs from quarter (Q)3 2012 until Q1 2017, including public and private insurance payments and patient out-of-pocket spending. The data were analyzed by drug-mix, sources of funding, growth, and intellectual property status. The Laspeyres Price Index was used to analyze expenditure growth. RESULTS We found 131 oncology drugs associated with 87,129 observations. Spending on drugs rose 120% from the first period, spanning from the first 3 quarters (Q3, Q4, Q1 2012-2013) to the last period (Q3, Q4, Q1 2016-2017), corresponding to an annualized rate of 19.2% and totaling US$398 million (in 2017 dollars). The public sector accounted for 84.2% of spending, which included 50 drugs in the official treatment protocols, whereas private insurance accounted for 7.3% in on-protocol drugs. The remaining 8.5% was paid out of pocket. In the public sector, more than 90% of growth resulted from increased use. Seven drugs, including 3 with nonexpired patents, accounted for 50% of total expenditures. CONCLUSION Increased use and access enabled by expanded public expenditures drove most of the growth in oncology drug expenditures. However, the rate of public expenditure growth may be fiscally unsustainable. Policies are urgently needed to promote the use of generic drugs, the appropriate mix of on-protocol versus off-protocol drugs, and the curbing of off-label prescribing.
Collapse
Affiliation(s)
- Veronica Vargas
- Department of Economics, Alberto Hurtado University, Santiago, Chile, and David Rockefeller Center for Latin America Studies, Harvard University, Boston, MA
| | - Christine Leopold
- Department of Population Medicine, Division of Health Policy and Insurance Research, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA
| | - Marianela Castillo-Riquelme
- Epidemiology Program, Institute of Population Health, School of Public Health, University of Chile, Santiago, Chile
| | - Jonathan J. Darrow
- Program on Regulation, Therapeutics, and Law, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
111
|
Kumar R, Harilal S, Gupta SV, Jose J, Thomas Parambi DG, Uddin MS, Shah MA, Mathew B. Exploring the new horizons of drug repurposing: A vital tool for turning hard work into smart work. Eur J Med Chem 2019; 182:111602. [PMID: 31421629 PMCID: PMC7127402 DOI: 10.1016/j.ejmech.2019.111602] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/07/2019] [Accepted: 08/07/2019] [Indexed: 02/07/2023]
Abstract
Drug discovery and development are long and financially taxing processes. On an average it takes 12-15 years and costs 1.2 billion USD for successful drug discovery and approval for clinical use. Many lead molecules are not developed further and their potential is not tapped to the fullest due to lack of resources or time constraints. In order for a drug to be approved by FDA for clinical use, it must have excellent therapeutic potential in the desired area of target with minimal toxicities as supported by both pre-clinical and clinical studies. The targeted clinical evaluations fail to explore other potential therapeutic applications of the candidate drug. Drug repurposing or repositioning is a fast and relatively cheap alternative to the lengthy and expensive de novo drug discovery and development. Drug repositioning utilizes the already available clinical trials data for toxicity and adverse effects, at the same time explores the drug's therapeutic potential for a different disease. This review addresses recent developments and future scope of drug repositioning strategy.
Collapse
Affiliation(s)
- Rajesh Kumar
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Seetha Harilal
- Department of Pharmacy, Kerala University of Health Sciences, Thrissur, Kerala, India
| | - Sheeba Varghese Gupta
- Department of Pharmaceutical Sciences, College of Pharmacy, University of South Florida, Tampa, FL, 33612, USA
| | - Jobin Jose
- Department of Pharmaceutics, NGSM Institute of Pharmaceutical Science, NITTE Deemed to be University, Manglore, 575018, India
| | - Della Grace Thomas Parambi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, Al Jouf, 2014, Saudi Arabia
| | - Md Sahab Uddin
- Department of Pharmacy, Southeast University, Dhaka, Bangladesh; Pharmakon Neuroscience Research Network, Dhaka, Bangladesh
| | - Muhammad Ajmal Shah
- Department of Pharmacogonosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan
| | - Bijo Mathew
- Division of Drug Design and Medicinal Chemistry Research Lab, Department of Pharmaceutical Chemistry, Ahalia School of Pharmacy, Palakkad, 678557, Kerala, India.
| |
Collapse
|
112
|
Choi J, Lee YJ, Yoon YJ, Kim CH, Park SJ, Kim SY, Doo Kim N, Cho Han D, Kwon BM. Pimozide suppresses cancer cell migration and tumor metastasis through binding to ARPC2, a subunit of the Arp2/3 complex. Cancer Sci 2019; 110:3788-3801. [PMID: 31571309 PMCID: PMC6890432 DOI: 10.1111/cas.14205] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 09/18/2019] [Accepted: 09/26/2019] [Indexed: 12/14/2022] Open
Abstract
ARPC2 is a subunit of the Arp2/3 complex, which is essential for lamellipodia, invadopodia and filopodia, and ARPC2 has been identified as a migrastatic target molecule. To identify ARPC2 inhibitors, we generated an ARPC2 knockout DLD-1 human colon cancer cell line using the clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 (CRISPR/Cas9) system and explored gene signature-based strategies, such as a connectivity map (CMap) using the gene expression profiling data of ARPC2 knockout and knockdown cells. From the CMap-based drug discovery strategy, we identified pimozide (a clinically used antipsychotic drug) as a migrastatic drug and ARPC2 inhibitor. Pimozide inhibited the migration and invasion of various cancer cells. Through drug affinity responsive target stability (DARTS) analysis and cellular thermal shift assay (CETSA), it was confirmed that pimozide directly binds to ARPC2. Pimozide increased the lag phase of Arp2/3 complex-dependent actin polymerization and inhibited the vinculin-mediated recruitment of ARPC2 to focal adhesions in cancer cells. To validate the likely binding of pimozide to ARPC2, mutant cells, including ARPC2F225A , ARPC2F247A and ARPC2Y250F cells, were prepared using ARPC2 knockout cells prepared by gene-editing technology. Pimozide strongly inhibited the migration of mutant cells because the mutated ARPC2 likely has a larger binding pocket than the wild-type ARPC2. Therefore, pimozide is a potential ARPC2 inhibitor, and ARPC2 is a new molecular target. Taken together, the results of the present study provide new insights into the molecular mechanism and target that are responsible for the antitumor and antimetastatic activity of pimozide.
Collapse
Affiliation(s)
- Jiyeon Choi
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Yu-Jin Lee
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Yae Jin Yoon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea
| | - Cheol-Hee Kim
- Department of Bioscience and Biotechnology, Chungnam National University, Daejeon, Korea
| | - Seung-Jin Park
- Korea Research Institute of Bioscience and Biotechnology, Personalized Genomic Medicine Research Center, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| | - Seon-Young Kim
- Korea Research Institute of Bioscience and Biotechnology, Personalized Genomic Medicine Research Center, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| | - Nam Doo Kim
- Daegu-Gyeongbuk Medical Innovation Foundation, Daegu, Korea
| | - Dong Cho Han
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| | - Byoung-Mog Kwon
- Laboratory of Chemical Biology and Genomics, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Korea.,University of Science and Technology, Daejeon, Korea
| |
Collapse
|
113
|
Decreased APE-1 by Nitroxoline Enhances Therapeutic Effect in a Temozolomide-resistant Glioblastoma: Correlation with Diffusion Weighted Imaging. Sci Rep 2019; 9:16613. [PMID: 31719653 PMCID: PMC6851184 DOI: 10.1038/s41598-019-53147-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 10/23/2019] [Indexed: 12/15/2022] Open
Abstract
Glioblastoma multiforme (GBM) is one of the most aggressive human tumors with poor survival rates. The current standard treatment includes chemotherapy with temozolomide (TMZ), but acquisition of resistance is a persistent clinical problem limiting the successful treatment of GBM. The purpose of our study was to investigate therapeutic effects of nitroxoline (NTX) against TMZ-resistant GBM in vitro and in vivo in TMZ-resistant GBM-bearing mouse model, which was correlated with diffusion-weighted imaging (DWI). For in vitro study, we used TMZ-resistant GBM cell lines and evaluated therapeutic effects of NTX by clonogenic and migration assays. Quantitative RT-PCR was used to investigate the expression level of TMZ-resistant genes after NTX treatment. For in vivo study, we performed 9.4 T MR imaging to obtain T2WI for tumor volume measurement and DWI for assessment of apparent diffusion coefficient (ADC) changes by NTX in TMZ-resistant GBM mice (n = 8). Moreover, we performed regression analysis for the relationship between ADC and histological findings, which reflects the changes in cellularity and apurinic/apyrimidinic endonuclease-1 (APE-1) expression. We observed the recovery of TMZ-induced morphological changes, a reduced number of colonies and a decreased rate of migration capacity in TMZ-resistant cells after NTX treatment. The expression of APE-1 was significantly decreased in TMZ-resistant cells after NTX treatment compared with those without treatment. In an in vivo study, NTX reduced tumor growth in TMZ-resistant GBM mice (P = 0.0122). Moreover, ADC was increased in the NTX-treated TMZ-resistant GBM mice compared to the control group (P = 0.0079), which was prior to a tumor volume decrease. The cellularity and APE-1 expression by histology were negatively correlated with the ADC value, which in turn resulted in longer survival in NTX group. The decreased expression of APE-1 by NTX leads to therapeutic effects and is inversely correlated with ADC in TMZ-resistant GBM. Therefore, NTX is suggested as potential therapeutic candidate against TMZ-resistant GBM.
Collapse
|
114
|
Harnessing the therapeutic potential of anticancer drugs through amorphous solid dispersions. Biochim Biophys Acta Rev Cancer 2019; 1873:188319. [PMID: 31678141 DOI: 10.1016/j.bbcan.2019.188319] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 10/28/2019] [Accepted: 10/28/2019] [Indexed: 12/19/2022]
Abstract
The treatment of cancer is still a major challenge. But tremendous progress in anticancer drug discovery and development has occurred in the last few decades. However, this progress has resulted in few effective oncology products due to challenges associated with anticancer drug delivery. Oral administration is the most preferred route for anticancer drug delivery, but the majority of anticancer drugs currently in product pipelines and the majority of those that have been commercially approved have inherently poor water solubility, and this cannot be mitigated without compromising their potency and stability. The poor water solubility of anticancer drugs, in conjunction with other factors, leads to suboptimal pharmacokinetic performance. Thus, these drugs have limited efficacy and safety when administered orally. The amorphous solid dispersion (ASD) is a promising formulation technology that primarily enhances the aqueous solubility of poorly water-soluble drugs. In this review, we discuss the challenges associated with the oral administration of anticancer drugs and the use of ASD technology in alleviating these challenges. We emphasize the ability of ASDs to improve not only the pharmacokinetics of poorly water-soluble anticancer drugs, but also their efficacy and safety. The goal of this paper is to rationalize the application of ASD technology in the formulation of anticancer drugs, thereby creating superior oncology products that lead to improved therapeutic outcomes.
Collapse
|
115
|
Lee JS, Roberts A, Juarez D, Vo TTT, Bhatt S, Herzog LO, Mallya S, Bellin RJ, Agarwal SK, Salem AH, Xu T, Jia J, Li L, Hanna JR, Davids MS, Fleischman AG, O'Brien S, Lam LT, Leverson JD, Letai A, Schatz JH, Fruman DA. Statins enhance efficacy of venetoclax in blood cancers. Sci Transl Med 2019; 10:10/445/eaaq1240. [PMID: 29899021 DOI: 10.1126/scitranslmed.aaq1240] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 05/11/2018] [Indexed: 12/14/2022]
Abstract
Statins have shown promise as anticancer agents in experimental and epidemiologic research. However, any benefit that they provide is likely context-dependent, for example, applicable only to certain cancers or in combination with specific anticancer drugs. We report that inhibition of 3-hydroxy-3-methylglutaryl coenzyme A reductase (HMGCR) using statins enhances the proapoptotic activity of the B cell lymphoma-2 (BCL2) inhibitor venetoclax (ABT-199) in primary leukemia and lymphoma cells but not in normal human peripheral blood mononuclear cells. By blocking mevalonate production, HMGCR inhibition suppressed protein geranylgeranylation, resulting in up-regulation of proapoptotic protein p53 up-regulated modulator of apoptosis (PUMA). In support of these findings, dynamic BH3 profiling confirmed that statins primed cells for apoptosis. Furthermore, in retrospective analyses of three clinical studies of chronic lymphocytic leukemia, background statin use was associated with enhanced response to venetoclax, as demonstrated by more frequent complete responses. Together, this work provides mechanistic justification and clinical evidence to warrant prospective clinical investigation of this combination in hematologic malignancies.
Collapse
Affiliation(s)
- J Scott Lee
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Andrew Roberts
- Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA.
| | - Dennis Juarez
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Thanh-Trang T Vo
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Shruti Bhatt
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Lee-Or Herzog
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Sharmila Mallya
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | | | | | - Ahmed Hamed Salem
- Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA.,Department of Clinical Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Tu Xu
- Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA
| | - Jia Jia
- Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA
| | - Lingxiao Li
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - John R Hanna
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Matthew S Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Angela G Fleischman
- Department of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Susan O'Brien
- Department of Medicine, University of California, Irvine, Irvine, CA 92697, USA
| | - Lloyd T Lam
- Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA
| | - Joel D Leverson
- Oncology Development, AbbVie Inc., North Chicago, IL 60064, USA
| | - Anthony Letai
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jonathan H Schatz
- Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
116
|
Turanli B, Altay O, Borén J, Turkez H, Nielsen J, Uhlen M, Arga KY, Mardinoglu A. Systems biology based drug repositioning for development of cancer therapy. Semin Cancer Biol 2019; 68:47-58. [PMID: 31568815 DOI: 10.1016/j.semcancer.2019.09.020] [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: 07/22/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 01/20/2023]
Abstract
Drug repositioning is a powerful method that can assists the conventional drug discovery process by using existing drugs for treatment of a disease rather than its original indication. The first examples of repurposed drugs were discovered serendipitously, however data accumulated by high-throughput screenings and advancements in computational biology methods have paved the way for rational drug repositioning methods. As chemotherapeutic agents have notorious side effects that significantly reduce quality of life, drug repositioning promises repurposed noncancer drugs with little or tolerable adverse effects for cancer patients. Here, we review current drug-related data types and databases including some examples of web-based drug repositioning tools. Next, we describe systems biology approaches to be used in drug repositioning for effective cancer therapy. Finally, we highlight examples of mostly repurposed drugs for cancer treatment and provide an overview of future expectations in the field for development of effective treatment strategies.
Collapse
Affiliation(s)
- Beste Turanli
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Department of Bioengineering, Marmara University, Istanbul, Turkey; Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Ozlem Altay
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | - Jan Borén
- Department of Molecular and Clinical Medicine, University of Gothenburg and Sahlgrenska University Hospital Gothenburg, Sweden
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum 25240, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden
| | | | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm SE-17121, Sweden; Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London SE1 9RT, United Kingdom.
| |
Collapse
|
117
|
Drug repurposing for breast cancer therapy: Old weapon for new battle. Semin Cancer Biol 2019; 68:8-20. [PMID: 31550502 PMCID: PMC7128772 DOI: 10.1016/j.semcancer.2019.09.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022]
Abstract
Despite tremendous resources being invested in prevention and treatment, breast cancer remains a leading cause of cancer deaths in women globally. The available treatment modalities are very costly and produces severe side effects. Drug repurposing that relate to new uses for old drugs has emerged as a novel approach for drug development. Repositioning of old, clinically approved, off patent non-cancer drugs with known targets, into newer indication is like using old weapons for new battle. The advances in genomics, proteomics and information computational biology has facilitated the process of drug repurposing. Repositioning approach not only fastens the process of drug development but also offers more effective, cheaper, safer drugs with lesser/known side effects. During the last decade, drugs such as alkylating agents, anthracyclins, antimetabolite, CDK4/6 inhibitor, aromatase inhibitor, mTOR inhibitor and mitotic inhibitors has been repositioned for breast cancer treatment. The repositioned drugs have been successfully used for the treatment of most aggressive triple negative breast cancer. The literature review suggest that serendipity plays a major role in the drug development. This article describes the comprehensive overview of the current scenario of drug repurposing for the breast cancer treatment. The strategies as well as several examples of repurposed drugs are provided. The challenges associated with drug repurposing are discussed.
Collapse
|
118
|
Wang X, Wang J, Pan J, Zhao F, Kan D, Cheng R, Zhang X, Sun SK. Rhenium Sulfide Nanoparticles as a Biosafe Spectral CT Contrast Agent for Gastrointestinal Tract Imaging and Tumor Theranostics in Vivo. ACS APPLIED MATERIALS & INTERFACES 2019; 11:33650-33658. [PMID: 31448891 DOI: 10.1021/acsami.9b10479] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Spectral computed tomography (CT) imaging as a novel imaging technique shows promising prospects in the accurate diagnosis of various diseases. However, clinically iodinated contrast agents suffer from poor signal-to-noise ratio, and emerging heavy-metal-based CT contrast agents arouse great biosafety concern. Herein, we show the fabrication of rhenium sulfide (ReS2) nanoparticles, a clinic radiotherapy sensitizer, as a biosafe spectral CT contrast agent for the gastrointestinal tract imaging and tumor theranostics in vivo by teaching old drugs new tricks. The ReS2 nanoparticles were fabricated in a one-pot facile method at room temperature, and exhibited sub-10 nm size, favorable monodispersity, admirable aqueous solubility, and strong X-ray attenuation capability. More importantly, the proposed nanoparticles possess an outstanding spectral CT imaging ability and undoubted biosafety as a clinic therapeutic agent. Besides, the ReS2 nanoparticles possess appealing photothermal performance due to their intense near-infrared absorption. The proposed nano-agent not only guarantees obvious contrast enhancement in gastrointestinal tract spectral CT imaging in vivo, but also allows effective CT imaging-guided tumor photothermal therapy. The proposed "teaching old drugs new tricks" strategy shortens the time and cuts the cost required for clinical application of nano-agents based on existing clinical toxicology testing and trial results, and lays down a low-cost, time-saving, and energy-saving method for the development of multifunctional nano-agents toward clinical applications.
Collapse
Affiliation(s)
| | - Jiaojiao Wang
- School of Medical Imaging , Tianjin Medical University , Tianjin 300203 , China
| | - Jinbin Pan
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging , Tianjin Medical University General Hospital , Tianjin 300052 , China
| | - Fangshi Zhao
- Department of Radiology, Tianjin Key Laboratory of Functional Imaging , Tianjin Medical University General Hospital , Tianjin 300052 , China
| | - Di Kan
- School of Medical Imaging , Tianjin Medical University , Tianjin 300203 , China
| | - Ran Cheng
- School of Medical Imaging , Tianjin Medical University , Tianjin 300203 , China
| | | | - Shao-Kai Sun
- School of Medical Imaging , Tianjin Medical University , Tianjin 300203 , China
| |
Collapse
|
119
|
Xia Y, Jia C, Xue Q, Jiang J, Xie Y, Wang R, Ran Z, Xu F, Zhang Y, Ye T. Antipsychotic Drug Trifluoperazine Suppresses Colorectal Cancer by Inducing G0/G1 Arrest and Apoptosis. Front Pharmacol 2019; 10:1029. [PMID: 31572198 PMCID: PMC6753363 DOI: 10.3389/fphar.2019.01029] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 08/12/2019] [Indexed: 02/05/2023] Open
Abstract
Repurposing existing drugs for cancer treatment is an effective strategy. An approved antipsychotic drug, trifluoperazine (TFP), has been reported to have potential anticancer effects against several cancer types. Here, we investigated the effect and molecular mechanism of TFP in colorectal cancer (CRC). In vitro studies showed that TFP induced G0/G1 cell cycle arrest to dramatically inhibit CRC cell proliferation through downregulating cyclin-dependent kinase (CDK) 2, CDK4, cyclin D1, and cyclin E and upregulating p27. TFP also induced apoptosis, decreased mitochondrial membrane potential, and increased reactive oxygen species levels in CRC cells, indicating that TFP induced mitochondria-mediated intrinsic apoptosis. Importantly, TFP significantly suppressed tumor growth in two CRC subcutaneous tumor models without side effects. Interestingly, TFP treatment increased the expression levels of programmed death-1 ligand 1 (PD-L1) in CRC cells and programmed death-1 (PD-1) in tumor-infiltrating CD4+ and CD8+ T cells, implying that the combination of TFP with an immune checkpoint inhibitor, such as an anti-PD-L1 or anti-PD-1 antibody, might have synergistic anticancer effects. Taken together, our study signifies that TFP is a novel treatment strategy for CRC and indicates the potential for using the combination treatment of TFP and immune checkpoint blockade to increase antitumor efficiency.
Collapse
Affiliation(s)
- Yong Xia
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Chengsen Jia
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China.,Key Laboratory of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Qiang Xue
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Jinrui Jiang
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yao Xie
- Department of Gynecology and Obstetrics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Ranran Wang
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Zhiqiang Ran
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Fuyan Xu
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yiwen Zhang
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Tinghong Ye
- Department of Rehabilitation Medicine and Laboratory of Liver Surgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| |
Collapse
|
120
|
Delou JMA, Souza ASO, Souza LCM, Borges HL. Highlights in Resistance Mechanism Pathways for Combination Therapy. Cells 2019; 8:E1013. [PMID: 31480389 PMCID: PMC6770082 DOI: 10.3390/cells8091013] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/15/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022] Open
Abstract
Combination chemotherapy has been a mainstay in cancer treatment for the last 60 years. Although the mechanisms of action and signaling pathways affected by most treatments with single antineoplastic agents might be relatively well understood, most combinations remain poorly understood. This review presents the most common alterations of signaling pathways in response to cytotoxic and targeted anticancer drug treatments, with a discussion of how the knowledge of signaling pathways might support and orient the development of innovative strategies for anticancer combination therapy. The ultimate goal is to highlight possible strategies of chemotherapy combinations based on the signaling pathways associated with the resistance mechanisms against anticancer drugs to maximize the selective induction of cancer cell death. We consider this review an extensive compilation of updated known information on chemotherapy resistance mechanisms to promote new combination therapies to be to discussed and tested.
Collapse
Affiliation(s)
- João M A Delou
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Alana S O Souza
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Leonel C M Souza
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil
| | - Helena L Borges
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil.
| |
Collapse
|
121
|
Nicolini A, Ferrari P, Carpi A. Limiting research on molecular targeted therapies in advanced solid cancers: beyond a cost–effectiveness ratio analysis. Biomark Med 2019; 13:887-890. [DOI: 10.2217/bmm-2019-0216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Andrea Nicolini
- Department of Oncology, Transplantations & New Technologies in Medicine, University of Pisa, Italy
| | - Paola Ferrari
- Department of Oncology, Transplantations & New Technologies in Medicine, University of Pisa, Italy
| | - Angelo Carpi
- Department of Clinical & Experimental Medicine, University of Pisa, Italy
| |
Collapse
|
122
|
Eslam M, George J. Genetic Insights for Drug Development in NAFLD. Trends Pharmacol Sci 2019; 40:506-516. [PMID: 31160124 DOI: 10.1016/j.tips.2019.05.002] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/10/2019] [Accepted: 05/06/2019] [Indexed: 12/21/2022]
Abstract
Drug development is a costly, time-consuming, and challenging endeavour, with only a few agents reaching the threshold of approval for clinical use. Therefore, approaches to more efficiently identify targets that are likely to translate to clinical benefit are required. Interrogation of the human genome in large patient cohorts has rapidly advanced our knowledge of the genetic architecture and underlying mechanisms of many diseases, including nonalcoholic fatty liver disease (NAFLD). There are no approved pharmacotherapies for NAFLD currently. Genetic insights provide a powerful and new approach to infer and prioritise candidate drugs, with such selection avoiding myriad pitfalls, while defining likely benefits. In this review, we discuss the prospects and challenges for the optimal utilisation of genetic findings for improving and accelerating the NAFLD drug discovery pipeline.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia.
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Westmead, NSW, Australia.
| |
Collapse
|
123
|
Halliday M, Mallucci GR. Reply: Trazodone to change the risk of neurodegeneration: bedside to bench. Brain 2019. [PMID: 28641370 DOI: 10.1093/brain/awx150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Mark Halliday
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK
| | - Giovanna R Mallucci
- MRC Toxicology Unit, Hodgkin Building, Lancaster Road, Leicester LE1 9HN, UK.,Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0AH, UK
| |
Collapse
|
124
|
Zhang L, Song Q, Zhang X, Li L, Xu X, Xu X, Li X, Wang Z, Lin Y, Li X, Li M, Su F, Wang X, Qiu P, Guan H, Tang Y, Xu W, Yang J, Zhao C. Zelnorm, an agonist of 5-Hydroxytryptamine 4-receptor, acts as a potential antitumor drug by targeting JAK/STAT3 signaling. Invest New Drugs 2019; 38:311-320. [PMID: 31087223 DOI: 10.1007/s10637-019-00790-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 05/06/2019] [Indexed: 12/14/2022]
Abstract
The Janus kinase (JAK)/signal transducer and activator of transcription 3 (STAT3) signaling pathway plays central roles in cancer cell growth and survival. Drug repurposing strategies have provided a valuable approach for developing antitumor drugs. Zelnorm (tegaserod maleate) was originally designed as an agonist of 5-hydroxytryptamine 4 receptor (5-HT4R) and approved by the FDA for treating irritable bowel syndrome with constipation (IBS-C). Through the use of a high-throughput drug screening system, Zelnorm was identified as a JAK/STAT3 signaling inhibitor. Moreover, the inhibition of STAT3 phosphorylation by Zelnorm was independent of its original target 5-HT4R. Zelnorm could cause G1 cell cycle arrest, induce cell apoptosis and inhibit the growth of a variety of cancer cells. The present study identifies Zelnorm as a novel JAK/STAT3 signaling inhibitor and reveals a new clinical application of Zelnorm upon market reintroduction.
Collapse
Affiliation(s)
- Lei Zhang
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Qiaoling Song
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Xinxin Zhang
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Li Li
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Ximing Xu
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Xiaohan Xu
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
| | - Xiaoyu Li
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Zhuoya Wang
- School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Yuxi Lin
- School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Xin Li
- School of Life Science, Lanzhou University, Lanzhou, 730000, Gansu, China
| | - Mengyuan Li
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Fan Su
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
| | - Xin Wang
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Peiju Qiu
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Huashi Guan
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Yu Tang
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Wenfang Xu
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Jinbo Yang
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China
| | - Chenyang Zhao
- School of Medicine and Pharmacy, Ocean University of China, 23 East Hong Kong Road, Qingdao, 266071, Shandong, China.
- Innovation Platform of Marine Drug Screening & Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266100, Shandong, China.
| |
Collapse
|
125
|
Formulation and evaluation of cyclodextrin complexes for improved anticancer activity of repurposed drug: Niclosamide. Carbohydr Polym 2019; 212:252-259. [DOI: 10.1016/j.carbpol.2019.02.041] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/28/2019] [Accepted: 02/12/2019] [Indexed: 12/14/2022]
|
126
|
Mishra S, Verma SS, Rai V, Awasthee N, Arya JS, Maiti KK, Gupta SC. Curcuma raktakanda Induces Apoptosis and Suppresses Migration in Cancer Cells: Role of Reactive Oxygen Species. Biomolecules 2019; 9:biom9040159. [PMID: 31018580 PMCID: PMC6523773 DOI: 10.3390/biom9040159] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/08/2019] [Accepted: 04/12/2019] [Indexed: 12/18/2022] Open
Abstract
Although over 100 species of Curcuma are reported, only Curcuma longa is extensively studied. Curcuma raktakanda, a poorly studied species, is most commonly distributed in the Kerala state of India. For the first time, we examined the efficacy of different fractions (acetone, hexane, and ethyl acetate) of C. raktakanda against glioma, cervical, and breast cancer cell lines. As determined by mitochondrial reductase activity assay, the viability of cancer cells was decreased in a concentration-dependent manner by the three fractions. The half maximal inhibitory concentration (IC-50) values after the treatment of C-6 glioma cells for 48 h was found to be 32.97 µg/mL (acetone extract), 40.63 µg/mL (hexane extract), and 51.65 µg/mL (ethyl acetate extract). Of the three fractions, the acetone fraction was more effective. The long-term colony formation of cancer cells was significantly suppressed by the acetone fraction. Analyses using DAPI (4',6-diamidino-2-phenylindole) staining, AO/PI (acridine orange/propidium iodide) staining, DNA laddering, and sub-G1 population revealed that the acetone extract induced apoptosis in glioma cells. The extract induced reactive oxygen species generation and suppressed the expression of cell survival proteins. The migration of cancer cells was also suppressed by the acetone extract. The gas chromatography-mass spectrometry (GC-MS) analysis indicated that tetracontane, dotriacontane, hexatriacontane, pentacosane, hexacosane, and eicosane are the major components in the acetone extract. Collectively, the extract from C. raktakanda exhibited anti-carcinogenic activities in cancer cells. We are exploring whether the phytoconstituents, individually, or collectively contribute to the anti-cancer activities of C. raktakanda.
Collapse
Affiliation(s)
- Shruti Mishra
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Sumit Singh Verma
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Vipin Rai
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Nikee Awasthee
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| | - Jayadev S Arya
- CSIR-National Institute for Interdisciplinary Science and Technology, Chemical Science and Technology Division, Organic Chemistry Section, Trivandrum-695019, India.
| | - Kaustabh K Maiti
- CSIR-National Institute for Interdisciplinary Science and Technology, Chemical Science and Technology Division, Organic Chemistry Section, Trivandrum-695019, India.
| | - Subash C Gupta
- Laboratory for Translational Cancer Research, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi-221 005, India.
| |
Collapse
|
127
|
Abdelaleem M, Ezzat H, Osama M, Megahed A, Alaa W, Gaber A, Shafei A, Refaat A. Prospects for repurposing CNS drugs for cancer treatment. Oncol Rev 2019; 13:411. [PMID: 31044029 PMCID: PMC6478007 DOI: 10.4081/oncol.2019.411] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Accepted: 04/09/2019] [Indexed: 02/08/2023] Open
Abstract
Drug repurposing is the idea of using an already approved drug for another disease or disorder away from its initial use. This new approach ensures the reduction in high cost required for developing a new drug in addition to the time consumed, especially in the tumor disorders that show an unceasing rising rate with an unmet success rate of new anticancer drugs. In our review, we will review the anti-cancer effect of some CNS drugs, including both therapeutic and preventive, by searching the literature for preclinical or clinical evidence for anticancer potential of central nervous system drugs over the last 8 years period (2010-2018) and including only evidence from Q1 journals as indicated by Scimago website (www.scimagojr.com). We concluded that Some Central Nervous system drugs show a great potential as anti-cancer in vitro, in vivo and clinical trials through different mechanisms and pathways in different types of cancer that reveal a promising evidence for the repurposing of CNS drugs for new indications.
Collapse
Affiliation(s)
| | - Hossam Ezzat
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | | | - Adel Megahed
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Waleed Alaa
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Ahmed Gaber
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Ayman Shafei
- Armed Forces College of Medicine (AFCM), Cairo, Egypt
| | - Alaa Refaat
- Armed Forces College of Medicine (AFCM), Cairo, Egypt.,Research Center, Misr International University (MIU), Cairo, Egypt.,Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
128
|
Pocket similarity identifies selective estrogen receptor modulators as microtubule modulators at the taxane site. Nat Commun 2019; 10:1033. [PMID: 30833575 PMCID: PMC6399299 DOI: 10.1038/s41467-019-08965-w] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 01/19/2019] [Indexed: 02/01/2023] Open
Abstract
Taxanes are a family of natural products with a broad spectrum of anticancer activity. This activity is mediated by interaction with the taxane site of beta-tubulin, leading to microtubule stabilization and cell death. Although widely used in the treatment of breast cancer and other malignancies, existing taxane-based therapies including paclitaxel and the second-generation docetaxel are currently limited by severe adverse effects and dose-limiting toxicity. To discover taxane site modulators, we employ a computational binding site similarity screen of > 14,000 drug-like pockets from PDB, revealing an unexpected similarity between the estrogen receptor and the beta-tubulin taxane binding pocket. Evaluation of nine selective estrogen receptor modulators (SERMs) via cellular and biochemical assays confirms taxane site interaction, microtubule stabilization, and cell proliferation inhibition. Our study demonstrates that SERMs can modulate microtubule assembly and raises the possibility of an estrogen receptor-independent mechanism for inhibiting cell proliferation. Taxanes are natural products which bind beta-tubulin, stabilize microtubules and have a broad spectrum of anticancer activity. Here authors employ a computational binding site similarity screen and cell-based assays to reveal a SERM cross-reactivity between the estrogen receptor and the beta-tubulin taxane binding pocket.
Collapse
|
129
|
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
|
130
|
Sourbron J, Partoens M, Scheldeman C, Zhang Y, Lagae L, de Witte P. Drug repurposing for Dravet syndrome in scn1Lab -/- mutant zebrafish. Epilepsia 2019; 60:e8-e13. [PMID: 30663052 PMCID: PMC6850687 DOI: 10.1111/epi.14647] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 12/22/2018] [Accepted: 12/24/2018] [Indexed: 11/28/2022]
Abstract
Dravet syndrome (DS) is a severe genetic epileptic encephalopathy with onset during the first year of life. Zebrafish models recapitulating human diseases are often used as drug discovery platforms, but also for drug repurposing testing. It was recently shown that pharmacological modulation of three serotonergic (5-HT) receptors (5-HT1D , 5-HT2C , 5-HT2A ) exerts antiseizure effects in a zebrafish scn1Lab-/- mutant model of DS. Using the zebrafish DS model, our aim was to examine the possibility of repurposing efavirenz (EFA), lisuride (LIS), and rizatriptan (RIZA), marketed medicines with a 5-HT on- or off-target profile, as antiepileptic drugs for DS. To examine whether these compounds have a broader antiseizure profile, they were tested in pentylenetetrazol and ethyl ketopentenoate (EKP) zebrafish models. Pharmacological effects were assessed by locomotor behavior, local field potential brain recordings, and bioluminescence. EFA was active in all models, whereas LIS was selectively active in the zebrafish DS model. Mainly, a poor response was observed to RIZA. Taken together, our preclinical results show that LIS could be a potential candidate for DS treatment. EFA was also active in the EKP model, characterized by a high level of treatment resistance, and hence these data are potentially important for future treatment of drug-resistant epilepsy.
Collapse
Affiliation(s)
- Jo Sourbron
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Michèle Partoens
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Chloë Scheldeman
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium.,Neurogenetics Research Group, VUB (Vrije Universiteit Brussel), Brussels, Belgium
| | - Yifan Zhang
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Lieven Lagae
- Department of Development and Regeneration, Section Pediatric Neurology, University of Leuven University Hospital, Leuven, Belgium
| | - Peter de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
131
|
Clinical efficacy and mechanism of Pralatrexate combined with Palbociclib Isethionate in treatment of bladder cancer patients. Oncol Lett 2019; 17:201-208. [PMID: 30655756 PMCID: PMC6313094 DOI: 10.3892/ol.2018.9617] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/27/2018] [Indexed: 02/03/2023] Open
Abstract
The clinical efficacy and mechanism of Pralatrexate (PTX) combined with Palbociclib Isethionate (PAL) in the treatment of bladder cancer patients was investigated. A retrospective analysis of medical records of 82 bladder cancer patients admitted to Shengjing Hospital of China Medical University from February 2015 to February 2018 was performed. Patients treated with PTX combined with PAL served as study group (42 cases) and patients with conventional GC (gemcitabine plus cisplatin) chemotherapy regimen were the control group (40 cases). Changes in liver function indexes before and after treatment were observed, including alanine aminotransferase (ALT), aspartate aminotransferase (AST), alkaline phosphatase (ALP), and total bilirubin (TBil). RT-qPCR was used for detection of relative expression levels of serum dihydrofolate reductase (DHFR) and vascular endothelial growth factor (VEGF) before and after treatment in the two groups. The clinical efficacy after treatment and adverse reactions during treatment were observed in the two groups. There was no significant difference in the clinical remission rate (RR) nor in the serum ALT, AST, ALP and TBil levels between the study and the control groups (P>0.05). Concentrations of serum ALT, AST, ALP and TBil were significantly higher than those before treatment in both groups (P<0.05). Serum ALT, AST, ALP and TBil concentrations in study group were significantly lower than those in control group (P<0.05). There was no significant difference in the incidence of thrombocytopenia and leukopenia between the two groups (P>0.05). There was no significant difference in relative expression levels of serum DHFR mRNA and VEGF mRNA before treatment between the study and control groups (P>0.05). Those after treatment were significantly lower than those before treatment in both groups (P<0.05), and those after treatment in study group were significantly lower than those in control group (P<0.05). PTX combined with PAL can reduce adverse reactions of nausea and vomiting and liver function impairment during treatment and suppress tumor neovascularization. This is achieved possibly by inhibiting expression levels of DHFR and VEGF, thereby killing cancer cells. PTX combined with PAL may become a new method for the treatment of bladder cancer patients. DHFR and VEGF are expected to become novel therapeutic targets for the treatment of bladder cancer.
Collapse
|
132
|
Sohraby F, Bagheri M, Aryapour H. Performing an In Silico Repurposing of Existing Drugs by Combining Virtual Screening and Molecular Dynamics Simulation. Methods Mol Biol 2019; 1903:23-43. [PMID: 30547434 DOI: 10.1007/978-1-4939-8955-3_2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Drug repurposing has become one of the most widely used methods that can make drug discovery more efficient and less expensive. Additionally, computational methods such as structure-based drug designing can be utilized to make drug discovery more efficient and more accurate. Now imagine what can be achieved by combining drug repurposing and computational methods together in drug discovery, "in silico repurposing." In this chapter, we tried to describe a method that combines structure-based virtual screening and molecular dynamics simulation which can find effective compounds among existing drugs that may affect on a specific molecular target. By using molecular docking as a tool for the screening process and then by calculating ligand binding in an active receptor site using scoring functions and inspecting the proper orientation of pharmacophores in the binding site, the potential compounds will be chosen. After that, in order to test the potential compounds in a realistic environment, molecular dynamics simulation and related analysis have to be carried out for separating the false positives and the true positives from each other and finally identifying true "Hit" compounds. It's good to emphasize that if any of these identified potential compounds turn out to have the efficacy to affect that specific molecular target, it can be taken to the phase 2 clinical trials straightaway.
Collapse
Affiliation(s)
- Farzin Sohraby
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| | - Milad Bagheri
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran
| | - Hassan Aryapour
- Department of Biology, Faculty of Science, Golestan University, Gorgan, Iran.
| |
Collapse
|
133
|
Wang X, Wu X, Zhang Z, Ma C, Wu T, Tang S, Zeng Z, Huang S, Gong C, Yuan C, Zhang L, Feng Y, Huang B, Liu W, Zhang B, Shen Y, Luo W, Wang X, Liu B, Lei Y, Ye Z, Zhao L, Cao D, Yang L, Chen X, Haydon RC, Luu HH, Peng B, Liu X, He TC. Monensin inhibits cell proliferation and tumor growth of chemo-resistant pancreatic cancer cells by targeting the EGFR signaling pathway. Sci Rep 2018; 8:17914. [PMID: 30559409 PMCID: PMC6297164 DOI: 10.1038/s41598-018-36214-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 11/14/2018] [Indexed: 02/05/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most deadly malignancies with <5% five-year survival rate due to late diagnosis, limited treatment options and chemoresistance. There is thus an urgent unmet clinical need to develop effective anticancer drugs to treat pancreatic cancer. Here, we study the potential of repurposing monensin as an anticancer drug for chemo-resistant pancreatic cancer. Using the two commonly-used chemo-resistant pancreatic cancer cell lines PANC-1 and MiaPaCa-2, we show that monensin suppresses cell proliferation and migration, and cell cycle progression, while solicits apoptosis in pancreatic cancer lines at a low micromole range. Moreover, monensin functions synergistically with gemcitabine or EGFR inhibitor erlotinib in suppressing cell growth and inducing cell death of pancreatic cancer cells. Mechanistically, monensin suppresses numerous cancer-associated pathways, such as E2F/DP1, STAT1/2, NFkB, AP-1, Elk-1/SRF, and represses EGFR expression in pancreatic cancer lines. Furthermore, the in vivo study shows that monensin blunts PDAC xenograft tumor growth by suppressing cell proliferation via targeting EGFR pathway. Therefore, our findings demonstrate that monensin can be repurposed as an effective anti-pancreatic cancer drug even though more investigations are needed to validate its safety and anticancer efficacy in pre-clinical and clinical models.
Collapse
Affiliation(s)
- Xin Wang
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, 610041, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhonglin Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Hepatobiliary & Pancreatic Surgery, Neurosurgery, and Otolaryngology, Head and Neck Surgery, the Affiliated Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chao Ma
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Hepatobiliary & Pancreatic Surgery, Neurosurgery, and Otolaryngology, Head and Neck Surgery, the Affiliated Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Tingting Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Hepatobiliary & Pancreatic Surgery, Neurosurgery, and Otolaryngology, Head and Neck Surgery, the Affiliated Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Shengli Tang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Hepatobiliary & Pancreatic Surgery, Neurosurgery, and Otolaryngology, Head and Neck Surgery, the Affiliated Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Zongyue Zeng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Shifeng Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Cheng Gong
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Hepatobiliary & Pancreatic Surgery, Neurosurgery, and Otolaryngology, Head and Neck Surgery, the Affiliated Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chengfu Yuan
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Biochemistry and Molecular Biology, China Three Gorges University School of Medicine, Yichang, 443002, China
| | - Linghuan Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Yixiao Feng
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bo Huang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
- Department of Clinical Laboratory Medicine, the Second Affiliated Hospital of Nanchang University, Nanchang, 330031, China
| | - Wei Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Bo Zhang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province, and the Departments of Orthopaedic Surgery and Obstetrics and Gynecology, the First and Second Hospitals of Lanzhou University, Lanzhou, 730030, China
| | - Yi Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, Xiangya Second Hospital of Central South University, Changsha, 410011, China
| | - Wenping Luo
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Chongqing Key Laboratory for Oral Diseases and Biomedical Sciences, and the Affiliated Hospital of Stomatology of Chongqing Medical University, Chongqing, China
| | - Xi Wang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Bo Liu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yan Lei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhenyu Ye
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of General Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, 215004, China
| | - Ling Zhao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Departments of Surgery, Clinical Laboratory Medicine, Orthopaedic Surgery, Plastic Surgery and Burn, Otolaryngology, Head and Neck Surgery, and Obstetrics and Gynecology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Daigui Cao
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Ministry of Education Key Laboratory of Diagnostic Medicine and School of Laboratory Medicine, and the Affiliated Hospitals of Chongqing Medical University, Chongqing, 400016, China
| | - Lijuan Yang
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Key Laboratory of Orthopaedic Surgery of Gansu Province, and the Departments of Orthopaedic Surgery and Obstetrics and Gynecology, the First and Second Hospitals of Lanzhou University, Lanzhou, 730030, China
| | - Xian Chen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Clinical Laboratory Medicine, the Affiliated Hospital of Qingdao University, Qingdao, 266061, China
| | - Rex C Haydon
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Hue H Luu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Bing Peng
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xubao Liu
- Department of Pancreatic Surgery, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA.
| |
Collapse
|
134
|
Seliger C, Schaertl J, Gerken M, Luber C, Proescholdt M, Riemenschneider MJ, Leitzmann MF, Hau P, Klinkhammer-Schalke M. Use of statins or NSAIDs and survival of patients with high-grade glioma. PLoS One 2018; 13:e0207858. [PMID: 30507932 PMCID: PMC6277074 DOI: 10.1371/journal.pone.0207858] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 11/07/2018] [Indexed: 12/21/2022] Open
Abstract
Background High-grade glioma (HGG) is associated with a limited prognosis. Drug repurposing has become of increasing interest to improve standard therapy. Statins and NSAIDs inhibit glioma cell growth in vitro and in vivo, but data on statin and NSAID treatment in relation to survival of patients with HGG are sparse. Methods We performed multivariable adjusted Cox-regression analyses among 1,093 patients with HGG from a regional cancer registry to obtain Hazard Ratios (HRs) with 95% Confidence Intervals (CIs) for overall survival (OS) and progression-free survival (PFS) according to treatment with statins or NSAIDs. Data on dose and duration of treatment was mostly lacking in our analysis, therefore we were not able to perform dose-response analyses. Results Use of statins was unrelated to OS or PFS of glioma patients. Use of aspirin was associated with prolonged OS and PFS in patients with WHO grade III, but not WHO grade IV glioma. Use of other NSAIDs (diclofenac, ibuprofen) or non-NSAID analgesics (paracetamol) was mostly unrelated to survival of glioma patients. Use of selective COX-2 inhibitors and metamizol was related to inferior patient survival in parts of the analyses. Conclusions Use of statins or NSAIDS, including aspirin, was not associated with prolonged OS or PFS of patients with WHO grade IV glioma in our selected cohort. There was an indication for improved survival in patients with WHO grade III glioma using aspirin, but further studies are needed to confirm our first observation.
Collapse
Affiliation(s)
- Corinna Seliger
- Department of Neurology, Regensburg University Medical Center, Regensburg, Germany
- Wilhelm Sander-NeuroOncology Unit, Regensburg University Medical Center, Regensburg, Germany
- * E-mail:
| | - Julia Schaertl
- Department of Neurology, Regensburg University Medical Center, Regensburg, Germany
| | - Michael Gerken
- Tumor Center, Institute for Quality Assurance and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Christian Luber
- Department of Neurology, Regensburg University Medical Center, Regensburg, Germany
| | - Martin Proescholdt
- Wilhelm Sander-NeuroOncology Unit, Regensburg University Medical Center, Regensburg, Germany
- Department of Neurosurgery, Regensburg University Medical Center, Regensburg, Germany
| | - Markus J. Riemenschneider
- Wilhelm Sander-NeuroOncology Unit, Regensburg University Medical Center, Regensburg, Germany
- Department of Neuropathology, Regensburg University Hospital, Regensburg, Germany
| | - Michael F. Leitzmann
- Department of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Peter Hau
- Department of Neurology, Regensburg University Medical Center, Regensburg, Germany
- Wilhelm Sander-NeuroOncology Unit, Regensburg University Medical Center, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Tumor Center, Institute for Quality Assurance and Health Services Research, University of Regensburg, Regensburg, Germany
| |
Collapse
|
135
|
Synthesis, antimalarial, antiproliferative, and apoptotic activities of benzimidazole-5-carboxamide derivatives. Med Chem Res 2018. [DOI: 10.1007/s00044-018-2258-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
136
|
Jang WJ, Jung SK, Vo TTL, Jeong CH. Anticancer activity of paroxetine in human colon cancer cells: Involvement of MET and ERBB3. J Cell Mol Med 2018; 23:1106-1115. [PMID: 30421568 PMCID: PMC6349215 DOI: 10.1111/jcmm.14011] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 07/18/2018] [Accepted: 08/16/2018] [Indexed: 12/28/2022] Open
Abstract
The concept of drug repositioning has recently received considerable attention in the field of oncology. In the present study, we propose that paroxetine can be used as a potent anticancer drug. Paroxetine, one of the selective serotonin reuptake inhibitors (SSRIs), has been widely prescribed for the treatment of depression and anxiety disorders. Recently, SSRIs have been reported to have anticancer activity in various types of cancer cells; however, the underlying mechanisms of their action are not yet known. In this study, we investigated the potential anticancer effect of paroxetine in human colorectal cancer cells, HCT116 and HT‐29. Treatment with paroxetine reduced cell viability, which was associated with marked increase in apoptosis, in both the cell lines. Also, paroxetine effectively inhibited colony formation and 3D spheroid formation. We speculated that the mode of action of paroxetine might be through the inhibition of two major receptor tyrosine kinases – MET and ERBB3 – leading to the suppression of AKT, ERK and p38 activation and induction of JNK and caspase‐3 pathways. Moreover, in vivo experiments revealed that treatment of athymic nude mice bearing HT‐29 cells with paroxetine remarkably suppressed tumour growth. In conclusion, paroxetine is a potential therapeutic option for patients with colorectal cancer.
Collapse
Affiliation(s)
- Won-Jun Jang
- College of Pharmacy, Keimyung University, Daegu, Korea
| | - Sung Keun Jung
- School of Food Science and Biotechnology, Kyungpook National University, Daegu, Korea
| | | | - Chul-Ho Jeong
- College of Pharmacy, Keimyung University, Daegu, Korea
| |
Collapse
|
137
|
Yeo DC, Wiraja C, Miao Q, Ning X, Pu K, Xu C. Anti-Scarring Drug Screening with Near-Infrared Molecular Probes Targeting Fibroblast Activation Protein-α. ACS APPLIED BIO MATERIALS 2018; 1:2054-2061. [DOI: 10.1021/acsabm.8b00528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- David C. Yeo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Qingqing Miao
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Xiaoyu Ning
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
- NTU Institute for Health Technologies, Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Drive, Singapore 637553, Singapore
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| |
Collapse
|
138
|
Li J, Liu R, Sun Z, Tang S, Wang L, Liu C, Zhao W, Yao Y, Sun C. The association between statin use and endometrial cancer survival outcome: A meta-analysis. Medicine (Baltimore) 2018; 97:e13264. [PMID: 30461633 PMCID: PMC6393075 DOI: 10.1097/md.0000000000013264] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Previous studies on the association between statin use and survival outcomes in gynecologic cancers have presented conflicting results. No independent studies to elucidate the association between statin use and survival outcomes of endometrial cancer (EC) have been conducted. METHODS To gather updated evidence, we carried out an extensive literature search on Medline (PubMed and OvidSP), Embase, Cochrane Library, China National Knowledge Infrastructure (CNKI), wanfang data, and Vip network to identify all potential studies on the effect of statins on the prognosis of endometrial carcinoma. The design and quality of all studies were evaluated, and a fixed-effects model was used to calculate pooled hazard ratios (HRs) for overall survival (OS) and disease-specific survival (DSS). RESULTS Of the 219 articles screened, 9 articles were eligible, including 8 articles and 1 abstract. A total of 5923 patients with endometrial cancer who used statins were identified. Statin use was related to increased overall survival (HR, 0.80; 95% confidence interval [CI], 0.66-0.95, without significant heterogeneity, I = 52%, P = .080). Statin users also had increased disease-specific survival (HR, 0.69; 95% CI, 0.61-0.79, I = 0.0%). CONCLUSION Statins are beneficial to the survival outcome of patients with endometrial cancer. The selection of statins as a 1st-line agent seems justified for endometrial carcinoma.
Collapse
Affiliation(s)
- Jia Li
- Weifang Medical University
| | - Ruijuan Liu
- Weifang Traditional Chinese Hospital, Weifang City
| | - Zhengdi Sun
- Weifang Traditional Chinese Hospital, Weifang City
| | - Shifeng Tang
- Weifang Traditional Chinese Hospital, Weifang City
| | - Lu Wang
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | - Cun Liu
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| | | | | | - Changgang Sun
- Shandong University of Traditional Chinese Medicine, Jinan City, Shandong Province, China
| |
Collapse
|
139
|
Priotti J, Baglioni MV, García A, Rico MJ, Leonardi D, Lamas MC, Menacho Márquez M. Repositioning of Anti-parasitic Drugs in Cyclodextrin Inclusion Complexes for Treatment of Triple-Negative Breast Cancer. AAPS PharmSciTech 2018; 19:3734-3741. [PMID: 30255471 DOI: 10.1208/s12249-018-1169-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 08/29/2018] [Indexed: 02/06/2023] Open
Abstract
Drug repositioning refers to the identification of new therapeutic indications for drugs already approved. Albendazole and ricobendazole have been used as anti-parasitic drugs for many years; their therapeutic action is based on the inhibition of microtubule formation. Therefore, the study of their properties as antitumor compounds and the design of an appropriate formulation for cancer therapy is an interesting issue to investigate. The selected compounds are poorly soluble in water, and consequently, they have low and erratic bioavailability. In order to improve their biopharmaceutics properties, several formulations employing cyclodextrin inclusion complexes were developed. To carefully evaluate the in vitro and in vivo antitumor activity of these drugs and their complexes, several studies were performed on a breast cancer cell line (4T1) and BALB/c mice. In vitro studies showed that albendazole presented improved antitumor activity compared with ricobendazole. Furthermore, albendazole:citrate-β-cyclodextrin complex decreased significantly 4T1 cell growth both in in vitro and in vivo experiments. Thus, new formulations for anti-parasitic drugs could help to reposition them for new therapeutic indications, offering safer and more effective treatments by using a well-known drug.
Collapse
|
140
|
Liu TP, Hsieh YY, Chou CJ, Yang PM. Systematic polypharmacology and drug repurposing via an integrated L1000-based Connectivity Map database mining. ROYAL SOCIETY OPEN SCIENCE 2018; 5:181321. [PMID: 30564416 PMCID: PMC6281908 DOI: 10.1098/rsos.181321] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 11/02/2018] [Indexed: 05/19/2023]
Abstract
Drug repurposing aims to find novel indications of clinically used or experimental drugs. Because drug data already exist, drug repurposing may save time and cost, and bypass safety concerns. Polypharmacology, one drug with multiple targets, serves as a basis for drug repurposing. Large-scale databases have been accumulated in recent years, and utilization and integration of these databases would be highly helpful for polypharmacology and drug repurposing. The Connectivity Map (CMap) is a database collecting gene-expression profiles of drug-treated human cancer cells, which has been widely used for investigation of polypharmacology and drug repurposing. In this study, we integrated the next-generation L1000-based CMap and an analytic Web tool, the L1000FWD, for systematic analyses of polypharmacology and drug repurposing. Two different types of anti-cancer drugs were used as proof-of-concept examples, including histone deacetylase (HDAC) inhibitors and topoisomerase inhibitors. We identified KM-00927 and BRD-K75081836 as novel HDAC inhibitors and mitomycin C as a topoisomerase IIB inhibitor. Our study provides a prime example of utilization and integration of the freely available public resources for systematic polypharmacology analysis and drug repurposing.
Collapse
Affiliation(s)
- Tsang-Pai Liu
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, Republic of China
- Department of Surgery, Mackay Memorial Hospital, Taipei, Taiwan, Republic of China
- Liver Medical Center, Mackay Memorial Hospital, Taipei, Taiwan, Republic of China
- Mackay Junior College of Medicine, Nursing and Management, New Taipei City, Taiwan, Republic of China
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan, Republic of China
| | - Yao-Yu Hsieh
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, Republic of China
- Division of Hematology and Oncology, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan, Republic of China
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan, Republic of China
| | - Chia-Jung Chou
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, Republic of China
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, Republic of China
- TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan, Republic of China
| | - Pei-Ming Yang
- PhD Program for Cancer Molecular Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University and Academia Sinica, Taipei, Taiwan, Republic of China
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan, Republic of China
- TMU Research Center of Cancer Translational Medicine, Taipei, Taiwan, Republic of China
- Author for correspondence: Pei-Ming Yang e-mail:
| |
Collapse
|
141
|
Seliger C, Luber C, Gerken M, Schaertl J, Proescholdt M, Riemenschneider MJ, Meier CR, Bogdahn U, Leitzmann MF, Klinkhammer-Schalke M, Hau P. Use of metformin and survival of patients with high-grade glioma. Int J Cancer 2018; 144:273-280. [PMID: 30091464 DOI: 10.1002/ijc.31783] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 07/03/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
High-grade glioma (HGG) is associated with poor prognosis. Drug repurposing evolves as new modality to improve standard therapy. The antidiabetic drug metformin has been found to inhibit glioma cell growth in vitro and in vivo. The aim of the present retrospective cohort study was to evaluate the survival of patients with HGG with or without treatment with metformin, based on a large cohort of a cancer registry. The analysis included 1,093 patients with HGG diagnosed between 1998 and 2013 from the population-based clinical cancer registry Regensburg (Germany), which covers 2.1 Mio inhabitants and 98% of all cancer diagnoses. We performed multivariable adjusted Cox-regression analyses. Hazard Ratios (HRs) with 95% Confidence Intervals (CIs) for overall survival (OS) and progression-free survival (PFS) of patients with HGG with or without treatment with metformin were obtained. Use of metformin was associated with a significantly better overall and progression-free survival of patients with WHO grade III glioma (HR for OS = 0.30; 95% CI = 0.11-0.81, HR for PFS = 0.29; 95% CI = 0.11-0.78), while there were no significant relations with OS (HR = 0.83; 95% CI = 0.57-1.20) or PFS (HR = 0.85; 95% CI = 0.59-1.22) in patients with WHO grade IV glioma. In conclusion, use of metformin is associated with better overall and progression-free survival of patients with WHO grade III. Possible underlying mechanisms include the higher prevalence of IDH mutations in WHO grade III glioma, which might sensitize to the metabolic drug metformin.
Collapse
Affiliation(s)
- Corinna Seliger
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Christian Luber
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Michael Gerken
- Institute for Quality Assurance and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Julia Schaertl
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Martin Proescholdt
- Department of Neurosurgery, Regensburg University Hospital, Regensburg, Germany
| | | | - Christoph R Meier
- Basel Pharmacoepidemiology Unit, Division of CIinical Pharmacy and Epidemiology, Department of Pharmaceutical Sciences, University of Basel, Switzerland.,Boston Collaborative Drug Surveillance Program, Boston University School of Public Health, Boston University, USA.,Department of Pharmaceutical Sciences, Hospital Pharmacy, University Hospital Basel, Switzerland
| | - Ulrich Bogdahn
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| | - Michael F Leitzmann
- Department of Epidemiology and Preventive Medicine, Institute of Epidemiology and Preventive Medicine, University of Regensburg, Regensburg, Germany
| | - Monika Klinkhammer-Schalke
- Institute for Quality Assurance and Health Services Research, University of Regensburg, Regensburg, Germany
| | - Peter Hau
- Department of Neurology and Wilhelm Sander-NeuroOncology Unit, Regensburg University Hospital, Regensburg, Germany
| |
Collapse
|
142
|
Hu D, Xu H, Xiao B, Li D, Zhou Z, Liu X, Tang J, Shen Y. Albumin-Stabilized Metal-Organic Nanoparticles for Effective Delivery of Metal Complex Anticancer Drugs. ACS APPLIED MATERIALS & INTERFACES 2018; 10:34974-34982. [PMID: 30238746 DOI: 10.1021/acsami.8b12812] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Many metal-organic complexes showed potent anticancer efficacy, but their clinical applications were limited by the lack of administration route because of their poor solubility. To make metal-organic nanoparticles (MONPs) comprising metal complex drugs is a new formulation strategy for their administration. Herein, we developed a facile synthesis of an MONP composed of bovine serum albumin (BSA), Cu2+, and an anticancer agent, 5-nitro-8-hydroxyquinoline (NQ) with albumin as a nanoreactor. The resultant BSA/Cu/NQ nanoparticle (BSA/Cu/NQ NP) showed good stability in different physiological buffers and could target tumors through the enhanced permeability and retention effect and receptor-mediated cellular uptake. As the BSA/Cu/NQ NP could be readily and efficiently internalized by cancer cells, it showed much higher cytotoxic cancer cells than the NQ + Cu(II) complex and NQ. Therefore, the treatment with BSA/Cu/NQ NP noticeably enhanced the anticancer efficacy without causing systemic toxicity, indicating that such a facile preparation method has great potential to prepare other metal complex nanoparticles for drug delivery.
Collapse
Affiliation(s)
- Ding Hu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Hongxia Xu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Bing Xiao
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Dongdong Li
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Zhuxian Zhou
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Xiangrui Liu
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Jianbin Tang
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| | - Youqing Shen
- Key Laboratory of Biomass Chemical Engineering of Ministry of Education, Center for Bionanoengineering, College of Chemical and Biological Engineering , Zhejiang University , Hangzhou 310027 , People's Republic of China
| |
Collapse
|
143
|
Feng Z, Xia Y, Gao T, Xu F, Lei Q, Peng C, Yang Y, Xue Q, Hu X, Wang Q, Wang R, Ran Z, Zeng Z, Yang N, Xie Z, Yu L. The antipsychotic agent trifluoperazine hydrochloride suppresses triple-negative breast cancer tumor growth and brain metastasis by inducing G0/G1 arrest and apoptosis. Cell Death Dis 2018; 9:1006. [PMID: 30258182 PMCID: PMC6158270 DOI: 10.1038/s41419-018-1046-3] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 02/08/2023]
Abstract
Women with aggressive triple-negative breast cancer (TNBC) are at high risk of brain metastasis, which has no effective therapeutic option partially due to the poor penetration of drugs across the blood-brain barrier. Trifluoperazine (TFP) is an approved antipsychotic drug with good bioavailability in brain and had shown anticancer effect in several types of cancer. It drives us to investigate its activities to suppress TNBC, especially the brain metastasis. In this study, we chose three TNBC cell lines MDA-MB-468, MDA-MB-231, and 4T1 to assess its anticancer activities along with the possible mechanisms. In vitro, it induced G0/G1 cell cycle arrest via decreasing the expression of both cyclinD1/CDK4 and cyclinE/CDK2, and stimulated mitochondria-mediated apoptosis. In vivo, TFP suppressed the growth of subcutaneous xenograft tumor and brain metastasis without causing detectable side effects. Importantly, it prolonged the survival of mice bearing brain metastasis. Immunohistochemical analysis of Ki67 and cleaved caspase-3 indicated TFP could suppress the growth and induce apoptosis of cancer cells in vivo. Taken together, TFP might be a potential available drug for treating TNBC with brain metastasis, which urgently needs novel treatment options.
Collapse
Affiliation(s)
- Zhanzhan Feng
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Yong Xia
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Tiantao Gao
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Fuyan Xu
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Qian Lei
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Cuiting Peng
- School of Chemical Engineering, Sichuan University, 610041, Chengdu, China
| | - Yufei Yang
- Sichuan Yuanda Shuyang Pharmaceutical Co., Ltd., 610041, Chengdu, China
| | - Qiang Xue
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Xi Hu
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Qianqian Wang
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China
| | - Ranran Wang
- West China School of Pharmacy, Sichuan University, 610041, Chengdu, China
| | - Zhiqiang Ran
- West China School of Pharmacy, Sichuan University, 610041, Chengdu, China
| | - Zhilin Zeng
- West China School of Pharmacy, Sichuan University, 610041, Chengdu, China
| | - Nan Yang
- West China School of Pharmacy, Sichuan University, 610041, Chengdu, China
| | - Zixin Xie
- West China School of Pharmacy, Sichuan University, 610041, Chengdu, China
| | - Luoting Yu
- Lab of Medicinal Chemistry, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, 610041, Chengdu, China.
| |
Collapse
|
144
|
Drug Repurposing of Metabolic Agents in Malignant Glioma. Int J Mol Sci 2018; 19:ijms19092768. [PMID: 30223473 PMCID: PMC6164672 DOI: 10.3390/ijms19092768] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 09/10/2018] [Accepted: 09/11/2018] [Indexed: 12/21/2022] Open
Abstract
Gliomas are highly invasive brain tumors with short patient survival. One major pathogenic factor is aberrant tumor metabolism, which may be targeted with different specific and unspecific agents. Drug repurposing is of increasing interest in glioma research. Drugs interfering with the patient’s metabolism may also influence glioma metabolism. In this review, we outline definitions and methods for drug repurposing. Furthermore, we give insights into important candidates for a metabolic drug repurposing, namely metformin, statins, non-steroidal anti-inflammatory drugs, disulfiram and lonidamine. Advantages and pitfalls of drug repurposing will finally be discussed.
Collapse
|
145
|
Yarmolinsky J, Wade KH, Richmond RC, Langdon RJ, Bull CJ, Tilling KM, Relton CL, Lewis SJ, Davey Smith G, Martin RM. Causal Inference in Cancer Epidemiology: What Is the Role of Mendelian Randomization? Cancer Epidemiol Biomarkers Prev 2018; 27:995-1010. [PMID: 29941659 PMCID: PMC6522350 DOI: 10.1158/1055-9965.epi-17-1177] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/15/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023] Open
Abstract
Observational epidemiologic studies are prone to confounding, measurement error, and reverse causation, undermining robust causal inference. Mendelian randomization (MR) uses genetic variants to proxy modifiable exposures to generate more reliable estimates of the causal effects of these exposures on diseases and their outcomes. MR has seen widespread adoption within cardio-metabolic epidemiology, but also holds much promise for identifying possible interventions for cancer prevention and treatment. However, some methodologic challenges in the implementation of MR are particularly pertinent when applying this method to cancer etiology and prognosis, including reverse causation arising from disease latency and selection bias in studies of cancer progression. These issues must be carefully considered to ensure appropriate design, analysis, and interpretation of such studies. In this review, we provide an overview of the key principles and assumptions of MR, focusing on applications of this method to the study of cancer etiology and prognosis. We summarize recent studies in the cancer literature that have adopted a MR framework to highlight strengths of this approach compared with conventional epidemiological studies. Finally, limitations of MR and recent methodologic developments to address them are discussed, along with the translational opportunities they present to inform public health and clinical interventions in cancer. Cancer Epidemiol Biomarkers Prev; 27(9); 995-1010. ©2018 AACR.
Collapse
Affiliation(s)
- James Yarmolinsky
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kaitlin H Wade
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ryan J Langdon
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Caroline J Bull
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Kate M Tilling
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Sarah J Lewis
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Richard M Martin
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom.
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
146
|
Lee TY, Martinez-Outschoorn UE, Schilder RJ, Kim CH, Richard SD, Rosenblum NG, Johnson JM. Metformin as a Therapeutic Target in Endometrial Cancers. Front Oncol 2018; 8:341. [PMID: 30211120 PMCID: PMC6121131 DOI: 10.3389/fonc.2018.00341] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 08/06/2018] [Indexed: 01/01/2023] Open
Abstract
Endometrial cancer is the most common gynecologic malignancy in developed countries. Its increasing incidence is thought to be related in part to the rise of metabolic syndrome, which has been shown to be a risk factor for the development of hyperestrogenic and hyperinsulinemic states. This has consequently lead to an increase in other hormone-responsive cancers as well e.g., breast and ovarian cancer. The correlation between obesity, hyperglycemia, and endometrial cancer has highlighted the important role of metabolism in cancer establishment and persistence. Tumor-mediated reprogramming of the microenvironment and macroenvironment can range from induction of cytokines and growth factors to stimulation of surrounding stromal cells to produce energy-rich catabolites, fueling the growth, and survival of cancer cells. Such mechanisms raise the prospect of the metabolic microenvironment itself as a viable target for treatment of malignancies. Metformin is a biguanide drug that is a first-line treatment for type 2 diabetes that has beneficial effects on various markers of the metabolic syndrome. Many studies suggest that metformin shows potential as an adjuvant treatment for uterine and other cancers. Here, we review the evidence for metformin as a treatment for cancers of the endometrium. We discuss the available clinical data and the molecular mechanisms by which it may exert its effects, with a focus on how it may alter the tumor microenvironment. The pleiotropic effects of metformin on cellular energy production and usage as well as intercellular and hormone-based interactions make it a promising candidate for reprogramming of the cancer ecosystem. This, along with other treatments aimed at targeting tumor metabolic pathways, may lead to novel treatment strategies for endometrial cancer.
Collapse
Affiliation(s)
- Teresa Y Lee
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | | | - Russell J Schilder
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Christine H Kim
- Department of Obstetrics and Gynecology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Scott D Richard
- Department of Obstetrics and Gynecology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Norman G Rosenblum
- Department of Obstetrics and Gynecology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Jennifer M Johnson
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
147
|
Mohammadi A, Sharifi A, Pourpaknia R, Mohammadian S, Sahebkar A. Manipulating macrophage polarization and function using classical HDAC inhibitors: Implications for autoimmunity and inflammation. Crit Rev Oncol Hematol 2018; 128:1-18. [DOI: 10.1016/j.critrevonc.2018.05.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/18/2018] [Accepted: 05/10/2018] [Indexed: 02/06/2023] Open
|
148
|
Stokes WA, Eguchi M, Amini A, Hararah MK, Ding D, McDermott JD, Bradley CJ, Karam SD. Survival impact and toxicity of metformin in head and neck cancer: An analysis of the SEER-Medicare dataset. Oral Oncol 2018; 84:12-19. [PMID: 30115470 DOI: 10.1016/j.oraloncology.2018.06.022] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 06/11/2018] [Accepted: 06/18/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES Recent preclinical research has renewed interest in the interplay between glucose dysregulation and cancer. Metformin holds promise as an adjunctive antineoplastic agent in head and neck cancer (HNC). We aimed to explore the impact of metformin in HNC patients from a population-based dataset. PATIENTS & METHODS Patients diagnosed with HNC from 2008 to 2011 were identified from the Surveillance, Epidemiology, and End Results (SEER)-Medicare linked dataset and categorized into three groups: non-diabetics (nD), diabetics not taking metformin (DnM), and diabetics taking metformin (D + M). Overall survival (OS) and cancer-specific survival (CSS) were compared between groups using Kaplan-Meier and Cox regression controlling for sociodemographic, clinical, and treatment covariates. The incidence of toxicities associated with HNC therapy was compared among groups using χ2 analysis. RESULTS Among 1646 patients, there were 1144 nD, 378 DnM, and 124 D + M. 2-year OS rates was 65.6% for nD, 57.7% for DnM, and 73.4% for D + M by Kaplan-Meier (p < 0.01), and corresponding rates of 2-year CSS were 73.7%, 66.1%, and 88.8% (p < 0.01), respectively. On Cox multivariable analysis, OS among the three groups did not significantly differ; however, CSS was significantly worse among both nD versus DnM as compared to D + M. Toxicity rates were not significantly increased among D + M. CONCLUSION HNC patients with diabetes taking metformin experience improved CSS. Prospective investigation of the addition of metformin to standard-of-care HNC therapy is warranted.
Collapse
Affiliation(s)
- William A Stokes
- Department of Radiation Oncology, University of Colorado Denver, USA
| | - Megan Eguchi
- Department of Health Systems, Management, and Policy, Colorado School of Public Health, USA
| | - Arya Amini
- Department of Radiation Oncology, City of Hope Cancer Center, USA
| | | | - Ding Ding
- Department of Radiation Oncology, University of Colorado Denver, USA
| | | | - Cathy J Bradley
- Department of Health Systems, Management, and Policy, Colorado School of Public Health, USA
| | - Sana D Karam
- Department of Radiation Oncology, University of Colorado Denver, USA.
| |
Collapse
|
149
|
El-Azab AS, Abdel-Aziz AAM, Abou-Zeid LA, El-Husseiny WM, El Morsy AM, El-Gendy MA, El-Sayed MAA. Synthesis, antitumour activities and molecular docking of thiocarboxylic acid ester-based NSAID scaffolds: COX-2 inhibition and mechanistic studies. J Enzyme Inhib Med Chem 2018; 33:989-998. [PMID: 29806488 PMCID: PMC6009944 DOI: 10.1080/14756366.2018.1474878] [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] [Indexed: 12/21/2022] Open
Abstract
A new series of NSAID thioesters were synthesized and evaluated for their in vitro antitumor effects against a panel of four human tumor cell lines, namely: HepG2, MCF-7, HCT-116 and Caco-2, using the MTT assay. Compared to the reference drugs 5-FU, afatinib and celecoxib, compounds 2b, 3b, 6a, 7a, 7b and 8a showed potent broad-spectrum antitumor activity against the selected tumour cell lines. Accordingly, these compounds were selected for mechanistic studies about COX inhibition and kinase assays. In vitro COX-1/COX-2 enzyme inhibition assay results indicated that compounds 2b, 3b, 6a, 7a, 7b, 8a and 8 b selectively inhibited the COX-2 enzyme (IC50 = ∼0.20–0.69 μM), with SI values of (>72.5–250) compared with celecoxib (IC50 = 0.16 μM, COX-2 SI: > 312.5); however, all the tested compounds did not inhibit the COX-1 enzyme (IC50 > 50 μM). On the other hand, EGFR, HER2, HER4 and cSrc kinase inhibition assays were evaluated at a 10 μM concentration. The selected candidates displayed limited activities against the various tested kinases; the compounds 2a, 3b, 6a, 7a, 7b and 8a showed no activity to weak activity (% inhibition = ∼0–10%). The molecular docking study revealed the importance of the thioester moiety for the interaction of the drugs with the amino acids in the active sites of COX-2. The aforementioned results indicated that thioester based on NSAID scaffolds derivatives may serve as new antitumor compounds.
Collapse
Affiliation(s)
- Adel S El-Azab
- a Department of Pharmaceutical Chemistry, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia.,b Department of Organic Chemistry, Faculty of Pharmacy , Al-Azhar University , Cairo , Egypt
| | - Alaa A-M Abdel-Aziz
- a Department of Pharmaceutical Chemistry, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia.,c Department of Medicinal Chemistry, Faculty of Pharmacy , Mansoura University , Mansoura , Egypt
| | - Laila A Abou-Zeid
- d Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Mansoura University , Mansoura , Egypt
| | - Walaa M El-Husseiny
- d Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Mansoura University , Mansoura , Egypt
| | - Ahmad M El Morsy
- b Department of Organic Chemistry, Faculty of Pharmacy , Al-Azhar University , Cairo , Egypt
| | - Manal A El-Gendy
- a Department of Pharmaceutical Chemistry, College of Pharmacy , King Saud University , Riyadh , Saudi Arabia
| | - Magda A-A El-Sayed
- d Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy , Mansoura University , Mansoura , Egypt.,e Department of Pharmaceutical Chemistry, Faculty of pharmacy , Horus university , New Damietta , Egypt
| |
Collapse
|
150
|
Nguyen TM, Muhammad SA, Ibrahim S, Ma L, Guo J, Bai B, Zeng B. DeCoST: A New Approach in Drug Repurposing From Control System Theory. Front Pharmacol 2018; 9:583. [PMID: 29922160 PMCID: PMC5996185 DOI: 10.3389/fphar.2018.00583] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 05/15/2018] [Indexed: 01/19/2023] Open
Abstract
In this paper, we propose DeCoST (Drug Repurposing from Control System Theory) framework to apply control system paradigm for drug repurposing purpose. Drug repurposing has become one of the most active areas in pharmacology since the last decade. Compared to traditional drug development, drug repurposing may provide more systematic and significantly less expensive approaches in discovering new treatments for complex diseases. Although drug repurposing techniques rapidly evolve from "one: disease-gene-drug" to "multi: gene, dru" and from "lazy guilt-by-association" to "systematic model-based pattern matching," mathematical system and control paradigm has not been widely applied to model the system biology connectivity among drugs, genes, and diseases. In this paradigm, our DeCoST framework, which is among the earliest approaches in drug repurposing with control theory paradigm, applies biological and pharmaceutical knowledge to quantify rich connective data sources among drugs, genes, and diseases to construct disease-specific mathematical model. We use linear-quadratic regulator control technique to assess the therapeutic effect of a drug in disease-specific treatment. DeCoST framework could classify between FDA-approved drugs and rejected/withdrawn drug, which is the foundation to apply DeCoST in recommending potentially new treatment. Applying DeCoST in Breast Cancer and Bladder Cancer, we reprofiled 8 promising candidate drugs for Breast Cancer ER+ (Erbitux, Flutamide, etc.), 2 drugs for Breast Cancer ER- (Daunorubicin and Donepezil) and 10 drugs for Bladder Cancer repurposing (Zafirlukast, Tenofovir, etc.).
Collapse
Affiliation(s)
- Thanh M Nguyen
- Department of Computer and Information Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Syed A Muhammad
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Pakistan
| | - Sara Ibrahim
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN, United States
| | - Lin Ma
- The 1st School of Medicine and School of Information and Engineering, Wenzhou Medical University, Zhejiang, China
| | - Jinlei Guo
- The 1st School of Medicine and School of Information and Engineering, Wenzhou Medical University, Zhejiang, China
| | - Baogang Bai
- The 1st School of Medicine and School of Information and Engineering, Wenzhou Medical University, Zhejiang, China
| | - Bixin Zeng
- Institute of Lasers and Biomedical Photonics, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|