251
|
Molecules and Mechanisms to Overcome Oxidative Stress Inducing Cardiovascular Disease in Cancer Patients. Life (Basel) 2021; 11:life11020105. [PMID: 33573162 PMCID: PMC7911715 DOI: 10.3390/life11020105] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/18/2021] [Accepted: 01/27/2021] [Indexed: 02/06/2023] Open
Abstract
Reactive oxygen species (ROS) are molecules involved in signal transduction pathways with both beneficial and detrimental effects on human cells. ROS are generated by many cellular processes including mitochondrial respiration, metabolism and enzymatic activities. In physiological conditions, ROS levels are well-balanced by antioxidative detoxification systems. In contrast, in pathological conditions such as cardiovascular, neurological and cancer diseases, ROS production exceeds the antioxidative detoxification capacity of cells, leading to cellular damages and death. In this review, we will first describe the biology and mechanisms of ROS mediated oxidative stress in cardiovascular disease. Second, we will review the role of oxidative stress mediated by oncological treatments in inducing cardiovascular disease. Lastly, we will discuss the strategies that potentially counteract the oxidative stress in order to fight the onset and progression of cardiovascular disease, including that induced by oncological treatments.
Collapse
|
252
|
The Protein-Binding Behavior of Platinum Anticancer Drugs in Blood Revealed by Mass Spectrometry. Pharmaceuticals (Basel) 2021; 14:ph14020104. [PMID: 33572935 PMCID: PMC7911130 DOI: 10.3390/ph14020104] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/22/2021] [Accepted: 01/26/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin and its analogues are widely used as chemotherapeutic agents in clinical practice. After being intravenously administrated, a substantial amount of platinum will bind with proteins in the blood. This binding is vital for the transport, distribution, and metabolism of drugs; however, toxicity can also occur from the irreversible binding between biologically active proteins and platinum drugs. Therefore, it is very important to study the protein-binding behavior of platinum drugs in blood. This review summarizes mass spectrometry-based strategies to identify and quantitate the proteins binding with platinum anticancer drugs in blood, such as offline high-performance liquid chromatography/inductively coupled plasma mass spectrometry (HPLC–ICP-MS) combined with electrospray ionization mass spectrometry (ESI-MS/MS) and multidimensional LC–ESI-MS/MS. The identification of in vivo targets in blood cannot be accomplished without first studying the protein-binding behavior of platinum drugs in vitro; therefore, relevant studies are also summarized. This knowledge will further our understanding of the pharmacokinetics and toxicity of platinum anticancer drugs, and it will be beneficial for the rational design of metal-based anticancer drugs.
Collapse
|
253
|
Ganzinelli M, Linardou H, Alvisi MF, Caiola E, Lo Russo G, Cecere FL, Bettini AC, Psyrri A, Milella M, Rulli E, Fabbri A, De Maglie M, Romanelli P, Murray S, Broggini M, Marabese M, Garassino MC. Single-arm, open label prospective trial to assess prediction of the role of ERCC1/XPF complex in the response of advanced NSCLC patients to platinum-based chemotherapy. ESMO Open 2021; 6:100034. [PMID: 33422766 PMCID: PMC7809372 DOI: 10.1016/j.esmoop.2020.100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/24/2020] [Accepted: 12/06/2020] [Indexed: 11/05/2022] Open
Abstract
Background Platinum-based therapy, combined or not with immune checkpoint inhibitors, represents a front-line choice for patients with non-small-cell lung cancer (NSCLC). Despite the improved outcomes in the last years for this malignancy, only a sub-group of patients have long-term benefit. Excision repair cross-complementation group 1 (ERCC1) has been considered a potential biomarker to predict the outcome of platinum-based chemotherapy in NSCLC. However, the ERCC1 gene is transcribed in four splice variants where the isoform 202 was described as the only one active and able to complex Xeroderma pigmentosum group F-complementing protein (XPF). Here, we prospectively investigated if the active form of ERCC1, as assessed by the ERCC1/XPF complex (ERCC1/XPF), could predict the sensitivity to platinum compounds. Patients and methods Prospectively enrolled, patients with advanced NSCLC treated with a first-line regimen containing platinum were centrally evaluated for ERCC1/XPF by a proximity ligation assay. Overall survival (OS), progression-free survival (PFS) and objective response rate (ORR) were analyzed. Results The absence of the ERCC1/XPF in the tumor suggested a trend of worst outcomes in terms of both OS [hazard ratio (HR) 1.41, 95% confidence interval (CI) 0.67-2.94, P = 0.373] and PFS (HR 1.61, 95% CI 0.88-3.03, P = 0.123). ORR was marginally influenced in ERCC1/XPF-negative and -positive groups [odds ratio (stable disease + progressive disease versus complete response + partial response) 0.87, 95% CI 0.25-3.07, P = 0.832]. Conclusion The lack of ERCC1/XPF complex in NSCLC tumor cells might delineate a group of patients with poor outcomes when treated with platinum compounds. ERCC1/XPF absence might well identify patients for whom a different therapeutic approach could be necessary. This is the first study investigating the ERCC1/XPF complex as a platinum-based therapy response biomarker in NSCLC. The lack of ERCC1/XPF complex might delineate a group of patients with poor outcomes when treated with platinum compounds. ERCC1/XPF absence might identify tumors for whom a different therapeutic approach than platinum compounds could be necessary.
Collapse
Affiliation(s)
- M Ganzinelli
- Unit of Thoracic Oncology, Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - H Linardou
- 4th Oncology Department, Metropolitan Hospital, Athens, Greece
| | - M F Alvisi
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - E Caiola
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - G Lo Russo
- Unit of Thoracic Oncology, Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - F L Cecere
- Division of Medical Oncology 1, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - A C Bettini
- UO Oncologia Medica, ASST Papa Giovanni XXIII, Bergamo, Italy
| | - A Psyrri
- Section of Oncology, Department of Internal Medicine, Attikon Hospital, National Kapodistrian University of Athens, Athens, Greece
| | - M Milella
- Department of Medicine, Section of Medical Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - E Rulli
- Laboratory of Methodology for Clinical Research, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - A Fabbri
- Department of Pathology and Laboratory Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - M De Maglie
- Mouse and Animal Pathology Lab, Fondazione Filarete, Milan, Italy; Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - P Romanelli
- Mouse and Animal Pathology Lab, Fondazione Filarete, Milan, Italy; Department of Veterinary Medicine, University of Milan, Milan, Italy
| | - S Murray
- Biomarker Solutions Ltd, London, UK
| | - M Broggini
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
| | - M Marabese
- Laboratory of Molecular Pharmacology, Department of Oncology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - M C Garassino
- Unit of Thoracic Oncology, Medical Oncology Department 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
254
|
Un H, Ugan RA, Gurbuz MA, Bayir Y, Kahramanlar A, Kaya G, Cadirci E, Halici Z. Phloretin and phloridzin guard against cisplatin-induced nephrotoxicity in mice through inhibiting oxidative stress and inflammation. Life Sci 2020; 266:118869. [PMID: 33309722 DOI: 10.1016/j.lfs.2020.118869] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 12/13/2022]
Abstract
AIM Cisplatin (Cis) is widely used chemotherapeutic and has some serious side effects as nephrotoxicity. Phloretin (PH) and Phloridzin (PZ) are known their anti-oxidant anti-inflammatory effects. We aimed to examine the protective effects of PH and PZ on cisplatin-induced nephrotoxicity. MAIN METHODS Totally, 48 Balb/C female mice were separated into eight groups (n = 6). First day, single dose of cisplatin (20 mg/kg intraperitoneal) was administered to induce toxicity. PH and PZ were given (50 and 100 mg/kg orally) to treatment groups during 3 days. After the experimental procedures serum renal function enzymes (BUN and Creatinine), oxidative parameters (SOD, GSH and MDA), nuclear agent NFKβ, inflammatory cytokines (Tnf-α and IL1β) and HSP70 expressions and histopathological assessments were analyzed. KEY FINDINGS Serum enzymes, tissue cytokines and oxidative stress were increased after the Cis treatment. PH and PZ treatments normalized all parameters compared to Cis administrated group. After the treatments, SOD activities and GSH levels were increased while MDA levels were decreased. PH and PZ treatments decreased Tnf-α, IL1β and NFKβ mRNA expressions. Cis significantly increased the HSP70 expression while PH and PZ administrations significantly decreased. Similar the biochemical and molecular results, PH and PZ showed positive effects on tissue pathological parameters. Cisplatin cause a lot of abnormal structures as tubular and glomeruli damages on the kidney. SIGNIFICANCE PH and PZ play important physiological roles in the prevention of nephrotoxicity. Antioxidant and anti-inflammatory effects of PH and PZ demonstrated visible protective effects in the cisplatin-induced nephrotoxicity model.
Collapse
Affiliation(s)
- Harun Un
- Agri Ibrahim Cecen University, Faculty of Pharmacy, Department of Biochemistry, Agri, Turkey.
| | - Rustem Anil Ugan
- Ataturk University, Faculty of Pharmacy, Department of Pharmacology, Erzurum, Turkey
| | - Muhammet Ali Gurbuz
- Ataturk University, Faculty of Medicine, Department of Histology and Embryology, Erzurum, Turkey
| | - Yasin Bayir
- Ataturk University, Faculty of Pharmacy, Department of Biochemistry, Erzurum, Turkey
| | - Aysenur Kahramanlar
- Ataturk University, Faculty of Pharmacy, Department of Biochemistry, Erzurum, Turkey
| | - Gokce Kaya
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey
| | - Elif Cadirci
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| | - Zekai Halici
- Ataturk University, Faculty of Medicine, Department of Pharmacology, Erzurum, Turkey; Clinical Research, Development and Design Application and Research Center, Ataturk University, Erzurum, Turkey
| |
Collapse
|
255
|
Shao F, Lyu X, Miao K, Xie L, Wang H, Xiao H, Li J, Chen Q, Ding R, Chen P, Xing F, Zhang X, Luo G, Zhu W, Cheng G, Lon NW, Martin SE, Wang G, Chen G, Dai Y, Deng C. Enhanced Protein Damage Clearance Induces Broad Drug Resistance in Multitype of Cancers Revealed by an Evolution Drug-Resistant Model and Genome-Wide siRNA Screening. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:2001914. [PMID: 33304752 PMCID: PMC7709997 DOI: 10.1002/advs.202001914] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/09/2020] [Indexed: 05/08/2023]
Abstract
Resistance to therapeutic drugs occurs in virtually all types of cancers, and the tolerance to one drug frequently becomes broad therapy resistance; however, the underlying mechanism remains elusive. Combining a whole whole-genome-wide RNA interference screening and an evolutionary drug pressure model with MDA-MB-231 cells, it is found that enhanced protein damage clearance and reduced mitochondrial respiratory activity are responsible for cisplatin resistance. Screening drug-resistant cancer cells and human patient-derived organoids for breast and colon cancers with many anticancer drugs indicates that activation of mitochondrion protein import surveillance system enhances proteasome activity and minimizes caspase activation, leading to broad drug resistance that can be overcome by co-treatment with a proteasome inhibitor, bortezomib. It is further demonstrated that cisplatin and bortezomib encapsulated into nanoparticle further enhance their therapeutic efficacy and alleviate side effects induced by drug combination treatment. These data demonstrate a feasibility for eliminating broad drug resistance by targeting its common mechanism to achieve effective therapy for multiple cancers.
Collapse
Affiliation(s)
- Fangyuan Shao
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Xueying Lyu
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Kai Miao
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Lisi Xie
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Haitao Wang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Hao Xiao
- Guangdong Key Laboratory of Animal Breeding and NutritionInstitute of Animal ScienceGuangdong Academy of Agricultural SciencesGuangzhou510640China
| | - Jie Li
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Qiang Chen
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Renbo Ding
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Ping Chen
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Fuqiang Xing
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Department of BiologySouthern University of Science and TechnologyShenzhen518055China
| | - Xu Zhang
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | | | | | - Gregory Cheng
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Ng Wai Lon
- Centro Hospitalar Conde de S. JanuárioMacau820004China
| | - Scott E. Martin
- Division of Pre‐Clinical InnovationNational Center for Advancing Translational Sciences (NCATS)National Institutes of HealthBethesdaMD20892USA
| | - Guanyu Wang
- Department of BiologySouthern University of Science and TechnologyShenzhen518055China
| | - Guokai Chen
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Yunlu Dai
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| | - Chu‐Xia Deng
- Cancer CenterFaculty of Health SciencesUniversity of MacauMacau999078China
- Center for Precision Medicine Research and TrainingFaculty of Health SciencesUniversity of MacauMacau999078China
| |
Collapse
|
256
|
Luo P, Wu S, Ji K, Yuan X, Li H, Chen J, Tian Y, Qiu Y, Zhong X. LncRNA MIR4435-2HG mediates cisplatin resistance in HCT116 cells by regulating Nrf2 and HO-1. PLoS One 2020; 15:e0223035. [PMID: 33232319 PMCID: PMC7685444 DOI: 10.1371/journal.pone.0223035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 08/16/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE Cisplatin resistance is still a serious problem in the clinic. However, the underlying mechanism remains unknown. In our study, we investigated cisplatin resistance by using the cisplatin-resistant cell line HCT116R. METHODS The HCT116 cell line, a colon cancer cell line, was purchased. Cell viability was determined using CCK-8 Assay Kit. The gene expression levels of MIR4435-2HG, Nrf2, and HO-1, and caspase activity were determined using qRT-PCR and Caspase 3 Assay Kit, respectively. RESULTS In this study, we found that the levels of the lncRNA MIR4435-2HG were dramatically increased in the cisplatin-resistant cell line HCT116R. Knockdown of MIR4435-2HG in HCT116R cells significantly restored the sensitivity to cisplatin, inhibited cell proliferation and promoted cell apoptosis. Furthermore, Nrf2 and HO-1 mRNA levels, as critical molecules in the oxidative stress pathway, were inhibited by siRNAs targeting MIR4435-2HG, suggesting that MIR4435-2HG-mediated cisplatin resistance occurs through the Nrf2/HO-1 pathway. CONCLUSION Our findings demonstrate that the lncRNA MIR4435-2HG is a main factor driving the cisplatin resistance of HCT116 cells.
Collapse
Affiliation(s)
- Ping Luo
- Department of Breast Tumor of Nanchang Third Hospital, Nanchang, Jiangxi, China
| | - Shugui Wu
- Department of Oncology of Ganzhou People’s Hospital, Ganzhou, Jiangxi, China
| | - Kaibao Ji
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Hubei, China
| | - Xia Yuan
- Department of Tumor Radiotherapy of Jiangxi Province Cancer Hospital, Nanchang, Jiangxi, China
| | - Hongmi Li
- Department of Tumor Radiotherapy of Jiangxi Province Cancer Hospital, Nanchang, Jiangxi, China
| | - Jinping Chen
- Department of Oncology of Yichun People’s Hospital, Yichun, Jiangxi, China
| | - Yunfei Tian
- Department of Interventional of Ganzhou People’s Hospital, Ganzhou, Jiangxi, China
| | - Yang Qiu
- Department of Tumor Radiotherapy of Jiangxi Province Cancer Hospital, Nanchang, Jiangxi, China
- * E-mail: (YQ); (XZ)
| | - Xiaoming Zhong
- Department of Tumor Radiotherapy of Jiangxi Province Cancer Hospital, Nanchang, Jiangxi, China
- * E-mail: (YQ); (XZ)
| |
Collapse
|
257
|
Kergreis A, Lord RM, Pike SJ. Influence of Ligand and Nuclearity on the Cytotoxicity of Cyclometallated C^N^C Platinum(II) Complexes. Chemistry 2020; 26:14938-14946. [PMID: 32520417 PMCID: PMC7756510 DOI: 10.1002/chem.202002517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Indexed: 01/25/2023]
Abstract
A series of cyclometallated mono- and di-nuclear platinum(II) complexes and the parent organic ligand, 2,6-diphenylpyridine 1 (HC^N^CH), have been synthesized and characterized. This library of compounds includes [(C^N^C)PtII (L)] (L=dimethylsulfoxide (DMSO) 2 and triphenylphosphine (PPh3 ) 3) and [((C^N^C)PtII )2 (L')] (where L'=N-heterocycles (pyrazine (pyr) 4, 4,4'-bipyridine (4,4'-bipy) 5 or diphosphine (1,4-bis(diphenylphosphino)butane (dppb) 6). Their cytotoxicity was assessed against four cancerous cell lines and one normal cell line, with results highlighting significantly increased antiproliferative activity for the dinuclear complexes (4-6), when compared to the mononucleated species (2 and 3). Complex 6 is the most promising candidate, displaying very high selectivity towards cancerous cells, with selectivity index (SI) values >29.5 (A2780) and >11.2 (A2780cisR), and outperforming cisplatin by >4-fold and >18-fold, respectively.
Collapse
Affiliation(s)
- Angélique Kergreis
- School of Chemistry and BiosciencesFaculty of Life SciencesUniversity of BradfordBradford, West YorkshireBD7 1DPUK
| | - Rianne M. Lord
- School of Chemistry and BiosciencesFaculty of Life SciencesUniversity of BradfordBradford, West YorkshireBD7 1DPUK
- School of ChemistryUniversity of East AngliaNorwich Research ParkNorwichNR4 7TJUK
| | - Sarah J. Pike
- School of Chemistry and BiosciencesFaculty of Life SciencesUniversity of BradfordBradford, West YorkshireBD7 1DPUK
- School of ChemistryUniversity of BirminghamEdgbastonBirminghamB15 2TTUK
| |
Collapse
|
258
|
Gierlich P, Mata AI, Donohoe C, Brito RMM, Senge MO, Gomes-da-Silva LC. Ligand-Targeted Delivery of Photosensitizers for Cancer Treatment. Molecules 2020; 25:E5317. [PMID: 33202648 PMCID: PMC7698280 DOI: 10.3390/molecules25225317] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/26/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising cancer treatment which involves a photosensitizer (PS), light at a specific wavelength for PS activation and oxygen, which combine to elicit cell death. While the illumination required to activate a PS imparts a certain amount of selectivity to PDT treatments, poor tumor accumulation and cell internalization are still inherent properties of most intravenously administered PSs. As a result, common consequences of PDT include skin photosensitivity. To overcome the mentioned issues, PSs may be tailored to specifically target overexpressed biomarkers of tumors. This active targeting can be achieved by direct conjugation of the PS to a ligand with enhanced affinity for a target overexpressed on cancer cells and/or other cells of the tumor microenvironment. Alternatively, PSs may be incorporated into ligand-targeted nanocarriers, which may also encompass multi-functionalities, including diagnosis and therapy. In this review, we highlight the major advances in active targeting of PSs, either by means of ligand-derived bioconjugates or by exploiting ligand-targeting nanocarriers.
Collapse
Affiliation(s)
- Piotr Gierlich
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Ana I. Mata
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| | - Claire Donohoe
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Rui M. M. Brito
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
- BSIM Therapeutics, Instituto Pedro Nunes, 3030-199 Coimbra, Portugal
| | - Mathias O. Senge
- Medicinal Chemistry, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, The University of Dublin, St. James’s Hospital, D08W9RT Dublin, Ireland;
| | - Lígia C. Gomes-da-Silva
- CQC, Coimbra Chemistry Center, Department of Chemistry, University of Coimbra, 3000-435 Coimbra, Portugal; (P.G.); (A.I.M.); (C.D.); (R.M.M.B.)
| |
Collapse
|
259
|
Therapeutic Potential of the Natural Compound S-Adenosylmethionine as a Chemoprotective Synergistic Agent in Breast, and Head and Neck Cancer Treatment: Current Status of Research. Int J Mol Sci 2020; 21:ijms21228547. [PMID: 33202711 PMCID: PMC7697526 DOI: 10.3390/ijms21228547] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
The present review summarizes the most recent studies focusing on the synergistic antitumor effect of the physiological methyl donor S-adenosylmethionine (AdoMet) in association with the main drugs used against breast cancer and head and neck squamous cell carcinoma (HNSCC), two highly aggressive and metastatic malignancies. In these two tumors the chemotherapy approach is recommended as the first choice despite the numerous side effects and recurrence of metastasis, so better tolerated treatments are needed to overcome this problem. In this regard, combination therapy with natural compounds, such as AdoMet, a molecule with pleiotropic effects on multiple cellular processes, is emerging as a suitable strategy to achieve synergistic anticancer efficacy. In this context, the analysis of studies conducted in the literature highlighted AdoMet as one of the most effective and promising chemosensitizing agents to be taken into consideration for inclusion in emerging antitumor therapeutic modalities such as nanotechnologies.
Collapse
|
260
|
Gąsiorkiewicz BM, Koczurkiewicz-Adamczyk P, Piska K, Pękala E. Autophagy modulating agents as chemosensitizers for cisplatin therapy in cancer. Invest New Drugs 2020; 39:538-563. [PMID: 33159673 PMCID: PMC7960624 DOI: 10.1007/s10637-020-01032-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 02/08/2023]
Abstract
Although cisplatin is one of the most common antineoplastic drug, its successful utilisation in cancer treatment is limited by the drug resistance. Multiple attempts have been made to find potential cisplatin chemosensitisers which would overcome cancer cells resistance thus improving antineoplastic efficacy. Autophagy modulation has become an important area of interest regarding the aforementioned topic. Autophagy is a highly conservative cellular self-digestive process implicated in response to multiple environmental stressors. The high basal level of autophagy is a common phenomenon in cisplatin-resistant cancer cells which is thought to grant survival benefit. However current evidence supports the role of autophagy in either promoting or limiting carcinogenesis depending on the context. This encourages the search of substances modulating the process to alleviate cisplatin resistance. Such a strategy encompasses not only simple autophagy inhibition but also harnessing the process to induce autophagy-dependent cell death. In this paper, we briefly describe the mechanism of cisplatin resistance with a special emphasis on autophagy and we give an extensive literature review of potential substances with cisplatin chemosensitising properties related to autophagy modulation.
Collapse
Affiliation(s)
- Bartosz Mateusz Gąsiorkiewicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland.
| | - Paulina Koczurkiewicz-Adamczyk
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Kamil Piska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| | - Elżbieta Pękala
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Kraków, Poland
| |
Collapse
|
261
|
Craig M, Jenner AL, Namgung B, Lee LP, Goldman A. Engineering in Medicine To Address the Challenge of Cancer Drug Resistance: From Micro- and Nanotechnologies to Computational and Mathematical Modeling. Chem Rev 2020; 121:3352-3389. [PMID: 33152247 DOI: 10.1021/acs.chemrev.0c00356] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Drug resistance has profoundly limited the success of cancer treatment, driving relapse, metastasis, and mortality. Nearly all anticancer drugs and even novel immunotherapies, which recalibrate the immune system for tumor recognition and destruction, have succumbed to resistance development. Engineers have emerged across mechanical, physical, chemical, mathematical, and biological disciplines to address the challenge of drug resistance using a combination of interdisciplinary tools and skill sets. This review explores the developing, complex, and under-recognized role of engineering in medicine to address the multitude of challenges in cancer drug resistance. Looking through the "lens" of intrinsic, extrinsic, and drug-induced resistance (also referred to as "tolerance"), we will discuss three specific areas where active innovation is driving novel treatment paradigms: (1) nanotechnology, which has revolutionized drug delivery in desmoplastic tissues, harnessing physiochemical characteristics to destroy tumors through photothermal therapy and rationally designed nanostructures to circumvent cancer immunotherapy failures, (2) bioengineered tumor models, which have benefitted from microfluidics and mechanical engineering, creating a paradigm shift in physiologically relevant environments to predict clinical refractoriness and enabling platforms for screening drug combinations to thwart resistance at the individual patient level, and (3) computational and mathematical modeling, which blends in silico simulations with molecular and evolutionary principles to map mutational patterns and model interactions between cells that promote resistance. On the basis that engineering in medicine has resulted in discoveries in resistance biology and successfully translated to clinical strategies that improve outcomes, we suggest the proliferation of multidisciplinary science that embraces engineering.
Collapse
Affiliation(s)
- Morgan Craig
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Adrianne L Jenner
- Department of Mathematics and Statistics, University of Montreal, Montreal, Quebec H3C 3J7, Canada.,Sainte-Justine University Hospital Research Centre, Montreal, Quebec H3S 2G4, Canada
| | - Bumseok Namgung
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Luke P Lee
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| | - Aaron Goldman
- Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States.,Department of Medicine, Harvard Medical School, Boston, Massachusetts 02139, United States
| |
Collapse
|
262
|
Lin Z, Lu S, Xie X, Yi X, Huang H. Noncoding RNAs in drug-resistant pancreatic cancer: A review. Biomed Pharmacother 2020; 131:110768. [PMID: 33152930 DOI: 10.1016/j.biopha.2020.110768] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is the fourth-leading cause of cancer-related deaths and is expected to be the second-leading cause of cancer-related deaths in Europe and the United States by 2030. The high fatality rate of pancreatic cancer is ascribed to untimely diagnosis, early metastasis and limited responses to both chemotherapy and radiotherapy. Although gemcitabine, 5-fluorouracil and some other drugs can profoundly improve patient prognosis, most pancreatic cancer patients eventually develop drug resistance, leading to poor clinical outcomes. The underlying mechanisms of pancreatic cancer drug resistance are complicated and inconclusive. Interestingly, accumulating evidence has demonstrated that different noncoding RNAs (ncRNAs), such as microRNAs (miRNAs), long noncoding RNAs (lncRNAs) and circular RNAs (circRNAs), play a crucial role in pancreatic cancer resistance to chemotherapy reagents. In this paper, we systematically summarize the molecular mechanism underlying the influence of ncRNAs on the generation and development of drug resistance in pancreatic cancer and discuss the potential role of ncRNAs as prognostic markers and new therapeutic targets for pancreatic cancer.
Collapse
Affiliation(s)
- Zhengjun Lin
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Shiyao Lu
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Xubin Xie
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - Xuyang Yi
- Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China.
| | - He Huang
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha, 410013, Hunan Province, China; State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, School of Pre-Clinical Medicine/ Second Affiliated Hospital, Xinjiang Medical University, Urumqi, Xinjiang, 830011, China.
| |
Collapse
|
263
|
Carrieri FA, Smack C, Siddiqui I, Kleinberg LR, Tran PT. Tumor Treating Fields: At the Crossroads Between Physics and Biology for Cancer Treatment. Front Oncol 2020; 10:575992. [PMID: 33215030 PMCID: PMC7664989 DOI: 10.3389/fonc.2020.575992] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/31/2020] [Indexed: 12/22/2022] Open
Abstract
Despite extraordinary advances that have been achieved in the last few decades, cancer continues to represent a leading cause of mortality worldwide. Lethal cancer types ultimately become refractory to standard of care approaches; thus, novel effective treatment options are desperately needed. Tumor Treating Fields (TTFields) are an innovative non-invasive regional anti-mitotic treatment modality with minimal systemic toxicity. TTFields are low intensity (1-3 V/cm), intermediate frequency (100-300 kHz) alternating electric fields delivered to cancer cells. In patients, TTFields are applied using FDA-approved transducer arrays, orthogonally positioned on the area surrounding the tumor region, with side effects mostly limited to the skin. The precise molecular mechanism of the anti-tumor effects of TTFields is not well-understood, but preclinical research on TTFields suggests it may act during two phases of mitosis: at metaphase, by disrupting the formation of the mitotic spindle, and at cytokinesis, by dielectrophoretic dislocation of intracellular organelles leading to cell death. This review describes the mechanism of action of TTFields and provides an overview of the most important in vitro studies that investigate the disruptive effects of TTFields in different cancer cells, focusing mainly on anti-mitotic roles. Lastly, we summarize completed and ongoing TTFields clinical trials on a variety of solid tumors.
Collapse
Affiliation(s)
- Francesca A. Carrieri
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caleb Smack
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ismaeel Siddiqui
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lawrence R. Kleinberg
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Phuoc T. Tran
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Program in Cancer Invasion and Metastasis, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Program in Cellular and Molecular Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
264
|
Pham D, Deter CJ, Reinard MC, Gibson GA, Kiselyov K, Yu W, Sandulache VC, St. Croix CM, Koide K. Using Ligand-Accelerated Catalysis to Repurpose Fluorogenic Reactions for Platinum or Copper. ACS CENTRAL SCIENCE 2020; 6:1772-1788. [PMID: 33145414 PMCID: PMC7596870 DOI: 10.1021/acscentsci.0c00676] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Indexed: 05/03/2023]
Abstract
The development of a fluorescent probe for a specific metal has required exquisite design, synthesis, and optimization of fluorogenic molecules endowed with chelating moieties with heteroatoms. These probes are generally chelation- or reactivity-based. Catalysis-based fluorescent probes have the potential to be more sensitive; however, catalytic methods with a biocompatible fluorescence turn-on switch are rare. Here, we have exploited ligand-accelerated metal catalysis to repurpose known fluorescent probes for different metals, a new approach in probe development. We used the cleavage of allylic and propargylic ethers as platforms that were previously designed for palladium. After a single experiment that combinatorially examined >800 reactions with two variables (metal and ligand) for each ether, we discovered a platinum- or copper-selective method with the ligand effect of specific phosphines. Both metal-ligand systems were previously unknown and afforded strong signals owing to catalytic turnover. The fluorometric technologies were applied to geological, pharmaceutical, serum, and live cell samples and were used to discover that platinum accumulates in lysosomes in cisplatin-resistant cells in a manner that appears to be independent of copper distribution. The use of ligand-accelerated catalysis may present a new blueprint for engineering metal selectivity in probe development.
Collapse
Affiliation(s)
- Dianne Pham
- Department
of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Carly J. Deter
- Department
of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Mariah C. Reinard
- Department
of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Gregory A. Gibson
- Department
of Cell Biology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| | - Kirill Kiselyov
- Department
of Biological Sciences, University of Pittsburgh, 4249 Fifth Avenue, Pittsburgh, Pennsylvania 15260, United States
| | - Wangjie Yu
- Bobby
R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Vlad C. Sandulache
- Bobby
R. Alford Department of Otolaryngology-Head and Neck Surgery, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Claudette M. St. Croix
- Department
of Cell Biology, University of Pittsburgh, 3500 Terrace Street, Pittsburgh, Pennsylvania 15261, United States
| | - Kazunori Koide
- Department
of Chemistry, University of Pittsburgh, 219 Parkman Avenue, Pittsburgh, Pennsylvania 15260, United States
| |
Collapse
|
265
|
Coates JTT, Rodriguez-Berriguete G, Puliyadi R, Ashton T, Prevo R, Wing A, Granata G, Pirovano G, McKenna GW, Higgins GS. The anti-malarial drug atovaquone potentiates platinum-mediated cancer cell death by increasing oxidative stress. Cell Death Discov 2020; 6:110. [PMID: 33133645 PMCID: PMC7591508 DOI: 10.1038/s41420-020-00343-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 10/02/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023] Open
Abstract
Platinum chemotherapies are highly effective cytotoxic agents but often induce resistance when used as monotherapies. Combinatorial strategies limit this risk and provide effective treatment options for many cancers. Here, we repurpose atovaquone (ATQ), a well-tolerated & FDA-approved anti-malarial agent by demonstrating that it potentiates cancer cell death of a subset of platinums. We show that ATQ in combination with carboplatin or cisplatin induces striking and repeatable concentration- and time-dependent cell death sensitization in vitro across a variety of cancer cell lines. ATQ induces mitochondrial reactive oxygen species (mROS), depleting intracellular glutathione (GSH) pools in a concentration-dependent manner. The superoxide dismutase mimetic MnTBAP rescues ATQ-induced mROS production and pre-loading cells with the GSH prodrug N-acetyl cysteine (NAC) abrogates the sensitization. Together, these findings implicate ATQ-induced oxidative stress as key mediator of the sensitizing effect. At physiologically achievable concentrations, ATQ and carboplatin furthermore synergistically delay the growth of three-dimensional avascular spheroids. Clinically, ATQ is a safe and specific inhibitor of the electron transport chain (ETC) and is concurrently being repurposed as a candidate tumor hypoxia modifier. Together, these findings suggest that ATQ is deserving of further study as a candidate platinum sensitizing agent.
Collapse
Affiliation(s)
| | | | - Rathi Puliyadi
- Department of Oncology, University of Oxford, Oxford, UK
| | - Thomas Ashton
- Department of Oncology, University of Oxford, Oxford, UK
| | - Remko Prevo
- Department of Oncology, University of Oxford, Oxford, UK
| | - Archie Wing
- Department of Oncology, University of Oxford, Oxford, UK
| | | | | | | | | |
Collapse
|
266
|
Harguindey S, Alfarouk K, Polo Orozco J, Fais S, Devesa J. Towards an Integral Therapeutic Protocol for Breast Cancer Based upon the New H +-Centered Anticancer Paradigm of the Late Post-Warburg Era. Int J Mol Sci 2020; 21:E7475. [PMID: 33050492 PMCID: PMC7589677 DOI: 10.3390/ijms21207475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/13/2022] Open
Abstract
A brand new approach to the understanding of breast cancer (BC) is urgently needed. In this contribution, the etiology, pathogenesis, and treatment of this disease is approached from the new pH-centric anticancer paradigm. Only this unitarian perspective, based upon the hydrogen ion (H+) dynamics of cancer, allows for the understanding and integration of the many dualisms, confusions, and paradoxes of the disease. The new H+-related, wide-ranging model can embrace, from a unique perspective, the many aspects of the disease and, at the same time, therapeutically interfere with most, if not all, of the hallmarks of cancer known to date. The pH-related armamentarium available for the treatment of BC reviewed here may be beneficial for all types and stages of the disease. In this vein, we have attempted a megasynthesis of traditional and new knowledge in the different areas of breast cancer research and treatment based upon the wide-ranging approach afforded by the hydrogen ion dynamics of cancer. The concerted utilization of the pH-related drugs that are available nowadays for the treatment of breast cancer is advanced.
Collapse
Affiliation(s)
- Salvador Harguindey
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Department of Pharmacology, Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah 42316, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Department of Oncology, Institute of Clinical Biology and Metabolism, 01004 Vitoria, Spain;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, 15886 Teo, Spain;
| |
Collapse
|
267
|
Afifah NN, Diantini A, Intania R, Abdulah R, Barliana MI. Genetic Polymorphisms and the Efficacy of Platinum-Based Chemotherapy: Review. Pharmgenomics Pers Med 2020; 13:427-444. [PMID: 33116759 PMCID: PMC7549502 DOI: 10.2147/pgpm.s267625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 09/07/2020] [Indexed: 11/23/2022] Open
Abstract
Previous studies have indicated that genetic variations in individuals may result in changes in gene expression and amino acids. The effect of these changes may lead to different responses to platinum-based chemotherapy. A vast response rate interval and a short survival rate indicate that the efficacy and efficiency of the selection of chemotherapy have not been optimized. This article aims to illustrate the potential relationship of various genetic polymorphisms in response to platinum-based chemotherapy for several types of cancer. This review was conducted using articles from the last three- and five-year periods (2014-2019) that use gene polymorphism and its relationship to the efficacy of platinum-based chemotherapy as their theme. A total of 26 out of 488 relevant articles were included based on specific criteria. Through various mechanisms, genes, including ERCC1, ERCC2/XPD, XPC, XPA, XRCC1, APE-1, PARP1, OGG1, ABCC2, MRP, GSTP1, GSTM1, GSTT1, MATE1, and OCT2, have been associated with patient response to platinum-based chemotherapy. We conclude that genetic polymorphism analysis is recommended for the management of cancer so that each patient can be administered therapy based on his or her genetic profile to achieve an effective and efficient outcome.
Collapse
Affiliation(s)
- Nadiya Nurul Afifah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| | - Ajeng Diantini
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Ruri Intania
- Dr. H.A. Rotinsulu Lung Hospital, Bandung, Indonesia
| | - Rizky Abdulah
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
| | - Melisa I Barliana
- Center of Excellence in Higher Education for Pharmaceutical Care Innovation, Universitas Padjadjaran, Bandung, Indonesia
- Department of Biological Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Bandung, Indonesia
| |
Collapse
|
268
|
Ma Y, Han J, Jiang J, Zheng Z, Tan Y, Liu C, Zhao Y. Ultrasound targeting of microbubble-bound anti PD-L1 mAb to enhance anti-tumor effect of cisplatin in cervical cancer xenografts treatment. Life Sci 2020; 262:118565. [PMID: 33038371 DOI: 10.1016/j.lfs.2020.118565] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 09/30/2020] [Accepted: 10/01/2020] [Indexed: 12/18/2022]
Abstract
AIMS Anti-PD-L1 monoclonal antibody (mAb)-conjugated ultrasound (US) lipid-shelled microbubbles (PD-L1-MBs) were successfully synthesized to investigate whether that PD-L1-MBs could enhance anti-tumor effect in combination therapy with cisplatin (CDDP) under ultrasound mediation. MAIN METHODS Based on affinity between biotin and streptavidin, we prepared microbubbles conjugated with anti-PD-L1 mAb by membrane hydration and mechanical oscillation. A subcutaneous tumor model was established to test the anti-tumor effect and immunological activity of this combination therapy. Bax and Bcl-2 expression were detected by RT-qPCR and Immunohistochemistry. Cells undergoing apoptosis in tissue section were determined by TUNEL. Proliferation of splenocytes was analyzed by Flow cytometry. A cytotoxic T lymphocyte assay was performed by CTL. Expression of PD-L1 and CD8 in tissue section was examined by immunologfluorescence. Expression of IFN-γ, TNF-α, CD86 and CD80 was also detected by RT-qPCR. KEY FINDINGS We observed that the growth of the subcutaneous tumor was significantly slower in combined group than that in the group treated with either drug or microbubbles. Moreover, higher antitumor activity was observed in the combined group than that in cisplatin alone, which could be reflected by the number of apoptotic cells in tumor tissues and over expression of bax in the combined group. This combination treatment also exhibited a better immunological activity, increasing the infiltration of CD8+ T cells and the expression of several revelant cytokines. SIGNIFICANCE The ultrasound lipid-shelled PD-L1-MBs may enhance anti-tumor effects of cisplatin by blocking the PD-L1 site and improving immune function.
Collapse
Affiliation(s)
- Yao Ma
- Medical College of China Three Gorges University, Yichang, China; Department of Ultrasonography, The First College of Clinical Medical Science, China Three Gorges University & Yichang Central People's Hospital, Yichang, China
| | - Jiaxuan Han
- Medical College of China Three Gorges University, Yichang, China
| | - Jinjun Jiang
- Medical College of China Three Gorges University, Yichang, China
| | - Zhiwei Zheng
- Medical College of China Three Gorges University, Yichang, China
| | - Yandi Tan
- Medical College of China Three Gorges University, Yichang, China
| | - Chaoqi Liu
- Medical College of China Three Gorges University, Yichang, China; Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, Yichang, China.
| | - Yun Zhao
- Medical College of China Three Gorges University, Yichang, China.
| |
Collapse
|
269
|
Synthesis, Antiproliferative Activity, and DNA Binding Studies of Nucleoamino Acid-Containing Pt(II) Complexes. Pharmaceuticals (Basel) 2020; 13:ph13100284. [PMID: 33007911 PMCID: PMC7600948 DOI: 10.3390/ph13100284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/25/2020] [Accepted: 09/28/2020] [Indexed: 11/17/2022] Open
Abstract
We here report our studies on the reaction with the platinum(II) ion of a nucleoamino acid constituted by the l-2,3-diaminopropanoic acid linked to the thymine nucleobase through a methylenecarbonyl linker. The obtained new platinum complexes, characterized by spectroscopic and mass spectrometric techniques, were envisaged to exploit synergistic effects due to the presence of both the platinum center and the nucleoamino acid moiety. The latter can be potentially useful to protect the complexes from early deactivation, as well as to facilitate their cell internalization. The biological activity of the complexes in terms of antiproliferative effects was evaluated in vitro on different cancer cell lines and healthy cells, showing the best results on human cervical adenocarcinoma (HeLa) cells along with good selectivity for cancer over normal cells. In contrast, the metal-free nucleoamino acid did not show any cytotoxicity on both normal and cancer cell lines. Finally, the ability of the novel Pt(II) complexes to bind various DNA model systems was investigated by circular dichroism (CD) spectroscopy and polyacrylamide gel electrophoresis analyses proving that the newly obtained compounds can potentially target DNA, similarly to other well-known anticancer Pt complexes, with a peculiar G-quadruplex vs. duplex selectivity.
Collapse
|
270
|
Mertens RT, Parkin S, Awuah SG. Cancer cell-selective modulation of mitochondrial respiration and metabolism by potent organogold(iii) dithiocarbamates. Chem Sci 2020; 11:10465-10482. [PMID: 34094305 PMCID: PMC8162438 DOI: 10.1039/d0sc03628e] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/04/2020] [Indexed: 12/28/2022] Open
Abstract
Metabolic reprogramming is a key cancer hallmark that has led to the therapeutic targeting of glycolysis. However, agents that target dysfunctional mitochondrial respiration for targeted therapy remains underexplored. We report the synthesis and characterization of ten (10) novel, highly potent organometallic gold(iii) complexes supported by dithiocarbamate ligands as selective inhibitors of mitochondrial respiration. The structure of dithiocarbamates employed dictates the biological stability and cellular cytotoxicity. Most of the compounds exhibit 50% inhibitory concentration (IC50) in the low-micromolar (0.50-2.9 μM) range when tested in a panel of aggressive cancer types with significant selectivity for cancer cells over normal cells. Consequently, there is great interest in the mechanism of action of gold chemotherapeutics, particularly, considering that DNA is not the major target of most gold complexes. We investigate the mechanism of action of representative complexes, 1a and 2a in the recalcitrant triple negative breast cancer (TNBC) cell line, MDA-MB-231. Whole-cell transcriptomics sequencing revealed genes related to three major pathways, namely: cell cycle, organelle fission, and oxidative phosphorylation. 2a irreversibly and rapidly inhibits maximal respiration in TNBC with no effect on normal epithelial cells, implicating mitochondrial OXPHOS as a potential target. Furthermore, the modulation of cyclin dependent kinases and G1 cell cycle arrest induced by these compounds is promising for the treatment of cancer. This work contributes to the need for mitochondrial respiration modulators in biomedical research and outlines a systematic approach to study the mechanism of action of metal-based agents.
Collapse
Affiliation(s)
- Randall T Mertens
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Sean Parkin
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
| | - Samuel G Awuah
- Department of Chemistry, University of Kentucky Lexington KY 40506 USA
- Center for Pharmaceutical Research and Innovation, College of Pharmacy and Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky Lexington Kentucky 40536 USA
| |
Collapse
|
271
|
Saleh DO, Mansour DF, Mostafa RE. Rosuvastatin and simvastatin attenuate cisplatin-induced cardiotoxicity via disruption of endoplasmic reticulum stress-mediated apoptotic death in rats: targeting ER-Chaperone GRP78 and Calpain-1 pathways. Toxicol Rep 2020; 7:1178-1186. [PMID: 32995293 PMCID: PMC7501485 DOI: 10.1016/j.toxrep.2020.08.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/29/2020] [Accepted: 08/11/2020] [Indexed: 12/18/2022] Open
Abstract
Cisplatin (CP) is a powerful antineoplastic chemotherapeutic agent with broad-spectrum properties. Acute and cumulative cardiotoxicity are major limiting factors for CP therapy. Various pathogenic pathways have been suggested to CP-induced cardiotoxicity; oxidative damage, ER stress, and programmed cell death/apoptosis. The present study aimed to assess the signaling mechanisms related to the advantageous effects of rosuvastatin (RSV) and simvastatin (SMV) against CP-related cardiac ER stress dependent apoptotic death in rats. Acute cardiotoxicity was induced by a single dose of CP (10 mg/kg, i.p.) on the 10th day of the experiment. RSV (10 mg/ kg/day) and SMV (10 mg/kg/day) were orally administered for 15 days. CP-treated rats showed significant alterations in electrocardiographic recordings and elevation in serum cardiac function biomarkers; troponin T content, lactate dehydrogenase and creatine kinase-MB levels as well as boost in the cardiac oxidative stress biomarkers. In addition, CP exposure resulted in GRP78 induction; an ER stress and elevation marker at calpain-1 content as well as activation of activated caspase-3 (ACASP3) and caspase-12 were reflected on CP-triggered apoptosis evidenced by elevation in the Bax/Bcl-2 ratio. However, RSV and SMV administration mitigate those adverse CP effects. Statins administration prominently alleviated CP-induced cardiac abnormalities exerting improvement in the ECG pattern and cardiac enzyme biomarkers. Interestingly, statins; RSV and SMV, disrupted CP-induced ER stress and the consequent apoptotic cell death evidenced by downregulation of ER-chaperone GRP78, calpain-1, ACASP3 and caspase-12 as well as decline in the Bax/Bcl-2 ratio. From all the previous findings, it can be suggested that statins namely; RSV and SMV, play protective role against CP-induced cardiac injury by regulating ER stress-mediated apoptotic pathways.
Collapse
Affiliation(s)
- Dalia O Saleh
- Department of Pharmacology, National Research Centre (ID: 60014618), 33 El Buhouth st-Dokki P.O:12622, Cairo, Egypt
| | - Dina F Mansour
- Department of Pharmacology, National Research Centre (ID: 60014618), 33 El Buhouth st-Dokki P.O:12622, Cairo, Egypt
| | - Rasha E Mostafa
- Department of Pharmacology, National Research Centre (ID: 60014618), 33 El Buhouth st-Dokki P.O:12622, Cairo, Egypt
| |
Collapse
|
272
|
Sayed A, Munir M, Eweis N, Wael D, Shazly O, Awad AK, Elbadawy MA, Eissa S. An overview on precision therapy in bladder cancer. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2020. [DOI: 10.1080/23808993.2020.1801346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- Ahmed Sayed
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Malak Munir
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Noor Eweis
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Doaa Wael
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Omar Shazly
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Ahmed K. Awad
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Marihan A. Elbadawy
- Faculty of Medicine, Undergraduate Medical Students, Ain Shams University, Cairo, Egypt
| | - Sanaa Eissa
- Faculty of Medicine, Professor of Medical Biochemistry and Molecular Biology, Ain Shams University, Cairo, Egypt
| |
Collapse
|
273
|
Eremina JA, Lider EV, Sukhikh TS, Klyushova LS, Perepechaeva ML, Sheven' DG, Berezin AS, Grishanova AY, Potkin VI. Water-soluble copper(II) complexes with 4,5-dichloro-isothiazole-3-carboxylic acid and heterocyclic N-donor ligands: Synthesis, crystal structures, cytotoxicity, and DNA binding study. Inorganica Chim Acta 2020. [DOI: 10.1016/j.ica.2020.119778] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
274
|
Lukanović D, Herzog M, Kobal B, Černe K. The contribution of copper efflux transporters ATP7A and ATP7B to chemoresistance and personalized medicine in ovarian cancer. Biomed Pharmacother 2020; 129:110401. [DOI: 10.1016/j.biopha.2020.110401] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/23/2020] [Accepted: 06/13/2020] [Indexed: 02/08/2023] Open
|
275
|
Murray D, Mirzayans R. Cellular Responses to Platinum-Based Anticancer Drugs and UVC: Role of p53 and Implications for Cancer Therapy. Int J Mol Sci 2020; 21:ijms21165766. [PMID: 32796711 PMCID: PMC7461110 DOI: 10.3390/ijms21165766] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 12/16/2022] Open
Abstract
Chemotherapy is intended to induce cancer cell death through apoptosis and other avenues. Unfortunately, as discussed in this article, moderate doses of genotoxic drugs such as cisplatin typical of those achieved in the clinic often invoke a cytostatic/dormancy rather than cytotoxic/apoptosis response in solid tumour-derived cell lines. This is commonly manifested by an extended apoptotic threshold, with extensive apoptosis only being seen after very high/supralethal doses of such agents. The dormancy response can be associated with senescence-like features, polyploidy and/or multinucleation, depending in part on the p53 status of the cells. In most solid tumour-derived cells, dormancy represents a long-term survival mechanism, ultimately contributing to disease recurrence. This review highlights the nonlinearity of key aspects of the molecular and cellular responses to bulky DNA lesions in human cells treated with chemotherapeutic drugs (e.g., cisplatin) or ultraviolet light-C (a widely used tool for unraveling details of the DNA damage-response) as a function of the level of genotoxic stress. Such data highlight the growing realization that targeting dormant cancer cells, which frequently emerge following conventional anticancer treatments, may represent a novel strategy to prevent or, at least, significantly suppress cancer recurrence.
Collapse
|
276
|
McMullen M, Madariaga A, Lheureux S. New approaches for targeting platinum-resistant ovarian cancer. Semin Cancer Biol 2020; 77:167-181. [DOI: 10.1016/j.semcancer.2020.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 08/15/2020] [Accepted: 08/25/2020] [Indexed: 12/12/2022]
|
277
|
Ng KTP, Yeung OWH, Liu J, Li CX, Liu H, Liu XB, Qi X, Ma YY, Lam YF, Lau MY, Qiu WQ, Shiu HC, Lai MK, Lo CM, Man K. Clinical significance and functional role of transmembrane protein 47 (TMEM47) in chemoresistance of hepatocellular carcinoma. Int J Oncol 2020; 57:956-966. [PMID: 32945373 PMCID: PMC7473756 DOI: 10.3892/ijo.2020.5104] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 07/13/2020] [Indexed: 12/15/2022] Open
Abstract
Chemoresistance is the main cause of chemotherapy failure in patients with hepatocellular carcinoma (HCC). The gene encoding transmembrane protein 47 (TMEM47) was previously identified to be significantly upregulated in HCC cell lines with acquired chemoresistance. The aim of the present study was to characterize the clinical significance and function of TMEM47 in HCC chemoresistance. The results demonstrated that the TMEM47 expression levels in the tumors of patients not responding to cisplatin-based transarterial chemoembolization (TACE) treatment was significantly higher compared with those in patients who responded to TACE treatment. Moreover, analyses from clinical samples and HCC cell lines indicated that TMEM47 expression may be upregulated in HCC in response to cisplatin treatment. Furthermore, the TMEM47 mRNA expression levels were positively correlated with the degree of cisplatin resistance of HCC cells. Overexpression of TMEM47 in HCC cells significantly promoted cisplatin resistance. The present study also demonstrated that targeted inhibition of TMEM47 could significantly reduce cisplatin resistance of cisplatin-resistant HCC cells via enhancing caspase-mediated apoptosis. In addition, targeted inhibition of TMEM47 enhanced the sensitivity of cisplatin-resistant cells to cisplatin via suppressing cisplatin-induced activation of the genes involved in drug efflux and metabolism. The present study also validated that TMEM47 expression was significantly correlated with multi-drug resistance-associated protein 1 in patients with HCC who received TACE treatment. In conclusion, the findings of the present study demonstrated that TMEM47 may be a useful biomarker for predicting the response to chemotherapy and a potential therapeutic target for overcoming HCC chemoresistance.
Collapse
Affiliation(s)
- Kevin Tak-Pan Ng
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Oscar Wai-Ho Yeung
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Jiang Liu
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Chang Xian Li
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Hui Liu
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Xiao Bing Liu
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Xiang Qi
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yuen Yuen Ma
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Yin Fan Lam
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Matthew Yh Lau
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Wen Qi Qiu
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Hoi Chung Shiu
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Man Kit Lai
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| | - Kwan Man
- Department of Surgery, The University of Hong Kong Shenzhen Hospital and LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, SAR, P.R. China
| |
Collapse
|
278
|
Acquisition of Cisplatin Resistance Shifts Head and Neck Squamous Cell Carcinoma Metabolism toward Neutralization of Oxidative Stress. Cancers (Basel) 2020; 12:cancers12061670. [PMID: 32599707 PMCID: PMC7352569 DOI: 10.3390/cancers12061670] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/18/2020] [Accepted: 06/20/2020] [Indexed: 12/24/2022] Open
Abstract
Background: Cisplatin (CDDP) is commonly utilized in the treatment of advanced solid tumors including head and neck squamous cell carcinoma (HNSCC). Cisplatin response remains highly variable among individual tumors and development of cisplatin resistance is common. We hypothesized that development of cisplatin resistance is partially driven by metabolic reprogramming. Methods: Using a pre-clinical HNSCC model and an integrated approach to steady state metabolomics, metabolic flux and gene expression data we characterized the interaction between cisplatin resistance and metabolic reprogramming. Results: Cisplatin toxicity in HNSCC was driven by generation of intra-cellular oxidative stress. This was validated by demonstrating that acquisition of cisplatin resistance generates cross-resistance to ferroptosis agonists despite the fact that cisplatin itself does not trigger ferroptosis. Acquisition of cisplatin resistance dysregulated the expression of genes involved in amino acid, fatty acid metabolism and central carbon catabolic pathways, enhanced glucose catabolism and serine synthesis. Acute cisplatin exposure increased intra-tumoral levels of S-methyl-5-thiadenosine (MTA) precursors and metabotoxins indicative of generalized oxidative stress. Conclusions: Acquisition of cisplatin resistance is linked to metabolic recovery from oxidative stress. Although this portends poor effectiveness for directed metabolic targeting, it supports the potential for biomarker development of cisplatin effectiveness using an integrated approach.
Collapse
|
279
|
Greenhalgh CJ, Karekla E, Miles GJ, Powley IR, Costa C, de Jesus J, Bailey MJ, Pritchard C, MacFarlane M, Pringle JH, Managh AJ. Exploration of Matrix Effects in Laser Ablation Inductively Coupled Plasma Mass Spectrometry Imaging of Cisplatin-Treated Tumors. Anal Chem 2020; 92:9847-9855. [DOI: 10.1021/acs.analchem.0c01347] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Calum J. Greenhalgh
- Department of Chemistry, Loughborough University, Loughborough, Leicestershire LE11 3TU, U.K
| | - Ellie Karekla
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Gareth J. Miles
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Ian R. Powley
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Catia Costa
- Ion Beam Centre, University of Surrey, Guildford GU2 7XH, U.K
| | - Janella de Jesus
- Department of Chemistry, University of Surrey, Guildford GU2 7XH, U.K
| | - Melanie J. Bailey
- Department of Chemistry, University of Surrey, Guildford GU2 7XH, U.K
| | - Catrin Pritchard
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | | | - J. Howard Pringle
- Leicester Cancer Research Centre, University of Leicester, Robert Kilpatrick Building, Leicester Royal Infirmary, Leicester LE2 7LX, U.K
| | - Amy J. Managh
- Department of Chemistry, Loughborough University, Loughborough, Leicestershire LE11 3TU, U.K
| |
Collapse
|
280
|
Zampieri LX, Grasso D, Bouzin C, Brusa D, Rossignol R, Sonveaux P. Mitochondria Participate in Chemoresistance to Cisplatin in Human Ovarian Cancer Cells. Mol Cancer Res 2020; 18:1379-1391. [PMID: 32471883 DOI: 10.1158/1541-7786.mcr-19-1145] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/16/2020] [Accepted: 05/21/2020] [Indexed: 11/16/2022]
Abstract
Ovarian cancer is an aggressive disease that affects about 300,000 patients worldwide, with a yearly death count of about 185,000. Following surgery, treatment involves adjuvant or neoadjuvant administration of taxane with platinum compounds cisplatin or carboplatin, which alkylate DNA through the same chemical intermediates. However, although platinum-based therapy can cure patients in a number of cases, a majority of them discontinues treatment owing to side effects and to the emergence of resistance. In this study, we focused on resistance to cisplatin and investigated whether metabolic changes could be involved. As models, we used matched pairs of cisplatin-sensitive (SKOV-3 and COV-362) and cisplatin-resistant (SKOV-3-R and COV-362-R) human ovarian carcinoma cells that were selected in vitro following exposure to increasing doses of the chemotherapy. Metabolic comparison revealed that resistant cells undergo a shift toward a more oxidative metabolism. The shift goes along with a reorganization of the mitochondrial network, with a generally increased mitochondrial compartment. More functional mitochondria in cisplatin-resistant compared with cisplatin-sensitive cells were associated to enzymatic changes affecting either the electron transport chain (SKOV-3/SKOV-3-R model) or mitochondrial coupling (COV-362/COV-362-R model). Our findings further indicate that the preservation of functional mitochondria in these cells could be due to an increased mitochondrial turnover rate, suggesting mitophagy inhibition as a potential strategy to tackle cisplatin-resistant human ovarian cancer progression. IMPLICATIONS: Besides classical mechanisms related to drug efflux and target modification, we report that preserving functional mitochondria is a strategy used by human ovarian cancer cells to resist to cisplatin chemotherapy.
Collapse
Affiliation(s)
- Luca X Zampieri
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Debora Grasso
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Caroline Bouzin
- IREC imaging platform (2IP), Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Davide Brusa
- IREC Flow Cytometry and Cell Sorting Platform, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium
| | | | - Pierre Sonveaux
- Pole of Pharmacology and Therapeutics, Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCLouvain), Brussels, Belgium.
| |
Collapse
|
281
|
Querino ALDA, Enes KB, Chaves OA, Dittz D, Couri MRC, Diniz R, Silva H. Modified pyrazole platinum(II) complex can circumvent albumin and glutathione: Synthesis, structure and cytotoxic activity. Bioorg Chem 2020; 100:103936. [PMID: 32438131 DOI: 10.1016/j.bioorg.2020.103936] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 12/18/2022]
Abstract
The synthesis and structural characterization of novel platinum complexes ([PtII(Pz)2Cl2] - C1, C2 and C3) featuring diphenyl-pyrazole derived ligands: para-fluorophenyl and para-substituted phenyl (CH3, F and Cl for L1, L2 and L3, respectively) were reported and it was also evaluated their potential antitumor activity. The elemental, molar conductivity and thermogravimetric analysis combined with FTIR, UV-vis, NMR and mass spectrometry are in agreement with the chemical structure indicated by single-crystal X-ray diffraction. The antiproliferative activities were assessed against tumor (B16F10 and 4T1) and non-tumor (BHK21) cell lines, and the cytotoxicity of the compounds was strongly increased after metal complexation displaying promising activity. It was also assessed the ability of extracellular bovine serum albumin (BSA) and glutathione (GSH) to decrease the cytotoxicity of the complexes against B16F10. It was highlighted that only the C3 activity was not disturbed in those conditions, being confirmed by flow cytometry using Anexin-V/PI to evaluate interferences in the apoptosis process, even it was not predicted by molecular docking simulations. The interaction of the synthesized compounds with calf-thymus DNA (ctDNA) and bovine serum albumin (BSA) was also investigated through spectrophotometric assays and molecular docking simulations, indicating that C1 and C2 presented better interaction with the biomacromolecules than the corresponding ligands. In addition, agarose gel electrophoresis with plasmid DNA revealed that C1-C3 are capable of interaction with DNA and modify its electrophoretic mobility.
Collapse
Affiliation(s)
| | - Karine Braga Enes
- Department of Chemistry, Universidade Federal de Juiz de Fora, 36036-900 Juiz de Fora, MG, Brazil
| | - Otávio Augusto Chaves
- Senai Innovation Institute for Green Chemistry, 20271-030 Rio de Janeiro, RJ, Brazil
| | - Dalton Dittz
- Department of Biochemistry and Pharmacology, Universidade Federal do Piauí, 64049-550 Terezina, PI, Brazil
| | - Mara Rubia Costa Couri
- Department of Chemistry, Universidade Federal de Juiz de Fora, 36036-900 Juiz de Fora, MG, Brazil
| | - Renata Diniz
- Department of Chemistry, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil
| | - Heveline Silva
- Department of Chemistry, Universidade Federal de Minas Gerais, 31270-901 Belo Horizonte, MG, Brazil.
| |
Collapse
|
282
|
He Y, Chen D, Yi Y, Zeng S, Liu S, Li P, Xie H, Yu P, Jiang G, Liu H. Histone Deacetylase Inhibitor Sensitizes ERCC1-High Non-small-Cell Lung Cancer Cells to Cisplatin via Regulating miR-149. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:448-459. [PMID: 32478168 PMCID: PMC7251316 DOI: 10.1016/j.omto.2020.05.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 05/01/2020] [Indexed: 02/06/2023]
Abstract
Resistance to platinum-based chemotherapy becomes a major obstacle in non-small-cell lung cancer (NSCLC) treatment. Overexpression of the excision repair cross-complementing 1 (ERCC1) gene is reported to negatively influence the effectiveness of cisplatin-based therapy for NSCLC cells. In this study, we confirm that high ERCC1 expression correlates with cisplatin resistance in NSCLC cells. Importantly, histone deacetylase inhibitors (HDACis) re-sensitize ERCC1-high NSCLC cells to cisplatin both in vitro and in vivo. Mechanistically, the HDACi induces the expression of miR-149 by acetylation and activation of E2F1, which directly targets ERCC1 and inhibits ERCC1 expression. Inhibition of miR-149 reverses the promotion effect of HDACis on cisplatin-induced DNA damage and cell apoptosis in ERCC1-high NSCLC cells. In conclusion, this study reveals a novel mechanism by which HDACis re-sensitizes ERCC1-high NSCLC cells to cisplatin via regulation of the E2F1/miR-149/ERCC1 axis, and we propose that combination of HDACis and cisplatin might hold promise to be a more effective therapeutic paradigm for ERCC1-high NSCLCs.
Collapse
Affiliation(s)
- Yuwen He
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Danyang Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Yanmei Yi
- Department of Histology and Embryology, Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Shanshan Zeng
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Shuang Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Pan Li
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
| | - Hui Xie
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Pengjiu Yu
- Department of Pharmacy, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China
| | - Guanmin Jiang
- Department of Clinical Laboratory, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 528000, Guangdong, China
- Corresponding author: Guanmin Jiang, Department of Clinical Laboratory, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai 528000, Guangdong, China.
| | - Hao Liu
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” Guangzhou 510095, Guangdong, China
- Corresponding author: Hao Liu, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou Key Laboratory of “Translational Medicine on Malignant Tumor Treatment,” No. 78 Engzhigang Road, Guangzhou 510095, Guangdong, China.
| |
Collapse
|
283
|
Sanz Del Olmo N, Bajo AM, Ionov M, García-Gallego S, Bryszewska M, Gómez R, Ortega P, de la Mata FJ. Cyclopentadienyl ruthenium(II) carbosilane metallodendrimers as a promising treatment against advanced prostate cancer. Eur J Med Chem 2020; 199:112414. [PMID: 32438200 DOI: 10.1016/j.ejmech.2020.112414] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/16/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022]
Abstract
In searching for efficient and selective antitumour drugs, a new family of carbosilane metallodendrimers functionalized with [Ru(η5-C5H5)(PTA)Cl] (PTA = 1,3,5-triaza-7-phosphatricyclo-[3.3.1.1] decane) is reported. Experiments of the biophysical characterization showed an ability to interact with biological membranes, as well as with proteins (e.g. human serum albumin) without affecting their usual biological activity. These metallodendrimers possessed potent and selective anticancer activity in vitro in a panel of tumour cell lines. Importantly, the first generation metallodendrimer, bearing 4 Ru(II) complexes, was remarkably active towards resistant prostate cancer cells, inhibiting both cell proliferation and metastasis to bone tissues. Such promising antitumour activity can be further improved when given with docetaxel, with in vitro cytotoxicity being in the nanomolar range. Furthermore, its intravenous administration to an advanced prostate cancer mice model inhibited tumour growth up to 25% and 45% when given 10 mg/kg/week and 7.5 mg/kg/4-5 days, respectively.
Collapse
Affiliation(s)
- Natalia Sanz Del Olmo
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, Madrid, Spain
| | - Ana M Bajo
- Department of Biology of Systems, Biochemistry and Molecular Biology Unit, University of Alcalá, Madrid, Spain
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Sandra García-Gallego
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, Madrid, Spain
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236, Lodz, Poland
| | - Rafael Gómez
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain and Institute "Ramón y Cajal" for Health Research (IRYCIS), Spain
| | - Paula Ortega
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain and Institute "Ramón y Cajal" for Health Research (IRYCIS), Spain.
| | - F Javier de la Mata
- Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), University of Alcalá, Madrid, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain and Institute "Ramón y Cajal" for Health Research (IRYCIS), Spain.
| |
Collapse
|
284
|
Huang H, Cao S, Zhang Z, Li L, Chen F, Wu Q. Multiple omics analysis of the protective effects of SFN on estrogen-dependent breast cancer cells. Mol Biol Rep 2020; 47:3331-3346. [DOI: 10.1007/s11033-020-05403-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/25/2020] [Indexed: 12/14/2022]
|
285
|
Delmas D, Xiao J, Vejux A, Aires V. Silymarin and Cancer: A Dual Strategy in Both in Chemoprevention and Chemosensitivity. Molecules 2020; 25:2009. [PMID: 32344919 PMCID: PMC7248929 DOI: 10.3390/molecules25092009] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 04/09/2020] [Accepted: 04/21/2020] [Indexed: 02/08/2023] Open
Abstract
Silymarin extracted from milk thistle consisting of flavonolignan silybin has shown chemopreventive and chemosensitizing activity against various cancers. The present review summarizes the current knowledge on the potential targets of silymarin against various cancers. Silymarin may play on the system of xenobiotics, metabolizing enzymes (phase I and phase II) to protect normal cells against various toxic molecules or to protect against deleterious effects of chemotherapeutic agents on normal cells. Furthermore, silymarin and its main bioactive compounds inhibit organic anion transporters (OAT) and ATP-binding cassettes (ABC) transporters, thus contributing to counteracting potential chemoresistance. Silymarin and its derivatives play a double role, namely, limiting the progression of cancer cells through different phases of the cycle-thus forcing them to evolve towards a process of cell death-and accumulating cancer cells in a phase of the cell cycle-thus making it possible to target a greater number of tumor cells with a specific anticancer agent. Silymarin exerts a chemopreventive effect by inducing intrinsic and extrinsic pathways and reactivating cell death pathways by modulation of the ratio of proapoptotic/antiapoptotic proteins and synergizing with agonists of death domains receptors. In summary, we highlight how silymarin may act as a chemopreventive agent and a chemosensitizer through multiple pathways.
Collapse
Affiliation(s)
- Dominique Delmas
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.V.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Dijon, Bioactive Molecules and Health research group, F-21000 Dijon, France
- Centre anticancéreux Georges François Leclerc Center, F-21000 Dijon, France
| | - Jianbo Xiao
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China;
| | - Anne Vejux
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.V.); (V.A.)
- Laboratoire Bio-PeroxIL“Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism”—EA 7270, UFR Sciences Vie Terre Environnement (SVTE), 6 Bd Gabriel, F-21000 Dijon, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.V.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Dijon, Bioactive Molecules and Health research group, F-21000 Dijon, France
| |
Collapse
|
286
|
Scagliarini A, Mathey A, Aires V, Delmas D. Xanthohumol, a Prenylated Flavonoid from Hops, Induces DNA Damages in Colorectal Cancer Cells and Sensitizes SW480 Cells to the SN38 Chemotherapeutic Agent. Cells 2020; 9:E932. [PMID: 32290112 PMCID: PMC7226974 DOI: 10.3390/cells9040932] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 12/21/2022] Open
Abstract
In spite of chemotherapy and systematic screening for people at risk, the mortality rate of colorectal cancer (CRC) remains consistently high, with 600,000 deaths per year. This low success rate in the treatment of CRC results from many failures associated with high resistance and the risk of metastasis. Therefore, in response to these therapeutic failures, new strategies have been under development for several years aimed at increasing the effect of anticancer compounds and/or at reducing their secondary effects on normal cells, thus enabling the host to better withstand chemotherapy. This study highlights that xanthohumol (Xn) concentrations under the IC50 values were able to induce apoptosis and to enhance the DNA-damage response (DDR). We demonstrate for the first time that Xn exerts its anticancer activity in models of colon cancer through activation of the ataxia telangiectasia mutated (ATM) pathway. Subsequently, the ability of Xn to restore DNA damage in CRC cells can sensitize them to anticancer agents such as SN38 (7-ethyl-10-hydroxycamptothecin) used in chemotherapy.
Collapse
Affiliation(s)
- Alessandra Scagliarini
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Aline Mathey
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (A.S.); (A.M.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, Bioactive Molecules and Health Research Group, F-21000 Dijon, France
- Centre Anticancéreux Georges François Leclerc, F-21000 Dijon, France
| |
Collapse
|
287
|
Doktorova TY, Oki NO, Mohorič T, Exner TE, Hardy B. A semi-automated workflow for adverse outcome pathway hypothesis generation: The use case of non-genotoxic induced hepatocellular carcinoma. Regul Toxicol Pharmacol 2020; 114:104652. [PMID: 32251711 DOI: 10.1016/j.yrtph.2020.104652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Revised: 01/10/2020] [Accepted: 03/29/2020] [Indexed: 02/07/2023]
Abstract
The utility of the Adverse Outcome Pathway (AOP) concept has been largely recognized by scientists, however, the AOP generation is still mainly done manually by screening through evidence and extracting probable associations. To accelerate this process and increase the reliability, we have developed an semi-automated workflow for AOP hypothesis generation. In brief, association mining methods were applied to high-throughput screening, gene expression, in vivo and disease data present in ToxCast and Comparative Toxicogenomics Database. This was supplemented by pathway mapping using Reactome to fill in gaps and identify events occurring at the cellular/tissue levels. Furthermore, in vivo data from TG-Gates was integrated to finally derive a gene, pathway, biochemical, histopathological and disease network from which specific disease sub-networks can be queried. To test the workflow, non-genotoxic-induced hepatocellular carcinoma (HCC) was selected as a case study. The implementation resulted in the identification of several non-genotoxic-specific HCC-connected genes belonging to cell proliferation, endoplasmic reticulum stress and early apoptosis. Biochemical findings revealed non-genotoxic-specific alkaline phosphatase increase. The explored non-genotoxic-specific histopathology was associated with early stages of hepatic steatosis, transforming into cirrhosis. This work illustrates the utility of computationally predicted constructs in supporting development by using pre-existing knowledge in a fast and unbiased manner.
Collapse
Affiliation(s)
- Tatyana Y Doktorova
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, Technology Park Basel, Basel, Switzerland.
| | - Noffisat O Oki
- American Association for the Advancement of Science, Science & Technology Policy Fellow, USA; National Institutes of Health, Rockville, MD, USA
| | - Tomaž Mohorič
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, Technology Park Basel, Basel, Switzerland
| | - Thomas E Exner
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, Technology Park Basel, Basel, Switzerland
| | - Barry Hardy
- Edelweiss Connect GmbH, Hochbergerstrasse 60C, Technology Park Basel, Basel, Switzerland
| |
Collapse
|
288
|
Paucarmayta A, Taitz H, McGlorthan L, Casablanca Y, Maxwell GL, Darcy KM, Syed V. Progesterone-Calcitriol Combination Enhanced Cytotoxicity of Cisplatin in Ovarian and Endometrial Cancer Cells In Vitro. Biomedicines 2020; 8:biomedicines8040073. [PMID: 32244545 PMCID: PMC7236602 DOI: 10.3390/biomedicines8040073] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 03/27/2020] [Accepted: 03/30/2020] [Indexed: 12/22/2022] Open
Abstract
: Initially, patients that respond to cisplatin (DDP) treatment later relapse and develop chemoresistance. Agents that enhance DDP effectiveness will have a significant impact on cancer treatment. We have shown pronounced inhibitory effects of the progesterone-calcitriol combination on endometrial and ovarian cancer cell growth. Here, we examined whether and how progesterone-calcitriol combination potentiates DDP anti-tumor effects in cancer cells. Ovarian and endometrial cancer cells treated with various concentrations of DDP showed a concentration-dependent decrease in cell proliferation. Concurrent treatment of cells with DDP and progesterone-calcitriol ombination potentiated anticancer effects of DDP compared to DDP-calcitriol, or DDP-progesterone treated groups. The anticancer effects were mediated by increased caspase-3, BAX, and decreased BCL2 and PARP-1 expression in DDP and progesterone-calcitriol combination-treated cells. Stimulation of the PI3K/AKT and MAPK/ERK pathways seen in cancer cells was reduced in DDP-progesterone-calcitriol treated cells. Pretreatment of cells with specific inhibitors further diminished AKT and ERK expression. Furthermore, progesterone-calcitriol potentiated the anti-growth effects of DDP on cancer cells by attenuating the expression of SMAD2/3, multidrug resistance protein- 1 (MDR-1), and ABC transporters (ABCG1, and ABCG2), thereby impeding the efflux of chemo drugs from cancer cells. These results suggest a potential clinical benefit of progesterone-calcitriol combination therapy when used in combination with DDP.
Collapse
Affiliation(s)
- Ana Paucarmayta
- Department of Obstetrics & Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (A.P.); (H.T.); (L.M.); (Y.C.); (K.M.D.)
| | - Hannah Taitz
- Department of Obstetrics & Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (A.P.); (H.T.); (L.M.); (Y.C.); (K.M.D.)
| | - Latoya McGlorthan
- Department of Obstetrics & Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (A.P.); (H.T.); (L.M.); (Y.C.); (K.M.D.)
| | - Yovanni Casablanca
- Department of Obstetrics & Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (A.P.); (H.T.); (L.M.); (Y.C.); (K.M.D.)
- Gynecologic Cancer Center of Excellence, Department of Obstetrics and Gynecology, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, USA
- John P. Murtha Cancer Center, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, USA;
- Gynecologic Cancer Center of Excellence, Women’s Health Integrated Research Center at Inova Health System, 3289 Woodburn Road, Suite 370, Annandale, VA 22003, USA
| | - G. Larry Maxwell
- John P. Murtha Cancer Center, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, USA;
- Gynecologic Cancer Center of Excellence, Women’s Health Integrated Research Center at Inova Health System, 3289 Woodburn Road, Suite 370, Annandale, VA 22003, USA
- Inova Fairfax Hospital, Department of Obstetrics & Gynecology, 3300 Gallows Road, Falls Church, VA 22042, USA
| | - Kathleen M. Darcy
- Department of Obstetrics & Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (A.P.); (H.T.); (L.M.); (Y.C.); (K.M.D.)
- John P. Murtha Cancer Center, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, USA;
- Gynecologic Cancer Center of Excellence, Women’s Health Integrated Research Center at Inova Health System, 3289 Woodburn Road, Suite 370, Annandale, VA 22003, USA
| | - Viqar Syed
- Department of Obstetrics & Gynecology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA; (A.P.); (H.T.); (L.M.); (Y.C.); (K.M.D.)
- John P. Murtha Cancer Center, Uniformed Services University of the Health Sciences and Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20889, USA;
- Department of Molecular and Cell Biology, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD 20814, USA
- Correspondence: ; Tel.: +301-295-3128; Fax: +301-295-6774
| |
Collapse
|
289
|
Lehmann DM, Williams WC. Physiological responses to cisplatin using a mouse hypersensitivity model. Inhal Toxicol 2020; 32:68-78. [PMID: 32188332 DOI: 10.1080/08958378.2020.1737762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Background: The physiological mechanisms underlying the development of respiratory hypersensitivity to cisplatin (CDDP) are not well-understood. It has been suggested that these reactions are likely the result of type I hypersensitivity, but other explanations are plausible and the potential for CDDP to induce type I hypersensitivity responses has not been directly evaluated in an animal model. Objectives and Methods: To investigate CDDP hypersensitivity, mice were topically sensitized through application of CDDP before being challenged by oropharyngeal aspiration (OPA) with CDDP. Before and immediately after OPA challenge, pulmonary responses were assessed using whole body plethysmography (WBP). Results: CDDP did not induce an immediate response or alter the respiratory rate in sensitized mice. Two days later, baseline enhanced pause (Penh) values were significantly elevated (p < 0.05) in mice challenged with CDDP. When challenged with methacholine (Mch) aerosol, Penh values were significantly elevated (p < 0.05) in sensitized mice and respiratory rate was reduced (p < 0.05). Lymph node cell counts and immunoglobulin E levels also indicated successful sensitization to CDDP. Irrespective of the sensitization state of the mice, the number of neutrophils increased significantly in bronchoalveolar lavage fluid (BALF) following CDDP challenge. BALF from sensitized mice also contained 2.46 (±0.8) × 104 eosinophils compared to less than 0.48 (±0.2) × 104 cells in non-sensitized mice (p < 0.05). Conclusions: The results from this study indicate that dermal exposure to CDDP induces immunological changes consistent with type I hypersensitivity and that a single respiratory challenge is enough to trigger pulmonary responses in dermally sensitized mice. These data provide previously unknown insights into the mechanisms of CDDP hypersensitivity.
Collapse
Affiliation(s)
- David M Lehmann
- Center for Public Health & Environmental Assessment (CPHEA), US - Environmental Protection Agency, Durham, NC, USA
| | - Wanda C Williams
- Center for Public Health & Environmental Assessment (CPHEA), US - Environmental Protection Agency, Durham, NC, USA
| |
Collapse
|
290
|
Harguindey S, Alfarouk K, Polo Orozco J, Hardonnière K, Stanciu D, Fais S, Devesa J. A New and Integral Approach to the Etiopathogenesis and Treatment of Breast Cancer Based upon Its Hydrogen Ion Dynamics. Int J Mol Sci 2020; 21:E1110. [PMID: 32046158 PMCID: PMC7036897 DOI: 10.3390/ijms21031110] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
Despite all efforts, the treatment of breast cancer (BC) cannot be considered to be a success story. The advances in surgery, chemotherapy and radiotherapy have not been sufficient at all. Indeed, the accumulated experience clearly indicates that new perspectives and non-main stream approaches are needed to better characterize the etiopathogenesis and treatment of this disease. This contribution deals with how the new pH-centric anticancer paradigm plays a fundamental role in reaching a more integral understanding of the etiology, pathogenesis, and treatment of this multifactorial disease. For the first time, the armamentarium available for the treatment of the different types and phases of BC is approached here from a Unitarian perspective-based upon the hydrogen ion dynamics of cancer. The wide-ranged pH-related molecular, biochemical and metabolic model is able to embrace most of the fields and subfields of breast cancer etiopathogenesis and treatment. This single and integrated approach allows advancing towards a unidirectional, concerted and synergistic program of treatment. Further efforts in this line are likely to first improve the therapeutics of each subtype of this tumor and every individual patient in every phase of the disease.
Collapse
Affiliation(s)
- Salvador Harguindey
- Institute of Clinical Biology and Metabolism, Postas 13, 01004 Vitoria, Spain;
| | - Khalid Alfarouk
- Al-Ghad International Colleges for Applied Medical Sciences, Al-Madinah Al-Munawarah, Saudi Arabia and Alfarouk Biomedical Research LLC, Tampa, FL 33617, USA;
| | - Julián Polo Orozco
- Institute of Clinical Biology and Metabolism, Postas 13, 01004 Vitoria, Spain;
| | - Kévin Hardonnière
- Université Paris-Saclay, Inserm, Inflammation, Microbiome and Immunosurveillance, 92290 Châtenay-Malabry, France;
| | - Daniel Stanciu
- Scientific Direction, MCS Foundation For Life, 5623KR Eindhoven, The Netherlands;
| | - Stefano Fais
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità (National Institute of Health), Viale Regina Elena, 299, 00161 Rome, Italy;
| | - Jesús Devesa
- Scientific Direction, Foltra Medical Centre, Travesía de Montouto 24, 15886 Teo, Spain;
| |
Collapse
|
291
|
Sharabi S, Guez D, Daniels D, Cooper I, Atrakchi D, Liraz-Zaltsman S, Last D, Mardor Y. The application of point source electroporation and chemotherapy for the treatment of glioma: a randomized controlled rat study. Sci Rep 2020; 10:2178. [PMID: 32034261 PMCID: PMC7005896 DOI: 10.1038/s41598-020-59152-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/23/2020] [Indexed: 11/28/2022] Open
Abstract
The prognosis of Glioblastoma Multiforme patients is poor despite aggressive therapy. Reasons include poor chemotherapy penetration across the blood-brain barrier and tumor infiltration into surrounding tissues. Here we studied the effects of combined point-source electroporation (EP) and systemic chemotherapy in glioma-bearing rats. 128 rats were studied. Treatment groups were administered systemic Cisplatin/Methotrexate before EP (either 90 or 180 pulses). Control groups were treated by EP, chemotherapy, or no treatment. Tumor volumes were determined by MRI. Tumors growth rates of the EP + Methotrexate group (1.02 ± 0.77) were significantly lower (p < 0.01) than the control (5.2 ± 1.0) 1-week post treatment. No significant difference was found compared to Methotrexate (1.7 ± 0.5). Objective response rates (ORR) were 40% and 57% for the Methotrexate and EP + Methotrexate groups respectively. Tumor growth rates and ORR of the EP + Cisplatin groups (90 pulses 0.98 ± 0.2, 57%, 180 pulses 1.2 ± 0.1, 33%) were significantly smaller than the control (6.4 ± 1.0, p < 0.01, p < 0.02, 0%) and Cisplatin (3.9 ± 1.0, p < 0.04, p < 0.05, 13%) groups. No significant differences were found between the control groups. Increased survival was found in the EP + Cisplatin group, Χ2 = 7.54, p < 0.006 (Log Rank). Point-source EP with systemic chemotherapy is a rapid, minimal-invasive treatment that was found to induce significant antineoplastic effects in a rat glioma model.
Collapse
Affiliation(s)
- Shirley Sharabi
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel.
| | - David Guez
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel
| | - Dianne Daniels
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel
| | - Itzik Cooper
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Interdisciplinary Center Herzliya, Herzliya, Israel
| | - Dana Atrakchi
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel
| | - Sigal Liraz-Zaltsman
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel Hashomer, Ramat Gan, Israel.,Gand Faculty of Health Profession, Ono Academic College, Kiryat Ono, Israel.,Department of Pharmacology, Institute for Drug Research, Hebrew University, Jerusalem, Israel
| | - David Last
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel
| | - Yael Mardor
- The Advanced Technology Center, Sheba Medical Center, Ramat-Gan, 52621, Israel.,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
292
|
Sioud F, Amor S, Toumia IB, Lahmar A, Aires V, Chekir-Ghedira L, Delmas D. A New Highlight of Ephedra alata Decne Properties as Potential Adjuvant in Combination with Cisplatin to Induce Cell Death of 4T1 Breast Cancer Cells In Vitro and In Vivo. Cells 2020; 9:E362. [PMID: 32033130 PMCID: PMC7072491 DOI: 10.3390/cells9020362] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 12/29/2022] Open
Abstract
Despite major advances in the last 10 years, whether in terms of prevention or treatment, the 5 year survival rate remains relatively low for a large number of cancers. These therapeutic failures can be the consequence of several factors associated with the cellular modifications or with the host by itself, especially for some anticancer drugs such as cisplatin, which induces a nephrotoxicity. In the strategy of research for active molecules capable both of exerting a protective action against the deleterious effects of cisplatin and exerting a chemosensitizing action with regard to cancer cells, we tested the potential effects of Ephedra alata Decne extract (E.A.) rich in polyphenolic compounds towards a 4T1 breast cancer model in vitro and in vivo. We showed that E.A. extract inhibited cell viability of 4T1 breast cancer cells and induced apoptosis in a caspase-dependent manner, which involved intrinsic pathways. Very interestingly, we observed a synergic antiproliferative and pro-apoptotic action with cisplatin. These events were associated with a strong decrease of breast tumor growth in mice treated with an E.A./cisplatin combination and simultaneously with a decrease of hepato- and nephrotoxicities of cisplatin.
Collapse
Affiliation(s)
- Fairouz Sioud
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
| | - Souheila Amor
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
| | - Imène ben Toumia
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
| | - Aida Lahmar
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
| | - Virginie Aires
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
| | - Leila Chekir-Ghedira
- Research Unit Bioactive Natural Products and Biotechnology UR17ES49, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne street, Monastir 5000, Tunisia; (F.S.); (I.b.T.); (A.L.); (L.C.-G.)
| | - Dominique Delmas
- Université de Bourgogne Franche-Comté, F-21000 Dijon, France; (S.A.); (V.A.)
- INSERM Research Center U1231—Cancer and Adaptive Immune Response Team, F-21000 Dijon, France
- Centre anticancéreux Georges François Leclerc Center, F-21000 Dijon, France
| |
Collapse
|
293
|
Kock FVC, Costa AR, de Oliveira KM, Batista AA, Ferreira AG, Venâncio T. A Supramolecular Interaction of a Ruthenium Complex With Calf-Thymus DNA: A Ligand Binding Approach by NMR Spectroscopy. Front Chem 2019; 7:762. [PMID: 31781544 PMCID: PMC6857657 DOI: 10.3389/fchem.2019.00762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/23/2019] [Indexed: 12/29/2022] Open
Abstract
Lawsone itself exhibits interesting biological activities, and its complexation with a metal center can improve the potency. In this context a cytotoxic Ru-complex, [Ru(law)(dppb)(bipy)] (law = lawsone, dppb = 1,4-bis(diphenylphosphino)butane and bipy = 2,2'-bipyridine), named as CBLAU, was prepared as reported. In this work, NMR binding-target studies were performed to bring to light the most accessible interaction sites of this Ru-complex toward Calf-Thymus DNA (CT-DNA, used as a model), in a similar approach used for other metallic complexes with anti-cancer activity, such as cisplatin and carboplatin. Advanced and robust NMR binding-target studies, among them Saturation Transfer Difference (STD)-NMR and longitudinal relaxometry (T1), were explored. The 1H and 31P -NMR data indicate that the structure of Ru-complex remains preserved in the presence of CT-DNA, and some linewidth broadening is also observed for all the signals, pointing out some interaction. Looking at the binding efficiency, the T1 values are highly influenced by the formation of the CBLAU-DNA adduct, decreasing from 11.4 s (without DNA) to 1.4 s (with DNA), where the difference is bigger for the lawsone protons. Besides, the STD-NMR titration experiments revealed a stronger interaction (KD = 5.9 mM) for CBLAU-DNA in comparison to non-complexed lawsone-DNA (KD = 34.0 mM). The epitope map, obtained by STD-NMR, shows that aromatic protons from the complexed lawsone exhibits higher saturation transfer, in comparison to other Ru-ligands (DPPB and bipy), suggesting the supramolecular contact with CT-DNA takes place by the lawsone face of the Ru-complex, possibly by a spatial π-π stacking involving π-bonds on nucleic acids segments of the DNA chain and the naphthoquinone group.
Collapse
Affiliation(s)
| | - Analu Rocha Costa
- Laboratory of Structure and Reactivity of Inorganic Compounds, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| | - Katia Mara de Oliveira
- Laboratory of Structure and Reactivity of Inorganic Compounds, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| | - Alzir Azevedo Batista
- Laboratory of Structure and Reactivity of Inorganic Compounds, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| | - Antônio Gilberto Ferreira
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| | - Tiago Venâncio
- Laboratory of Nuclear Magnetic Resonance, Department of Chemistry, Federal University of São Carlos, São Carlos, Brazil
| |
Collapse
|