1
|
Kessler L, Koo C, Richter CP, Tan X. Hearing loss during chemotherapy: prevalence, mechanisms, and protection. Am J Cancer Res 2024; 14:4597-4632. [PMID: 39417180 PMCID: PMC11477841 DOI: 10.62347/okgq4382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/23/2024] [Indexed: 10/19/2024] Open
Abstract
Ototoxicity is an often-underestimated sequela for cancer patients undergoing chemotherapy, with an incidence rate exceeding 50%, affecting approximately 4 million individuals worldwide each year. Despite the nearly 2,000 publications on chemotherapy-related ototoxicity in the past decade, the understanding of its prevalence, mechanisms, and preventative or therapeutic measures remains ambiguous and subject to debate. To date, only one drug, sodium thiosulfate, has gained FDA approval for treating ototoxicity in chemotherapy. However, its utilization is restricted. This review aims to offer clinicians and researchers a comprehensive perspective by thoroughly and carefully reviewing available data and current evidence. Chemotherapy-induced ototoxicity is characterized by four primary symptoms: hearing loss, tinnitus, vertigo, and dizziness, originating from both auditory and vestibular systems. Hearing loss is the predominant symptom. Amongst over 700 chemotherapeutic agents documented in various databases, only seven are reported to induce hearing loss. While the molecular mechanisms of the hearing loss caused by the two platinum-based drugs are extensively explored, the pathways behind the action of the other five drugs are primarily speculative, rooted in their therapeutic properties and side effects. Cisplatin attracts the majority of attention among these drugs, encompassing around two-thirds of the literature regarding ototoxicity in chemotherapy. Cisplatin ototoxicity chiefly manifests through the loss of outer hair cells, possibly resulting from damages directly by cisplatin uptake or secondary effects on the stria vascularis. Both direct and indirect influences contribute to cisplatin ototoxicity, while it is still debated which path is dominant or where the primary target of cisplatin is located. Candidates for hearing protection against cisplatin ototoxicity are also discussed, with novel strategies and methods showing promise on the horizon.
Collapse
Affiliation(s)
- Lexie Kessler
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Chail Koo
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
| | - Claus-Peter Richter
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Biomedical Engineering, Northwestern UniversityEvanston, Illinois 60208, USA
- Department of Communication Sciences and Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| | - Xiaodong Tan
- Department of Otolaryngology-Head and Neck Surgery, Feinberg School of Medicine, Northwestern UniversityChicago, Illinois 60611, USA
- Hugh Knowles Center for Clinical and Basic Science in Hearing and Its Disorders, Northwestern UniversityEvanston, Illinois 60208, USA
| |
Collapse
|
2
|
Duan M, Leng S, Mao P. Cisplatin in the era of PARP inhibitors and immunotherapy. Pharmacol Ther 2024; 258:108642. [PMID: 38614254 DOI: 10.1016/j.pharmthera.2024.108642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Platinum compounds such as cisplatin, carboplatin and oxaliplatin are widely used in chemotherapy. Cisplatin induces cytotoxic DNA damage that blocks DNA replication and gene transcription, leading to arrest of cell proliferation. Although platinum therapy alone is effective against many tumors, cancer cells can adapt to the treatment and gain resistance. The mechanisms for cisplatin resistance are complex, including low DNA damage formation, high DNA repair capacity, changes in apoptosis signaling pathways, rewired cell metabolisms, and others. Drug resistance compromises the clinical efficacy and calls for new strategies by combining cisplatin with other therapies. Exciting progress in cancer treatment, particularly development of poly (ADP-ribose) polymerase (PARP) inhibitors and immune checkpoint inhibitors, opened a new chapter to combine cisplatin with these new cancer therapies. In this Review, we discuss how platinum synergizes with PARP inhibitors and immunotherapy to bring new hope to cancer patients.
Collapse
Affiliation(s)
- Mingrui Duan
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA
| | - Shuguang Leng
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| | - Peng Mao
- Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| |
Collapse
|
3
|
Wang P, Ouyang J, Jia Z, Zhang A, Yang Y. Roles of DNA damage in renal tubular epithelial cells injury. Front Physiol 2023; 14:1162546. [PMID: 37089416 PMCID: PMC10117683 DOI: 10.3389/fphys.2023.1162546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
The prevalence of renal diseases including acute kidney injury (AKI) and chronic kidney disease (CKD) is increasing worldwide. However, the pathogenesis of most renal diseases is still unclear and effective treatments are still lacking. DNA damage and the related DNA damage response (DDR) have been confirmed as common pathogenesis of acute kidney injury and chronic kidney disease. Reactive oxygen species (ROS) induced DNA damage is one of the most common types of DNA damage involved in the pathogenesis of acute kidney injury and chronic kidney disease. In recent years, several developments have been made in the field of DNA damage. Herein, we review the roles and developments of DNA damage and DNA damage response in renal tubular epithelial cell injury in acute kidney injury and chronic kidney disease. In this review, we conclude that focusing on DNA damage and DNA damage response may provide valuable diagnostic biomarkers and treatment strategies for renal diseases including acute kidney injury and chronic kidney disease.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Jing Ouyang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yunwen Yang
- Department of Nephrology, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Nanjing Key Laboratory of Pediatrics, Children’s Hospital of Nanjing Medical University, Nanjing, China
- Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| |
Collapse
|
4
|
Horibe S, Ishikawa K, Nakada K, Wake M, Takeda N, Tanaka T, Kawauchi S, Sasaki N, Rikitake Y. Mitochondrial DNA mutations are involved in the acquisition of cisplatin resistance in human lung cancer A549 cells. Oncol Rep 2021; 47:32. [PMID: 34935060 PMCID: PMC8717125 DOI: 10.3892/or.2021.8243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 11/17/2021] [Indexed: 11/21/2022] Open
Abstract
The efficacy of cisplatin (CDDP) has been demonstrated in the treatment of various cancers as monotherapy and combination therapy with immunotherapy. However, acquired CDDP resistance is a major obstacle to successful treatment. In the present study, the mechanisms underlying acquired CDDP resistance were examined using ACR20 cells, which are CDDP-resistant cells derived from A549 lung cancer cells. CDDP induces cytotoxicity by binding nuclear DNA and generating reactive oxygen species (ROS). Contrary to our expectation, ROS levels were elevated in ACR20 cells not treated with CDDP. Pretreatment with an ROS inhibitor enhanced the sensitivity of ACR20 cells to CDDP and prevented the activation of nuclear factor (NF)-кB signaling and upregulation of inhibitor of apoptosis proteins (IAPs). Notably, evaluation of the mitochondrial oxygen consumption rate and mitochondrial superoxide levels revealed a deterioration of mitochondrial function in ACR20 cells. Mitochondrial DNA PCR-RFLP analysis revealed four mutations with varying percentage levels in ACR20 cells. In addition, in cytoplasmic hybrids with mitochondria from ACR20 cells, intrinsic ROS levels were elevated, expression of IAPs was increased, and complex I activity and sensitivity to CDDP were decreased. Analysis of three-dimensional structure data indicated that a mutation (ND2 F40L) may impact the proton translocation pathway, thereby affecting mitochondrial complex I activity. Together, these findings suggest that intrinsic ROS levels were elevated by mitochondrial DNA mutations, which decreased the sensitivity to CDDP via activation of NF-κB signaling and induction of IAP expression in ACR20 cells. These findings indicate that newly identified mutations in mitochondrial DNA may lead to acquired cisplatin resistance in cancer.
Collapse
Affiliation(s)
- Sayo Horibe
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada‑ku, Kobe, Hyogo 658‑8558, Japan
| | - Kaori Ishikawa
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki 305‑8572, Japan
| | - Kazuto Nakada
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tennodai, Tsukuba, Ibaraki 305‑8572, Japan
| | - Masaki Wake
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Yakushiji, Shimotsuke‑shi, Tochigi 329‑0498, Japan
| | - Norihiko Takeda
- Division of Cardiology and Metabolism, Center for Molecular Medicine, Jichi Medical University, Yakushiji, Shimotsuke‑shi, Tochigi 329‑0498, Japan
| | - Toru Tanaka
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada‑ku, Kobe, Hyogo 658‑8558, Japan
| | - Shoji Kawauchi
- Comprehensive Education and Research Center, Kobe Pharmaceutical University, Higashinada‑ku, Kobe, Hyogo 658‑8558, Japan
| | - Naoto Sasaki
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada‑ku, Kobe, Hyogo 658‑8558, Japan
| | - Yoshiyuki Rikitake
- Laboratory of Medical Pharmaceutics, Kobe Pharmaceutical University, Higashinada‑ku, Kobe, Hyogo 658‑8558, Japan
| |
Collapse
|
5
|
Regression Modeling of the Antioxidant-to-Nephroprotective Relation Shows the Pivotal Role of Oxidative Stress in Cisplatin Nephrotoxicity. Antioxidants (Basel) 2021; 10:antiox10091355. [PMID: 34572987 PMCID: PMC8464812 DOI: 10.3390/antiox10091355] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/20/2021] [Accepted: 08/23/2021] [Indexed: 12/14/2022] Open
Abstract
The clinical utility of the chemotherapeutic drug cisplatin is significantly limited by its nephrotoxicity, which is characterized by electrolytic disorders, glomerular filtration rate decline, and azotemia. These alterations are consequences of a primary tubulopathy causing injury to proximal and distal epithelial cells, and thus tubular dysfunction. Oxidative stress plays a role in cisplatin nephrotoxicity and cytotoxicity, but its relative contribution to overall toxicity remains unknown. We studied the relation between the degree of oxidative reduction (provided by antioxidant treatment) and the extent of nephrotoxicity amelioration (i.e., nephroprotection) by means of a regression analysis of studies in animal models. Our results indicate that a linear relation exists between these two parameters, and that this relation very nearly crosses the value of maximal nephroprotection at maximal antioxidant effect, suggesting that oxidative stress seems to be a pivotal and mandatory mechanism of cisplatin nephrotoxicity, and, hence, an interesting, rationale-based target for clinical use. Our model also serves to identify antioxidants with enhanced effectiveness by comparing their actual nephroprotective power with that predicted by their antioxidant effect. Among those, this study identified nanoceria, erythropoietin, and maltol as highly effective candidates affording more nephroprotection than expected from their antioxidant effect for prospective clinical development.
Collapse
|
6
|
Gai Z, Gui T, Kullak-Ublick GA, Li Y, Visentin M. The Role of Mitochondria in Drug-Induced Kidney Injury. Front Physiol 2020; 11:1079. [PMID: 33013462 PMCID: PMC7500167 DOI: 10.3389/fphys.2020.01079] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/05/2020] [Indexed: 12/11/2022] Open
Abstract
The kidneys utilize roughly 10% of the body’s oxygen supply to produce the energy required for accomplishing their primary function: the regulation of body fluid composition through secreting, filtering, and reabsorbing metabolites and nutrients. To ensure an adequate ATP supply, the kidneys are particularly enriched in mitochondria, having the second highest mitochondrial content and thus oxygen consumption of our body. The bulk of the ATP generated in the kidneys is consumed to move solutes toward (reabsorption) or from (secretion) the peritubular capillaries through the concerted action of an array of ATP-binding cassette (ABC) pumps and transporters. ABC pumps function upon direct ATP hydrolysis. Transporters are driven by the ion electrochemical gradients and the membrane potential generated by the asymmetric transport of ions across the plasma membrane mediated by the ATPase pumps. Some of these transporters, namely the polyspecific organic anion transporters (OATs), the organic anion transporting polypeptides (OATPs), and the organic cation transporters (OCTs) are highly expressed on the proximal tubular cell membranes and happen to also transport drugs whose levels in the proximal tubular cells can rapidly rise, thereby damaging the mitochondria and resulting in cell death and kidney injury. Drug-induced kidney injury (DIKI) is a growing public health concern and a major cause of drug attrition in drug development and post-marketing approval. As part of the article collection “Mitochondria in Renal Health and Disease,” here, we provide a critical overview of the main molecular mechanisms underlying the mitochondrial damage caused by drugs inducing nephrotoxicity.
Collapse
Affiliation(s)
- Zhibo Gai
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China.,Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Ting Gui
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Gerd A Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Mechanistic Safety, CMO & Patient Safety, Global Drug Development, Novartis Pharma, Basel, Switzerland
| | - Yunlun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China.,The Third Department of Cardiovascular Diseases, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Michele Visentin
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
7
|
Luo K, Guo W, Yu Y, Xu S, Zhou M, Xiang K, Niu K, Zhu X, Zhu G, An Z, Yu Q, Gan Z. Reduction-sensitive platinum (IV)-prodrug nano-sensitizer with an ultra-high drug loading for efficient chemo-radiotherapy of Pt-resistant cervical cancer in vivo. J Control Release 2020; 326:25-37. [PMID: 32531414 DOI: 10.1016/j.jconrel.2020.06.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 06/01/2020] [Accepted: 06/05/2020] [Indexed: 01/03/2023]
Abstract
Cisplatin is widely used in the chemoradiotherapy (CRT) of cervical cancers. However, despite the severe systemic side effects, the therapeutic efficacy of cisplatin is often compromised by the development of drug resistance, which is closely related to the elevated intracellular thiol-containing species (especially glutathione (GSH)) and the adenosine triphosphate (ATP)-dependent glutathione S-conjugate pumps. The construction of a safe and redox-sensitive nano-sensitizer with high disulfide density and high Pt(IV) prodrug loading capacity (up to 16.50% Pt and even higher), as described herein, is a promising way to overcome the cisplatin resistance and enhance the CRT efficacy. The optimized nanoparticles (NPs) (referred to as SSCV5) with moderate Pt loading (7.62% Pt) and median size (c.a. 40 nm) was screened out and used for further biological evaluation. Compared with free cisplatin, more drugs could be transported and released inside the cisplatin resistant cells (Hela-CDDP) by SSCV5 NPs. With the synergistic effect of GSH scavenging and mitochondrial damage, SSCV5 NPs can easily reverse the cisplatin resistance. Moreover, the higher nucleus DNA binding Pt content of SSCV5 NPs not only caused the DNA damage and apoptosis of Hela-CDDP cells but also sensitized these cells to X-Ray radiation. The in vivo safety and efficacy results showed that SSCV5 NPs effectively accumulated inside tumor and inhibited the growth of cisplatin resistant xenograft models while alleviating the serious side effect associated with cisplatin (the maximum tolerated cisplatin equivalent of single injection is higher than 20 mg/kg body weight). The intervention of exogenous radiation further improved the anticancer efficacy of SSCV5 NPs and caused the shrinkage of tumor volume, thus making this safe and facile nano-sensitizer a promising route for the neoadjuvant CRT of cervical cancers.
Collapse
Affiliation(s)
- Kejun Luo
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Wenxuan Guo
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Yanting Yu
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Simeng Xu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Min Zhou
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Keqi Xiang
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Kun Niu
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Xianqi Zhu
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China
| | - Guangying Zhu
- Department of radiation oncology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zheng An
- Proton therapy center, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qingsong Yu
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China.
| | - Zhihua Gan
- State Key Laboratory of Organic-Inorganic Composite Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing 100029, China.
| |
Collapse
|
8
|
Links between cancer metabolism and cisplatin resistance. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 354:107-164. [PMID: 32475471 DOI: 10.1016/bs.ircmb.2020.01.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cisplatin is one of the most potent and widely used chemotherapeutic agent in the treatment of several solid tumors, despite the high toxicity and the frequent relapse of patients due to the onset of drug resistance. Resistance to chemotherapeutic agents, either intrinsic or acquired, is currently one of the major problems in oncology. Thus, understanding the biology of chemoresistance is fundamental in order to overcome this challenge and to improve the survival rate of patients. Studies over the last 30 decades have underlined how resistance is a multifactorial phenomenon not yet completely understood. Recently, tumor metabolism has gained a lot of interest in the context of chemoresistance; accumulating evidence suggests that the rearrangements of the principal metabolic pathways within cells, contributes to the sensitivity of tumor to the drug treatment. In this review, the principal metabolic alterations associated with cisplatin resistance are highlighted. Improving the knowledge of the influence of metabolism on cisplatin response is fundamental to identify new possible metabolic targets useful for combinatory treatments, in order to overcome cisplatin resistance.
Collapse
|
9
|
Ongnok B, Chattipakorn N, Chattipakorn SC. Doxorubicin and cisplatin induced cognitive impairment: The possible mechanisms and interventions. Exp Neurol 2019; 324:113118. [PMID: 31756316 DOI: 10.1016/j.expneurol.2019.113118] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 10/18/2019] [Accepted: 11/15/2019] [Indexed: 12/24/2022]
Abstract
Chemotherapy has significantly increased the number of cancer survivors. However, chemotherapy itself carries various adverse effects that limit the efficacy of treatment and quality of life of the cancer patients. Most patients who have received chemotherapy report some cognitive deficit characterized by dysfunction in memory, learning, concentration, and reasoning. The phenomenon of cognitive decline developed from chemotherapy treatment is referred to as chemotherapy-induced cognitive impairment (CICI) or chemobrain. The two most common cancers occurring worldwide are lung and breast cancer. The predominant chemotherapeutic drugs used to treat lung and breast cancer are doxorubicin and cisplatin. There is evidence to suggest that both drugs potentially induce chemobrain. The evidence around the proposed pathogenesis of chemobrain caused by these two drugs is inconsistent. Understanding the underlying mechanisms involved in the development of chemobrain would aid in the prevention or treatment of the adverse effects of chemotherapy on brain. This review will summarize and discuss controversial findings and possible mechanisms involved in the development of chemobrain and the interventions which could limit it from in vitro, in vivo, and clinical studies.
Collapse
Affiliation(s)
- Benjamin Ongnok
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Neuroelectrophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
10
|
Protective Effects of ACY-1215 Against Chemotherapy-Related Cognitive Impairment and Brain Damage in Mice. Neurochem Res 2019; 44:2460-2469. [DOI: 10.1007/s11064-019-02882-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/29/2019] [Accepted: 09/18/2019] [Indexed: 11/26/2022]
|
11
|
Cseh AM, Fabian Z, Quintana-Cabrera R, Szabo A, Eros K, Soriano ME, Gallyas F, Scorrano L, Sumegi B. PARP Inhibitor PJ34 Protects Mitochondria and Induces DNA-Damage Mediated Apoptosis in Combination With Cisplatin or Temozolomide in B16F10 Melanoma Cells. Front Physiol 2019; 10:538. [PMID: 31133874 PMCID: PMC6514236 DOI: 10.3389/fphys.2019.00538] [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: 01/27/2019] [Accepted: 04/15/2019] [Indexed: 12/24/2022] Open
Abstract
PARP-1 inhibition has recently been employed in both mono- and combination therapies in various malignancies including melanoma with both promising and contradicting results reported. Although deeper understanding of the underlying molecular mechanisms may help improving clinical modalities, the complex cellular effects of PARP inhibitors make disentangling of the mechanisms involved in combination therapies difficult. Here, we used two cytostatic agents used in melanoma therapies in combination with PARP inhibition to have an insight into cellular events using the B16F10 melanoma model. We found that, when used in combination with cisplatin or temozolomide, pharmacologic blockade of PARP-1 by PJ34 augmented the DNA-damaging and cytotoxic effects of both alkylating compounds. Interestingly, however, this synergism unfolds relatively slowly and is preceded by molecular events that are traditionally believed to support cell survival including the stabilization of mitochondrial membrane potential and morphology. Our data indicate that the PARP inhibitor PJ34 has, apparently, opposing effects on the mitochondrial structure and cell survival. While, initially, it stimulates mitochondrial fusion and hyperpolarization, hallmarks of mitochondrial protection, it enhances the cytotoxic effects of alkylating agents at later stages. These findings may contribute to the optimization of PARP inhibitor-based antineoplastic modalities.
Collapse
Affiliation(s)
- Anna Maria Cseh
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary.,Department of Biology, University of Padova, Padua, Italy
| | - Zsolt Fabian
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Ruben Quintana-Cabrera
- Institute of Functional Biology and Genomics, University of Salamanca, Consejo Superior de Investigaciones Científicas, Salamanca, Spain.,Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, Consejo Superior de Investigaciones Científicas, Salamanca, Spain.,CIBERFES, Instituto de Salud Carlos III, Madrid, Spain
| | - Aliz Szabo
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Krisztian Eros
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Maria Eugenia Soriano
- Department of Biology, University of Padova, Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy
| | - Ferenc Gallyas
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Luca Scorrano
- Department of Biology, University of Padova, Padua, Italy.,Venetian Institute of Molecular Medicine, Padua, Italy
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, Medical School, University of Pécs, Pécs, Hungary.,Nuclear-Mitochondrial Interactions Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
12
|
Sheng J, Shen L, Sun L, Zhang X, Cui R, Wang L. Inhibition of PI3K/mTOR increased the sensitivity of hepatocellular carcinoma cells to cisplatin via interference with mitochondrial-lysosomal crosstalk. Cell Prolif 2019; 52:e12609. [PMID: 31033054 PMCID: PMC6536453 DOI: 10.1111/cpr.12609] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 01/31/2019] [Accepted: 02/13/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVES The genotoxicity of cisplatin towards nuclear DNA is not sufficient to explain the cisplatin resistance of hepatocellular carcinoma (HCC) cells; cisplatin interacts with many organelles, which can influence the sensitivity. Here, we explored the role of mitochondrial-lysosomal crosstalk in the cisplatin resistance of HCC cells. MATERIALS AND METHODS Huh7 and HepG2 cells were subjected to different treatments. Flow cytometry was conducted to detect mitochondrial reactive oxygen species, mitochondrial mass, lysosomal function, mitochondrial membrane potential and apoptosis. Western blotting was performed to evaluate protein levels. The oxygen consumption rate was measured to evaluate mitochondrial function. RESULTS Cisplatin activated mitophagy and lysosomal biogenesis, resulting in crosstalk between mitochondria and lysosomes and cisplatin resistance in HCC cells. Furthermore, a combination of cisplatin with the phosphatidylinositol-3-kinase/mammalian target of rapamycin (PI3K/mTOR) inhibitor PKI-402 induced lysosomal membrane permeabilization. This effect changed the role of the lysosome from a protective one to that of a cell death promoter, completely destroying the mitochondrial-lysosomal crosstalk and significantly enhancing the sensitivity of HCC cells to cisplatin. CONCLUSIONS This is the first evidence of the importance of mitochondrial-lysosomal crosstalk in the cisplatin resistance of HCC cells and of the destruction of this crosstalk by a PI3K/mTOR inhibitor to increase the sensitivity of HCC cells to cisplatin. This mechanism could be developed as a novel target for treatment of HCC in the future.
Collapse
Affiliation(s)
- Jiyao Sheng
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Luyan Shen
- Department of Pathophysiology, College of Basic Medical SciencesJilin UniversityChangchunJilinChina
| | - Liankun Sun
- Department of Pathophysiology, College of Basic Medical SciencesJilin UniversityChangchunJilinChina
| | - Xuewen Zhang
- Department of Hepatobiliary and Pancreatic SurgeryThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical GeneticThe Second Hospital of Jilin UniversityChangchunJilinChina
| | - Lizhong Wang
- Jilin Provincial Key Laboratory on Molecular and Chemical GeneticThe Second Hospital of Jilin UniversityChangchunJilinChina
| |
Collapse
|
13
|
Abstract
Background:
Since the serendipitous discovery of the antitumor activity of cisplatin
there has been a continuous surge in studies aimed at the development of new cytotoxic
metal complexes. While the majority of these complexes have been designed to interact with
nuclear DNA, other targets for anticancer metallodrugs attract increasing interest. In cancer
cells the mitochondrial metabolism is deregulated. Impaired apoptosis, insensitivity to antigrowth
signals and unlimited proliferation have been linked to mitochondrial dysfunction. It
is therefore not surprising that mitochondria have emerged as a major target for cancer therapy.
Mitochondria-targeting agents are able to bypass resistance mechanisms and to (re-) activate
cell-death programs.
Methods:
Web-based literature searching tools such as SciFinder were used to search for reports
on cytotoxic metal complexes that are taken up by the mitochondria and interact with
mitochondrial DNA or mitochondrial proteins, disrupt the mitochondrial membrane potential,
facilitate mitochondrial membrane permeabilization or activate mitochondria-dependent celldeath
signaling by unbalancing the cellular redox state. Included in the search were publications
investigating strategies to selectively accumulate metallodrugs in the mitochondria.
Results:
This review includes 241 references on antimitochondrial metal complexes, the use
of mitochondria-targeting carrier ligands and the formation of lipophilic cationic complexes.
Conclusion:
Recent developments in the design, cytotoxic potency, and mechanistic understanding
of antimitochondrial metal complexes, in particular of cyclometalated Au, Ru, Ir and
Pt complexes, Ru polypyridine complexes and Au-N-heterocyclic carbene and phosphine
complexes are summarized and discussed.
Collapse
Affiliation(s)
- Andrea Erxleben
- School of Chemistry, National University of Ireland, Galway, Ireland
| |
Collapse
|
14
|
Pathak RK, Basu U, Ahmad A, Sarkar S, Kumar A, Surnar B, Ansari S, Wilczek K, Ivan ME, Marples B, Kolishetti N, Dhar S. A designer bow-tie combination therapeutic platform: An approach to resistant cancer treatment by simultaneous delivery of cytotoxic and anti-inflammatory agents and radiation. Biomaterials 2018; 187:117-129. [DOI: 10.1016/j.biomaterials.2018.08.062] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/28/2018] [Accepted: 08/29/2018] [Indexed: 12/20/2022]
|
15
|
Basu U, Banik B, Wen R, Pathak RK, Dhar S. The Platin-X series: activation, targeting, and delivery. Dalton Trans 2018; 45:12992-3004. [PMID: 27493131 DOI: 10.1039/c6dt01738j] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Anticancer platinum (Pt) complexes have long been considered to be one of the biggest success stories in the history of medicinal inorganic chemistry. Yet there remains the hunt for the "magic bullet" which can satisfy the requirements of an effective chemotherapeutic drug formulation. Pt(iv) complexes are kinetically more inert than the Pt(ii) congeners and offer the opportunity to append additional functional groups/ligands for prodrug activation, tumor targeting, or drug delivery. The ultimate aim of functionalization is to enhance the tumor selective action and attenuate systemic toxicity of the drugs. Moreover, an increase in cellular accumulation to surmount the resistance of the tumor against the drugs is also of paramount importance in drug development and discovery. In this review, we will address the attempts made in our lab to develop Pt(iv) prodrugs that can be activated and delivered using targeted nanotechnology-based delivery platforms.
Collapse
Affiliation(s)
- Uttara Basu
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Bhabatosh Banik
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Ru Wen
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Rakesh K Pathak
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| | - Shanta Dhar
- NanoTherapeutics Research Laboratory, Department of Chemistry, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
16
|
Geoghegan F, Buckland RJ, Rogers ET, Khalifa K, O'Connor EB, Rooney MF, Behnam-Motlagh P, Nilsson TK, Grankvist K, Porter RK. Bioenergetics of acquired cisplatin resistant H1299 non-small cell lung cancer and P31 mesothelioma cells. Oncotarget 2017; 8:94711-94725. [PMID: 29212260 PMCID: PMC5706906 DOI: 10.18632/oncotarget.21885] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 09/21/2017] [Indexed: 01/31/2023] Open
Abstract
Acquired cisplatin resistance is a common feature of tumours following cancer treatment with cisplatin and also of non-small cell lung cancer (H1299) and mesothelioma (P31) cell lines exposed to cisplatin. To elucidate the cellular basis of acquired cisplatin resistance, a comprehensive bioenergetic analysis was undertaken. We demonstrate that cellular oxygen consumption was significantly decreased in cisplatin resistant cells and that the reduction was primarily due to reduced mitochondrial activity as a result of reduced mitochondrial abundance. The differential mitochondrial abundance was supported by data showing reduced sirtuin 1 (SIRT1), peroxisome-proliferator activator receptor-γ co-activator 1-alpha (PGC1α), sirtuin 3 (SIRT3) and mitochondrial transcription factor A (TFAM) protein expression in resistant cells. Consistent with these data we observed increased reactive oxygen species (ROS) production and increased hypoxia inducible factor 1-alpha (HIF1α) stabilization in cisplatin resistant cells when compared to cisplatin sensitive controls. We also observed an increase in AMP kinase subunit α2 (AMPKα2) transcripts and protein expression in resistant H1299 cells. mRNA expression was also reduced for cisplatin resistant H1299 cells in these genes, however the pattern was not consistent in resistant P31 cells. There was very little change in DNA methylation of these genes, suggesting that the cells are not stably reprogrammed epigenetically. Taken together, our data demonstrate reduced oxidative metabolism, reduced mitochondrial abundance, potential for increased glycolytic flux and increased ROS production in acquired cisplatin resistant cells. This suggests that the metabolic changes are a result of reduced SIRT3 expression and increased HIF-1α stabilization.
Collapse
Affiliation(s)
- Fintan Geoghegan
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Robert J Buckland
- Dept of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Eric T Rogers
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Karima Khalifa
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Emma B O'Connor
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | - Mary F Rooney
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| | | | - Torbjörn K Nilsson
- Dept of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Kjell Grankvist
- Dept of Medical Biosciences, Clinical Chemistry, Umeå University, Umeå, Sweden
| | - Richard K Porter
- School of Biochemistry and Immunology, Trinity Biomedical Science Institute (TBSI), Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
17
|
Noh MR, Kim KY, Han SJ, Kim JI, Kim HY, Park KM. Methionine Sulfoxide Reductase A Deficiency Exacerbates Cisplatin-Induced Nephrotoxicity via Increased Mitochondrial Damage and Renal Cell Death. Antioxid Redox Signal 2017; 27:727-741. [PMID: 28158949 DOI: 10.1089/ars.2016.6874] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
AIMS Methionine sulfoxide reductase A (MsrA), which is abundantly localized in the mitochondria, reduces methionine-S-sulfoxide, scavenging reactive oxygen species (ROS). Cisplatin, an anticancer drug, accumulates at high levels in the mitochondria of renal cells, causing mitochondrial impairment that ultimately leads to nephrotoxicity. Here, we investigated the role of MsrA in cisplatin-induced mitochondrial damage and kidney cell death using MsrA gene-deleted (MsrA-/-) mice. RESULTS Cisplatin injection resulted in increases of ROS production, methionine oxidation, and oxidative damage in the kidneys. This oxidative stress was greater in MsrA-/- mouse kidneys than in wild-type (MsrA+/+) mouse kidneys. MsrA gene deletion exacerbated cisplatin-induced reductions in the expression and activity of MsrA and MsrBs, and the expression of thioredoxin 1, glutathione peroxidase 1 and 4, mitochondrial superoxide dismutase, cystathionine-β-synthase, and cystathionine-γ-lyase. Cisplatin induced swelling, cristae loss, and fragmentation of mitochondria with increased lipid peroxidation, more so in MsrA-/- than in MsrA+/+ kidneys. The ratio of mitochondrial fission regulator (Fis1) to fusion regulator (Opa1) was higher in MsrA-/- than MsrA+/+ mice. MsrA deletion exacerbated cisplatin-induced increases in Bax to Bcl-2 ratio, cleaved caspase-3 level, and apoptosis, whereas MsrA overexpression attenuated cisplatin-induced oxidative stress and apoptosis. INNOVATION MsrA gene deletion in mice exacerbates cisplatin-induced renal injury through increases of mitochondrial susceptibility, whereas MsrA overexpression protects cells against cisplatin. CONCLUSION This study demonstrates that MsrA protects kidney cells against cisplatin-induced methionine oxidation, oxidative stress, mitochondrial damage, and apoptosis, suggesting that MsrA could be a useful target protein for the treatment of cisplatin-induced nephrotoxicity. Antioxid. Redox Signal. 27, 727-741.
Collapse
Affiliation(s)
- Mi Ra Noh
- 1 Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine , Junggu, Daegu, Republic of Korea
| | - Ki Young Kim
- 2 Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine , Namgu, Daegu, Republic of Korea
| | - Sang Jun Han
- 1 Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine , Junggu, Daegu, Republic of Korea
| | - Jee In Kim
- 3 Department of Molecular Medicine and MRC, Keimyung University School of Medicine , Dalseogu, Daegu, Republic of Korea
| | - Hwa-Young Kim
- 2 Department of Biochemistry and Molecular Biology, Yeungnam University College of Medicine , Namgu, Daegu, Republic of Korea
| | - Kwon Moo Park
- 1 Department of Anatomy and BK21 Plus, Kyungpook National University School of Medicine , Junggu, Daegu, Republic of Korea
| |
Collapse
|
18
|
Cisplatin selects short forms of the mitochondrial DNA OriB variant (16184-16193 poly-cytosine tract), which confer resistance to cisplatin. Sci Rep 2017; 7:46240. [PMID: 28393913 PMCID: PMC5385546 DOI: 10.1038/srep46240] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/14/2017] [Indexed: 11/09/2022] Open
Abstract
A number of alternations in mitochondrial DNA (mtDNA) have been reported in different types of cancers, and the role of mtDNA in cancer has been attracting increasing interest. In order to investigate the relationship between mtDNA alternations and chemosensitivity, we constructed cybrid (trans-mitochondrial hybrid) cell lines carrying a HeLa nucleus and the mtDNA of healthy individuals because of the presence of somatic alternations in the mtDNA of many cancer cells. After a treatment with 1.0 μg/mL cisplatin for 10 days, we isolated 100 cisplatin-resistant clones, 70 of which carried the shorter mtDNA OriB variant (16184–16193 poly-cytosine tract), which was located in the control region of mtDNA. Whole mtDNA sequencing of 10 clones revealed no additional alternations. Re-construction of the HeLa nucleus and mtDNA from cisplatin-resistant cells showed that cisplatin resistance was only acquired by mtDNA alternations in the control region, and not by possible alternation(s) in the nuclear genome.
Collapse
|
19
|
Gao W, Wu MH, Wang N, Ying MZ, Zhang YY, Hua J, Chuan L, Wang YJ. Mitochondrial transcription factor A contributes to cisplatin resistance in patients with estrogen receptor-positive breast cancer. Mol Med Rep 2016; 14:5304-5310. [DOI: 10.3892/mmr.2016.5881] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Accepted: 09/19/2016] [Indexed: 11/05/2022] Open
|
20
|
Morphine, a potential antagonist of cisplatin cytotoxicity, inhibits cisplatin-induced apoptosis and suppression of tumor growth in nasopharyngeal carcinoma xenografts. Sci Rep 2016; 6:18706. [PMID: 26729257 PMCID: PMC4700493 DOI: 10.1038/srep18706] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/24/2015] [Indexed: 12/21/2022] Open
Abstract
Morphine is an opioid analgesic drug often used for pain relief in cancer patients. However, there is growing evidence that morphine may modulate tumor growth, progression and metastasis. In this study, we evaluated whether morphine modulates cisplatin-induced apoptosis in human nasopharyngeal carcinoma CNE-2 cells and whether morphine affects the antitumor activity of cisplatin on tumor growth in human nasopharyngeal carcinoma CNE-2 xenografts in nude mice. We showed that a pretreatment with morphine (1 μg/ml) inhibited the sensitivity of CNE-2 cells to cisplatin by inhibiting cisplatin-induced CNE-2 cell apoptosis, decreasing caspase-3 activity and increasing the Bcl-2/Bax ratio. However, a high dose of morphine (1000 μg/ml) had the opposite effect. We also showed that at a low dose, morphine enhances chemoresistance in an in vivo nasopharyngeal carcinoma (NPC) model by inhibiting cisplatin-induced apoptosis and decreasing neovascularization. Taken together, our results indicate that a low dose of morphine may lead to chemoresistance of cisplatin in NPC models in vitro and in vivo by inhibiting cisplatin-induced apoptosis and decreasing neovascularization.
Collapse
|
21
|
Mitochondrial Transcription Factor A and Mitochondrial Genome as Molecular Targets for Cisplatin-Based Cancer Chemotherapy. Int J Mol Sci 2015; 16:19836-50. [PMID: 26307971 PMCID: PMC4581328 DOI: 10.3390/ijms160819836] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 07/28/2015] [Accepted: 08/07/2015] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are important cellular organelles that function as control centers of the energy supply for highly proliferative cancer cells and regulate apoptosis after cancer chemotherapy. Cisplatin is one of the most important chemotherapeutic agents and a key drug in therapeutic regimens for a broad range of solid tumors. Cisplatin may directly interact with mitochondria, which can induce apoptosis. The direct interactions between cisplatin and mitochondria may account for our understanding of the clinical activity of cisplatin and development of resistance. However, the basis for the roles of mitochondria under treatment with chemotherapy is poorly understood. In this review, we present novel aspects regarding the unique characteristics of the mitochondrial genome in relation to the use of platinum-based chemotherapy and describe our recent work demonstrating the importance of the mitochondrial transcription factor A (mtTFA) expression in cancer cells.
Collapse
|
22
|
Gatti L, Cassinelli G, Zaffaroni N, Lanzi C, Perego P. New mechanisms for old drugs: Insights into DNA-unrelated effects of platinum compounds and drug resistance determinants. Drug Resist Updat 2015; 20:1-11. [PMID: 26003720 DOI: 10.1016/j.drup.2015.04.001] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/27/2015] [Accepted: 04/29/2015] [Indexed: 01/11/2023]
Abstract
Platinum drugs have been widely used for the treatment of several solid tumors. Although DNA has been recognized as the primary cellular target for these agents, there are unresolved issues concerning their effects and the molecular mechanisms underlying the antitumor efficacy. These cytotoxic agents interact with sub-cellular compartments other than the nucleus. Here, we review how such emerging phenomena contribute to the pharmacologic activity as well as to drug resistance phenotypes. DNA-unrelated effects of platinum drugs involve alterations at the plasma membrane and in endo-lysosomal compartments. A direct interaction with the mitochondria also appears to be implicated in drug-induced cell death. Moreover, the pioneering work of a few groups has shown that platinum drugs can act on the tumor microenvironment as well, and potentiate antitumor activity of the immune system. These poorly understood aspects of platinum drug activity sites may be harnessed to enhance their antitumor efficacy. A complete understanding of DNA-unrelated effects of platinum compounds might reveal new aspects of drug resistance allowing the implementation of the antitumor therapeutic efficacy of platinum compound-based regimens and minimization of their toxic side effects.
Collapse
Affiliation(s)
- Laura Gatti
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42/via Venezian 1, 20133 Milan, Italy
| | - Giuliana Cassinelli
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42/via Venezian 1, 20133 Milan, Italy
| | - Nadia Zaffaroni
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42/via Venezian 1, 20133 Milan, Italy
| | - Cinzia Lanzi
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42/via Venezian 1, 20133 Milan, Italy
| | - Paola Perego
- Molecular Pharmacology Unit, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Amadeo 42/via Venezian 1, 20133 Milan, Italy.
| |
Collapse
|
23
|
Maurmann L, Belkacemi L, Adams NR, Majmudar PM, Moghaddas S, Bose RN. A novel cisplatin mediated apoptosis pathway is associated with acid sphingomyelinase and FAS proapoptotic protein activation in ovarian cancer. Apoptosis 2015; 20:960-74. [DOI: 10.1007/s10495-015-1124-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
24
|
How do changes in the mtDNA and mitochondrial dysfunction influence cancer and cancer therapy? Challenges, opportunities and models. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2015; 764:16-30. [DOI: 10.1016/j.mrrev.2015.01.001] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 12/28/2022]
|
25
|
Wisnovsky SP, Wilson JJ, Radford RJ, Pereira MP, Chan MR, Laposa RR, Lippard SJ, Kelley SO. Targeting mitochondrial DNA with a platinum-based anticancer agent. ACTA ACUST UNITED AC 2013; 20:1323-8. [PMID: 24183971 DOI: 10.1016/j.chembiol.2013.08.010] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 08/07/2013] [Accepted: 08/10/2013] [Indexed: 02/04/2023]
Abstract
An analog of the anticancer drug cisplatin (mtPt) was delivered to mitochondria of human cells using a peptide specifically targeting this organelle. mtPt induces apoptosis without damaging nuclear DNA, indicating that mtDNA damage is sufficient to mediate the activity of a platinum-based chemotherapeutic. This study demonstrates the specific delivery of a platinum drug to mitochondria and investigates the effects of directing this agent outside the nucleus.
Collapse
Affiliation(s)
- Simon P Wisnovsky
- Department of Biochemistry, Faculty of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Sancho-Martínez SM, Prieto-García L, Prieto M, López-Novoa JM, López-Hernández FJ. Subcellular targets of cisplatin cytotoxicity: An integrated view. Pharmacol Ther 2012; 136:35-55. [DOI: 10.1016/j.pharmthera.2012.07.003] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 06/28/2012] [Indexed: 12/29/2022]
|
27
|
Wintzell M, Löfstedt L, Johansson J, Pedersen AB, Fuxe J, Shoshan M. Repeated cisplatin treatment can lead to a multiresistant tumor cell population with stem cell features and sensitivity to 3-bromopyruvate. Cancer Biol Ther 2012; 13:1454-62. [PMID: 22954696 DOI: 10.4161/cbt.22007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cisplatin is used in treatment of several types of cancer, including epithelial ovarian carcinoma (EOC). In order to mimic clinical treatment and to investigate longterm effects of cisplatin in surviving cancer cells, two EOC cell lines were repeatedly treated with low doses. In the SKOV-3 cell line originating from malignant ascites, but not in A2780 cells from a primary tumor, this led to emergence of a stable population (SKOV-3-R) which in the absence of cisplatin showed increased motility, epithelial-mesenchymal transition (EMT) and expression of cancer stem cell markers CD117, CD44 and ALDH1. Accordingly, the cells formed self-renewing spheres in serum-free stem cell medium. Despite upregulation of mitochondrial mass and cytochrome c, and no upregulation of Bcl-2/Bcl-xL, SKOV-3-R were multiresistant to antineoplastic drugs. Cancer stem cells, or tumor-initiating cells (TICs) are highly chemoresistant and are believed to cause relapse into disseminated and resistant EOC. Our second aim was therefore to target resistance in these TIC-like cells. Resistance could be correlated with upregulation of hexokinase-II and VDAC, which are known to form a survival-promoting mitochondrial complex. The cells were thus sensitive to 3-bromopyruvate, which dissociates hexokinase-II from this complex, and were particularly sensitive to combination treatment with cisplatin at doses down to 0.1 x IC 50. 3-bromopyruvate might thus be of use in targeting the especially aggressive TIC populations.
Collapse
Affiliation(s)
- My Wintzell
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
28
|
Rodríguez-Enríquez S, Hernández-Esquivel L, Marín-Hernández A, Dong LF, Akporiaye ET, Neuzil J, Ralph SJ, Moreno-Sánchez R. Molecular mechanism for the selective impairment of cancer mitochondrial function by a mitochondrially targeted vitamin E analogue. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2012; 1817:1597-607. [PMID: 22627082 DOI: 10.1016/j.bbabio.2012.05.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 04/30/2012] [Accepted: 05/10/2012] [Indexed: 12/31/2022]
Abstract
The effects of α-tocopheryl succinate (α-TOS), α-tocopheryl acetyl ether (α-TEA) and triphenylphosphonium-tagged vitamin E succinate (mitochondrially targeted vitamin E succinate; MitoVES) on energy-related mitochondrial functions were determined in mitochondria isolated from AS-30D hepatoma and rat liver, bovine heart sub-mitochondrial particles (SMPs), and in rodent and human carcinoma cell lines and rat hepatocytes. In isolated mitochondria, MitoVES stimulated basal respiration and ATP hydrolysis, but inhibited net state 3 (ADP-stimulated) respiration and Ca(2+) uptake, by collapsing the membrane potential at low doses (1-10μM). Uncoupled mitochondrial respiration and basal respiration of SMPs were inhibited by the three drugs at concentrations at least one order of magnitude higher and with different efficacy: MitoVES>α-TEA>α-TOS. At high doses (>10μM), the respiratory complex II (CII) was the most sensitive MitoVES target. Acting as an uncoupler at low doses, this agent stimulated total O(2) uptake, collapsed ∆ψ(m), inhibited oxidative phosphorylation and induced ATP depletion in rodent and human cancer cells more potently than in normal rat hepatocytes. These findings revealed that in situ tumor mitochondria are preferred targets of the drug, indicating its clinical relevance.
Collapse
|
29
|
Abstract
Mitochondria control essential cellular activities including generation of ATP via oxidative phosphorylation. Mitochondrial DNA (mtDNA) mutations in the regulatory D-loop region and somatic mtDNA mutations are common in primary human cancers. The biological impact of a given mutation may vary, depending on the nature of the mutation and the proportion of mutant mtDNAs carried by the cell. Identification of mtDNA mutations in precancerous lesions supports their early contribution to cell transformation and cancer progression. Introduction of mtDNA mutations in transformed cells has been associated with increased ROS production and tumor growth. Studies reveal that increased and altered mtDNA plays a role in the development of cancer but further work is required to establish the functional significance of specific mitochondrial mutations in cancer and disease progression. This review offers some insight into the extent of mtDNA mutations, their functional consequences in tumorigenesis, mitochondrial therapeutics, and future clinical application.
Collapse
Affiliation(s)
- Aditi Chatterjee
- Department of Otolaryngology-Head and Neck Surgery, Johns Hopkins University, Baltimore, MD, USA
| | | | | |
Collapse
|
30
|
Yoshida Y, Hasegawa J, Nezu R, Kim YK, Hirota M, Kawano K, Izumi H, Kohno K. Clinical usefulness of mitochondrial transcription factor A expression as a predictive marker in colorectal cancer patients treated with FOLFOX. Cancer Sci 2011; 102:578-82. [DOI: 10.1111/j.1349-7006.2010.01835.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
31
|
Chapman EG, Hostetter AA, Osborn MF, Miller AL, DeRose VJ. Binding of kinetically inert metal ions to RNA: the case of platinum(II). Met Ions Life Sci 2011; 9:347-77. [PMID: 22010278 PMCID: PMC4080900 DOI: 10.1039/9781849732512-00347] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In this chapter several aspects of Pt(II) are highlighted that focus on the properties of Pt(II)-RNA adducts and the possibility that they influence RNA-based processes in cells. Cellular distribution of Pt(II) complexes results in significant platination of RNA, and localization studies find Pt(II) in the nucleus, nucleolus, and a distribution of other sites in cells. Treatment with Pt(II) compounds disrupts RNA-based processes including enzymatic processing, splicing, and translation, and this disruption may be indicative of structural changes to RNA or RNA-protein complexes. Several RNA-Pt(II) adducts have been characterized in vitro by biochemical and other methods. Evidence for Pt(II) binding in non-helical regions and for Pt(II) cross-linking of internal loops has been found. Although platinated sites have been identified, there currently exists very little in the way of detailed structural characterization of RNA-Pt(II) adducts. Some insight into the details of Pt(II) coordination to RNA, especially RNA helices, can be gained from DNA model systems. Many RNA structures, however, contain complex tertiary folds and common, purine-rich structural elements that present suitable Pt(II) nucleophiles in unique arrangements which may hold the potential for novel types of platinum-RNA adducts. Future research aimed at structural characterization of platinum-RNA adducts may provide further insights into platinum-nucleic acid binding motifs, and perhaps provide a rationale for the observed inhibition by Pt(II) complexes of splicing, translation, and enzymatic processing.
Collapse
Affiliation(s)
- Erich G. Chapman
- Department of Chemistry University of Oregon Eugene OR 97403 USA
| | | | - Maire F. Osborn
- Department of Chemistry University of Oregon Eugene OR 97403 USA
| | - Amanda L. Miller
- Department of Chemistry University of Oregon Eugene OR 97403 USA
| | | |
Collapse
|
32
|
Prasad SB, Rosangkima G, Kharbangar A. Structural and biochemical changes in mitochondria after cisplatin treatment of Dalton’s lymphoma-bearing mice. Mitochondrion 2010; 10:38-45. [DOI: 10.1016/j.mito.2009.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2009] [Revised: 09/07/2009] [Accepted: 09/11/2009] [Indexed: 10/20/2022]
|
33
|
Tarladacalisir YT, Kanter M, Uygun M. Protective Effects of Vitamin C on Cisplatin-Induced Renal Damage: A Light and Electron Microscopic Study. Ren Fail 2009; 30:1-8. [DOI: 10.1080/08860220701742070] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
34
|
LaRiviere FJ, Newman AG, Watts ML, Bradley SQ, Juskewitch JE, Greenwood PG, Millard JT. Quantitative PCR analysis of diepoxybutane and epihalohydrin damage to nuclear versus mitochondrial DNA. Mutat Res 2009; 664:48-54. [PMID: 19428380 PMCID: PMC2727856 DOI: 10.1016/j.mrfmmm.2009.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2008] [Revised: 12/04/2008] [Accepted: 02/06/2009] [Indexed: 10/21/2022]
Abstract
The bifunctional alkylating agents diepoxybutane (DEB) and epichlorohydrin (ECH) are linked to the elevated incidence of certain cancers among workers in the synthetic polymer industry. Both compounds form interstrand cross-links within duplex DNA, an activity suggested to contribute to their cytotoxicity. To assess the DNA targeting of these compounds in vivo, we assayed for damage within chicken erythro-progenitor cells at three different sites: one within mitochondrial DNA, one within expressed nuclear DNA, and one within unexpressed nuclear DNA. We determined the degree of damage at each site via a quantitative polymerase chain reaction, which compares amplification of control, untreated DNA to that from cells exposed to the agent in question. We found that ECH and the related compound epibromohydrin preferentially target nuclear DNA relative to mitochondrial DNA, whereas DEB reacts similarly with the two genomes. Decreased reactivity of the mitochondrial genome could contribute to the reduced apoptotic potential of ECH relative to DEB. Additionally, formation of lesions by all agents occurred at comparable levels for unexpressed and expressed nuclear loci, suggesting that alkylation is unaffected by the degree of chromatin condensation.
Collapse
Affiliation(s)
| | - Adam G. Newman
- Department of Chemistry, Colby College, Waterville ME 04901
| | - Megan L. Watts
- Department of Chemistry, Colby College, Waterville ME 04901
| | | | | | | | | |
Collapse
|
35
|
Scolaro C, Chaplin AB, Hartinger CG, Bergamo A, Cocchietto M, Keppler BK, Sava G, Dyson PJ. Tuning the hydrophobicity of ruthenium(II)-arene (RAPTA) drugs to modify uptake, biomolecular interactions and efficacy. Dalton Trans 2007:5065-72. [PMID: 17992291 DOI: 10.1039/b705449a] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The antitumour activity of the organometallic ruthenium(ii)-arene mixed phosphine complexes, [Ru(eta(6)-p-cymene)Cl(PTA)(PPh(3))]BF(4) and [Ru(eta(6)-C(6)H(5)CH(2)CH(2)OH)Cl(PTA)(PPh(3))]BF(4) (PTA = 1,3,5-triaza-7-phosphaadamantane), have been evaluated in vitro and compared to their RAPTA analogues, [Ru(eta(6)-p-cymene)Cl(2)(PTA)] and [Ru(eta(6)-C(6)H(5)CH(2)CH(2)OH)Cl(2)(PTA)] . The results show that the addition of the PPh(3) ligand to increases the cytotoxicity towards the TS/A adenocarcinoma cancer cells, which correlates with increased uptake, but also increases cytotoxicity to non-tumourigenic HBL-100 cells, thus decreasing selectivity. The decrease in selectivity has been correlated to increased DNA interactions relative to proteins, demonstrated by reactivity of the compounds with a 14-mer oligonucleotide and the model proteins ubiquitin and cytochrome-c.
Collapse
Affiliation(s)
- Claudine Scolaro
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015, Lausanne, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Cullen KJ, Yang Z, Schumaker L, Guo Z. Mitochondria as a critical target of the chemotheraputic agent cisplatin in head and neck cancer. J Bioenerg Biomembr 2007; 39:43-50. [PMID: 17318397 DOI: 10.1007/s10863-006-9059-5] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Cisplatin is among the most important chemotherapeutic agents ever developed. It is a critical component of therapeutic regimens in a broad range of malignancies. However, more than a generation after its clinical introduction, the exact mechanism of cisplatin action on tumor cells is not fully defined. The preponderance of research over the last three decades has focused on cisplatin interactions with nuclear DNA which are felt to lead to apoptotic cell death in sensitive cells. However, recent data have shown that cisplatin may have important direct interactions with mitochondria which can induce apoptosis and may account for a significant portion of the clinical activity associated with this drug. These direct interactions between cisplatin and mitochondria may have critical implications for our understanding of this class of drugs and the development of new therapeutic agents.
Collapse
Affiliation(s)
- Kevin J Cullen
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, Baltimore, MD, USA.
| | | | | | | |
Collapse
|
37
|
Yang Z, Schumaker LM, Egorin MJ, Zuhowski EG, Guo Z, Cullen KJ. Cisplatin preferentially binds mitochondrial DNA and voltage-dependent anion channel protein in the mitochondrial membrane of head and neck squamous cell carcinoma: possible role in apoptosis. Clin Cancer Res 2006; 12:5817-25. [PMID: 17020989 DOI: 10.1158/1078-0432.ccr-06-1037] [Citation(s) in RCA: 194] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Cisplatin adducts to nuclear DNA (nDNA) are felt to be the molecular lesions that trigger apoptosis, but the mechanism linking nDNA adduct formation and cell death is unclear. Some literature in the last decade has suggested a possible direct effect of cisplatin on mitochondria independent of nDNA interaction. In this study, we define separately the sequelae of cisplatin interactions with nDNA and with mitochondria in head and neck squamous cell carcinoma (HNSCC) cell lines. EXPERIMENTAL DESIGN Cisplatin binding to mitochondrial DNA (mtDNA) and proteins was analyzed by atomic absorption spectroscopy and other methods. RESULTS Following 1 hour of exposure to cisplatin, platinum adducts to mtDNA were 300- to 500-fold more abundant than adducts to nDNA; these differences were not due to differences in rates of adduct repair. Whereas HNSCC cell cytoplasts free of nDNA retained the same dose-dependent cisplatin sensitivity as parental cells, HNSCC rho(0) cells free of mtDNA were 4- to 5-fold more resistant to cisplatin than parental cells. Isolated mitochondria released cytochrome c within minutes of exposure to cisplatin, and ultrastructural analysis of intact HNSCC cells by electron microscopy showed marked mitochondrial disruption after 4 hours of cisplatin treatment, whereas the nucleus and other cellular structures remain intact. The very prompt release of cytochrome c from isolated mitochondria implies that apoptosis does not require alteration in mitochondrial gene transcription. Further, cisplatin binds preferentially to mitochondrial membrane proteins, particularly the voltage-dependent anion channel. CONCLUSIONS Cisplatin binding to nDNA is not necessary for induction of apoptosis in HNSCC, which can result from direct action of cisplatin on mitochondria.
Collapse
Affiliation(s)
- Zejia Yang
- University of Maryland Marlene and Stewart Greenebaum Cancer Center, 22 South Greene Street, Baltimore, MD 21201, USA
| | | | | | | | | | | |
Collapse
|
38
|
Dzamitika S, Salerno M, Pereira-Maia E, Le Moyec L, Garnier-Suillerot A. Preferential energy- and potential-dependent accumulation of cisplatin-gutathione complexes in human cancer cell lines (GLC4 and K562): A likely role of mitochondria. J Bioenerg Biomembr 2006; 38:11-21. [PMID: 16732471 DOI: 10.1007/s10863-006-9001-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Accepted: 01/19/2006] [Indexed: 11/28/2022]
Abstract
cis-Diamminedichloroplatinum(II) (CDDP) is an important chemotherapeutic agent used in the treatment of a wide variety of solid tumors. We have recently shown that aquated forms of cisplatin (aqua-Pt) rapidly accumulate in K562 and GLC4 cultured cells, in comparison to CDDP. Thus, when cells are incubated with aquated forms of cisplatin a gradient of concentration is observed after a short time, approximately 40 min, with an intracellular concentration of aqua-Pt of 20-30 times higher than that of extracellular aqua-Pt. The same gradient of concentration is observed when cells are incubated with CDDP but it takes a longer time, i.e., about 24 h. Therefore, the question arises as to the identity of the intracellular sites of accumulation of aqua-Pt. Using several agents to modulate membrane potential, acidic compartment pH and/or ATP level, we obtained evidence that aqua-Pt may accumulate rapidly inside mitochondria as this accumulation is energy- and membrane-potential-dependent. However, aqua-Pt complexes are not characterized by a delocalized charge and a lipophilic character that would permit their movement through the inner membrane. Therefore, it is suggested that intracellular aqua-Pt reacts rapidly with glutathione with the resultant complex being transported inside the mitochondria via one of the known glutathione transporters, i.e., dicarboxylate and/or 2-oxoglutarate transporters present in the inner membrane.
Collapse
Affiliation(s)
- Simplice Dzamitika
- Biophysique Moléculaire Cellulaire et Tissulaire (BioMoCeTi UMR-CNRS 7033), Université Paris 13 et Université Paris 6, UFR SMBH, 74 rue Marcel Cachin, 93017, Bobigny, France
| | | | | | | | | |
Collapse
|
39
|
Kalayda GV, Zhang G, Abraham T, Tanke HJ, Reedijk J. Application of fluorescence microscopy for investigation of cellular distribution of dinuclear platinum anticancer drugs. J Med Chem 2005; 48:5191-202. [PMID: 16078838 DOI: 10.1021/jm050216h] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The dinuclear platinum complexes with aliphatic diamines [{cis-Pt(NH(3))(2)Cl}(2)(mu-H(2)N(CH(2))(6)NH(2))](NO(3))(2) (1,1/c,c) and [{trans-Pt(NH(3))(2)Cl}(2)(mu-H(2)N(CH(2))(4)NH(2))](NO(3))(2) (1,1/t,t), which are known to be highly active in vitro against several cancer cell lines, have been modified with a fluorogenic reporter (carboxyfluorescein diacetate, CFDA) and a hapten (dinitrophenyl, DNP). These labeled complexes have been designed for fluorescence microscopy investigation of cellular pathways of promising dinuclear platinum anticancer drugs and present the first example of labeling biologically active dinuclear platinum complexes with a fluorescent reporter. The modified compounds interact with a guanine model base similarly to the label-free parent complexes. The uptake of the complexes with a fluorescent label and the respective unlabeled complexes in the U2-OS human osteosarcoma cell line and its cisplatin-resistant derivative, U2-OS/Pt cell line has been investigated. Cellular processing of the CFDA- and DNP-modified dinuclear platinum complexes in U2-OS and U2-OS/Pt cells has been studied.
Collapse
Affiliation(s)
- Ganna V Kalayda
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA Leiden, The Netherlands
| | | | | | | | | |
Collapse
|
40
|
Schwerdt G, Freudinger R, Schuster C, Weber F, Thews O, Gekle M. Cisplatin-Induced Apoptosis Is Enhanced by Hypoxia and by Inhibition of Mitochondria in Renal Collecting Duct Cells. Toxicol Sci 2005; 85:735-42. [PMID: 15716484 DOI: 10.1093/toxsci/kfi117] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Cisplatin is a widely used chemotherapeutic agent. Here we show that cisplatin induces apoptosis in renal collecting duct-derived cells (MDCK-C7 cells, resembling principal cells) in a dose-dependent manner. Additionally, we studied the role of mitochondria in this process by inhibition of the mitochondrial respiratory chain, the F1F(o)-ATP synthase or by uncoupling. The role of intra- and extracellular pH in apoptosis induction was investigated. Activation of caspase-3 and DNA ladder formation were used to monitor the apoptotic response. When cells were incubated with inhibitors of the mitochondrial respiratory chain or an inhibitor of the ATP-synthase, cisplatin-induced apoptosis was markedly enhanced. Mitochondrial blockade led to enhanced production of lactic acid. Also, anoxia potentiated the cisplatin-induced caspase-3 activation. Neither intra- nor extracellular pH had an influence on caspase-3 activation at low cisplatin concentrations. Acidic conditions (pH 6.8) potentiated the caspase-3 activation when high (100 microM) cisplatin concentrations were used. We demonstrate that intact mitochondria are important to prevent cisplatin-induced apoptosis in MDCK-C7 cells and that acidic conditions can aggravate the toxic effects of cisplatin.
Collapse
Affiliation(s)
- Gerald Schwerdt
- Physiologisches Institut, Universität Würzburg, Röntgenring 9, D-97070 Würzburg, Germany.
| | | | | | | | | | | |
Collapse
|
41
|
Kalayda GV, Jansen BAJ, Molenaar C, Wielaard P, Tanke HJ, Reedijk J. Dinuclear platinum complexes with N,N′-bis(aminoalkyl)-1,4-diaminoanthraquinones as linking ligands. Part II. Cellular processing in A2780 cisplatin-resistant human ovarian carcinoma cells: new insights into the mechanism of resistance. J Biol Inorg Chem 2004; 9:414-22. [PMID: 15071768 DOI: 10.1007/s00775-004-0540-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2003] [Accepted: 03/11/2004] [Indexed: 11/24/2022]
Abstract
The cellular processing of three fluorescent N, N'-bis(aminoalkyl)-1,4-diaminoanthraquinones (aminoalkyl=2-aminoethyl, 3-aminoprop-1-yl or 4-aminobut-1-yl) and their dinuclear platinum complexes in A2780 human ovarian carcinoma cells with acquired resistance to cisplatin has been monitored over time by time-lapse fluorescence microscopy. The results were compared with the previously reported observations in the parent A2780 cell line. The cellular distribution pattern for the free ligands is similar in sensitive and resistant cells, whereas significant differences in cellular distribution were observed in the case of the platinum complexes. In the cisplatin-resistant cell line the platinum complexes were found to be sequestrated in acidic vesicles in the cytosol from the very beginning of the incubation. This sequestration was not observed in the case of sensitive cells. Platinum accumulation in vesicles possibly presents a mechanism of resistance to platinum complexes. This mechanism appears to be unrelated to the mechanism of deactivation of platinum compounds by glutathione. Encapsulation of the dinuclear platinum complexes in lysosomal vesicles provides a plausible explanation for the decreased activity of these compounds in the resistant cell line, as compared to the sensitive cell line.
Collapse
Affiliation(s)
- Ganna V Kalayda
- Leiden Institute of Chemistry, Gorlaeus Laboratories, Leiden University, P.O. Box 9502, 2300 RA, Leiden, The Netherlands
| | | | | | | | | | | |
Collapse
|
42
|
Marín-Hernández A, Gracia-Mora I, Ruiz-Ramírez L, Moreno-Sánchez R. Toxic effects of copper-based antineoplastic drugs (Casiopeinas) on mitochondrial functions. Biochem Pharmacol 2003; 65:1979-89. [PMID: 12787878 DOI: 10.1016/s0006-2952(03)00212-0] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
To elucidate some of the subcellular and biochemical mechanisms of toxicity of metal-based antineoplastic drugs, mitochondria and cells were exposed to Casiopeinas), a new class of copper-based compounds with high antineoplastic activity. The rates of respiration and swelling, the H(+) gradient, and the activities of succinate (SDH) and 2-oxoglutarate dehydrogenases (2-OGDH) and ATPase were measured in mitochondria isolated from rat liver, kidney, heart, and hepatoma AS-30D. Also, oligomycin-sensitive respiration and ATP content in hepatoma AS-30D cells were determined. Casiopeinas) (CS) II-gly and III-i inhibited the rates of state 3 and uncoupled respiration in mitochondria. CS II was 10 times more potent than CS III. The sensitivity to CS II was 4-5-fold higher in mitochondria incubated with 2-OG than with succinate. Thus, at low concentrations (< or =10 nmol (mg protein)(-1); 10 microM), CS II disturbed mitochondrial functions only when 2-OG was present, due to a specific inhibition of 2-OGDH. At high concentrations (> or =15nmol (mg protein)(-1)), CS II-induced stimulation of basal respiration, followed by a strong inhibition, which correlated with K(+)-dependent swelling and cytochrome c release, respectively; K(+)-channel openers induce a similar mitochondrial response. Mitochondria from liver, kidney and hepatoma showed a similar sensitivity towards CS II, whereas heart mitochondria were more resistant. Oxidative phosphorylation and ATP content were also decreased in tumor cells by CS II. The data suggested that CS affected several different mitochondrial sites, bringing about inhibition of respiration and ATP synthesis, which could compromise energy-dependent processes such as cellular duplication.
Collapse
Affiliation(s)
- Alvaro Marín-Hernández
- Departamento de Bioquímica, Instituto Nacional de Cardiología, Juan Badiano # 1, Col. Sección XVI, Tlalpan, México D.F. 14080, Mexico
| | | | | | | |
Collapse
|
43
|
Jakupec MA, Galanski MS, Keppler BK. Tumour-inhibiting platinum complexes--state of the art and future perspectives. Rev Physiol Biochem Pharmacol 2003; 146:1-54. [PMID: 12605304 DOI: 10.1007/s10254-002-0001-x] [Citation(s) in RCA: 308] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Thirty years after the onset of the first clinical studies with cisplatin, the development of antineoplastic platinum drugs continues to be a productive field of research. This article reviews the current preclinical and clinical status, including a discussion of the molecular basis for the activity of the parent drug cisplatin and platinum drugs of the second and third generation, in particular their interaction with DNA. Further emphasis is laid on the development of third generation platinum drugs with activity in cisplatin-resistant tumours, particularly on chelates containing 1,2-diaminocyclohexane (DACH) and on the promising and more recently evolving field of non-classic ( trans- and multinuclear) platinum complexes. The development of oral platinum drugs and drug targeting strategies using liposomes, polymers or low-molecular-weight carriers in order to improve the therapeutic index of platinum chemotherapy are also covered.
Collapse
Affiliation(s)
- M A Jakupec
- Institute of Inorganic Chemistry, University of Vienna, Währinger Strasse 42, 1090, Vienna, Austria
| | | | | |
Collapse
|
44
|
Schwerdt G, Freudinger R, Schuster C, Silbernagl S, Gekle M. Inhibition of mitochondria prevents cell death in kidney epithelial cells by intra- and extracellular acidification. Kidney Int 2003; 63:1725-35. [PMID: 12675848 DOI: 10.1046/j.1523-1755.2003.00934.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Nephrotoxic substances like cisplatin or ochratoxin A (OTA) induce cell death in human proximal tubule-derived cells (IHKE cells). Mitochondria play a significant role in apoptosis and loss of their function may influence OTA- or cisplatin-induced apoptosis. Extracellular pH also plays an important role in tumor genesis. Therefore, we investigated the role of mitochondria and intra- and extracellular pH on cell death induction by cisplatin or OTA. METHODS IHKE cells were incubated in the presence of OTA or cisplatin, together with inhibitors of the mitochondrial metabolism, and the activity of caspase-3 was measured and DNA laddering was monitored. Adenosine triphosphate (ATP) content of the cells, lactate release into the media, and glucose consumption was determined. In addition, media and cells were acidified or alkalized artificially to investigate the effect of intra- and extracellular pH on cell death induction. Cytochrome C was immunodetected in cellular compartments. RESULTS Inhibition of the mitochondrial function reduced OTA- or cisplatin-induced cell death and led to considerable lactic acid production and extracellular acidification. Intra- and extracellular acidification prevented cells from cell death induced by OTA or cisplatin. No cytochrome C release from mitochondria could be detected during 24 hours of exposure to OTA or cisplatin. CONCLUSION We conclude that OTA- or cisplatin-induced cell death is dependent on functional and intact, ATP-producing mitochondria and that intra- and extracellular pH is crucial for induction of cell death in IHKE cells.
Collapse
Affiliation(s)
- Gerald Schwerdt
- Physiologisches Institut, Universität Würzburg, Würzburg, Germany. gerald
| | | | | | | | | |
Collapse
|
45
|
Meijera C, van Luyn MJ, Nienhuis EF, Blom N, Mulder NH, de Vries EG. Ultrastructural morphology and localisation of cisplatin-induced platinum-DNA adducts in a cisplatin-sensitive and -resistant human small cell lung cancer cell line using electron microscopy. Biochem Pharmacol 2001; 61:573-8. [PMID: 11239500 DOI: 10.1016/s0006-2952(00)00584-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Ultrastructural morphology (transmission electron microscopy) and localisation of cisplatin-induced platinum (Pt)-DNA adducts (immunoelectron microscopy) were analysed in the human small cell lung cancer cell line GLC(4) and its 40-fold in vitro acquired cisplatin-resistant subline GLC(4)-CDDP, which is characterised by, among other things, a decreased DNA platination. Immunolabelling of Pt-DNA adducts was performed with the polyclonal antibody GPt, known to detect the main Pt-containing intrastrand and interstrand DNA adducts. Morphological analysis of GLC(4) and GLC(4)-CDDP at the ultrastructural level showed cells with a high nucleus/cytoplasm ratio with the majority of nuclei containing one or more nucleoli. GLC(4)-CDDP showed, in contrast to GLC(4), an extensive Golgi apparatus and an increased number of mitochondria. DNA platination was detectable in both GLC(4) and GLC(4)-CDDP. Immunoelectron microscopy showed Pt-DNA adducts primarily in the nucleus, preferentially at loci with high-density chromatin (e.g. heterochromatin, pars granulosa around nucleoli, condensed DNA in proliferating and apoptotic cells), and in mitochondria. The level of detectable Pt-DNA adducts was cell cycle status-dependent. In both cell lines, Pt-DNA adduct levels increased from non-dividing interphase cells to dividing cells and were highest in cells undergoing apoptosis. Overall localisation of Pt-DNA adducts was comparable in GLC(4) and GLC(4)-CDDP cells.
Collapse
Affiliation(s)
- C Meijera
- Department of Medical Oncology, P.O. Box 30.001, University Hospital Groningen, 9700RB, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
46
|
lizuka N, Miyamoto K, Tangoku A, Hayashi H, Hazama S, Yoshino S, Yoshimura K, Hirose K, Yoshida H, Oka M. Downregulation of intracellular nm23-H1 prevents cisplatin-induced DNA damage in oesophageal cancer cells: possible association with Na(+), K(+)-ATPase. Br J Cancer 2000; 83:1209-15. [PMID: 11027435 PMCID: PMC2363580 DOI: 10.1054/bjoc.2000.1436] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Previously, we showed that expression of nm23-H1 is associated inversely with sensitivity to cisplatin in human oesophageal squamous cell carcinoma (OSCC). The present study was undertaken to investigate the association of nm23-H1 expression with cisplatin-induced DNA damage in OSCC using antisense nm23-H1 transfectants. YES-2/AS-12, an antisense nm23-H1-transfected OSCC cell line, showed significantly reduced expression of intracellular nm23-H1 protein compared with that in parental YES-2 cells and YES-2/Neo transfectants. Surface expression of nm23-H1 protein was not observed in any of the three cell lines. PCR analysis for DNA damage demonstrated that YES-2/AS-12 cells were more resistant to nuclear and mitochondrial DNA damage by cisplatin than were YES-2/Neo cells. In addition, mitochondrial membrane potentials and DNA fragmentation assays confirmed that YES-2/AS-12 was more resistant than YES-2/Neo to apoptosis induced by cisplatin. In contrast, YES-2/AS-12 was more sensitive to ouabain, a selective inhibitor of Na(+), K(+)-ATPase, than YES-2 and YES-2/Neo. Pre-treatment with ouabain resulted in no differences in cisplatin sensitivity between the three cell lines examined. Intracellular platinum level in YES-2/AS-12 was significantly lower than that in YES-2 and YES-2/Neo following incubation with cisplatin, whereas ouabain pre-treatment resulted in no differences in intracellular platinum accumulations between the three cell lines. Our data support the conclusion that reduced expression of intracellular nm23-H1 in OSCC cells is associated with cisplatin resistance via the prevention of both nuclear and mitochondrial DNA damage and suggest that it may be related to Na(+), K(+)-ATPase activity, which is responsible for intracellular cisplatin accumulation.
Collapse
Affiliation(s)
- N lizuka
- Department of Bioregulatory Function, Department of Surgery II, Yamaguchi University School of Medicine, 1-1-1 Minami-kogushi, Ube, Yamaguchi, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Iizuka N, Hirose K, Noma T, Hazama S, Tangoku A, Hayashi H, Abe T, Yamamoto K, Oka M. The nm23-H1 gene as a predictor of sensitivity to chemotherapeutic agents in oesophageal squamous cell carcinoma. Br J Cancer 1999; 81:469-75. [PMID: 10507772 PMCID: PMC2362931 DOI: 10.1038/sj.bjc.6690717] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Recently, nm23-H1, an anti-metastasis gene, has been reported to correlate with sensitivity to chemotherapeutic agents including cisplatin in human breast and ovarian carcinoma cells. The aim of this study was to evaluate a role for nm23-H1 in responsiveness to cisplatin-based chemotherapy in patients with oesophageal squamous cell carcinoma (OSCC). The expression of nm23-H1 protein was examined immunohistochemically in 32 eligible patients with OSCC who underwent adjuvant chemotherapy with cisplatin, etoposide, and 5-fluorouracil after tumour resection. Fifteen (46.9%) of 32 patients were positive for nm23-H1 staining and 17 (53.1%) were negative. Both disease-free survival and overall survival rates of nm23-H1-negative patients were significantly shorter than in nm23-H1-positive patients (P < 0.01 for both). There was no significant difference in clinicopathologic characteristics between nm23-H1-positive and nm23-H1-negative groups. Multivariate analysis also showed that nm23-H1 expression was the most significant factor for overall survival of OSCC patients included in this study (P = 0.0007). To further study the role of nm23-H1, a human OSCC cell line (YES-2) was transfected with a plasmid containing a fragment of the nm23-H1 cDNA in an antisense orientation. Reduced expression of nm23-H1 protein in the antisense-transfected (AS) clones was found by Western blot analysis as compared to wild-type YES-2 and YES-2/Neo (clone transfected with the neomycin resistance gene alone). MTT (3-(4,5-dimethyl-2-thiazol)-2,5-diphenyl-2H tetrazolium bromide) assay showed that reduced expression of the nm23-H1 protein in AS clones was consistent with the degree of increased resistance to cisplatin but not etoposide or 5-fluorouracil. These data support the conclusion that reduced expression of nm23-H1 may be associated with resistance to cisplatin, suggesting the value of nm23-H1 expression as a prognostic marker for OSCC patients who are to undergo cisplatin-based chemotherapy.
Collapse
MESH Headings
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/surgery
- Chemotherapy, Adjuvant
- Cisplatin/administration & dosage
- Combined Modality Therapy
- DNA, Complementary/genetics
- Disease-Free Survival
- Drug Resistance, Neoplasm/genetics
- Esophageal Neoplasms/drug therapy
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/mortality
- Esophageal Neoplasms/pathology
- Esophageal Neoplasms/surgery
- Etoposide/administration & dosage
- Female
- Fluorouracil/administration & dosage
- Humans
- Male
- Middle Aged
- Monomeric GTP-Binding Proteins
- NM23 Nucleoside Diphosphate Kinases
- Neoplasm Proteins/genetics
- Nucleoside-Diphosphate Kinase
- Oligonucleotides, Antisense/pharmacology
- Survival Analysis
- Survival Rate
- Transcription Factors/genetics
- Transfection
- Treatment Outcome
- Tumor Cells, Cultured/drug effects
Collapse
Affiliation(s)
- N Iizuka
- Department of Bioregulatory Function, Yamaguchi University School of Medicine, Ube, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Affiliation(s)
- L A Marcelino
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge 02139, USA
| | | |
Collapse
|
49
|
Woynarowski JM, Chapman WG, Napier C, Herzig MC, Juniewicz P. Sequence- and region-specificity of oxaliplatin adducts in naked and cellular DNA. Mol Pharmacol 1998; 54:770-7. [PMID: 9804612 DOI: 10.1124/mol.54.5.770] [Citation(s) in RCA: 188] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Oxaliplatin is a clinical anticancer drug with a pharmacological profile distinct from that of cisplatin. Our studies compared site- and region-specificity of lesions induced by oxaliplatin and cisplatin in naked and intracellular DNA, respectively. Oxaliplatin adducts in naked Simian virus 40 (SV40 DNA) were mapped by repetitive primer extension. The sites of oxaliplatin adducts were nearly identical to the sites of cisplatin adducts and were focused in G clusters and GNG motifs probably reflecting intrastrand cross-links. Although alkaline agarose electrophoresis of specific SV40 fragments showed that oxaliplatin formed interstrand cross-links, the levels of this lesion type were low. Drug-induced lesions in discrete loci of cellular DNA were assessed by the polymerase chain reaction stop assay in human tumor A2780 cells. Oxaliplatin at 200 microM induced approximately 1300, approximately 1500, approximately 800, and approximately 300 lesions/10(6) bp in the human beta-globin, c-myc, and HPRT genes and in mitochondrial DNA, respectively. Cisplatin formed two to six times more lesions in the same regions. For both drugs, lesion frequencies seem to parallel the density of drug-binding motifs in the nuclear regions, whereas mitochondrial DNA was disproportionately less affected. Despite less potent induction of DNA lesions, oxaliplatin was more cytotoxic than cisplatin against A2780 cells. Because our findings clearly demonstrate that oxaliplatin forms covalent adducts with a similar sequence- and region-specificity to that of cisplatin, other properties of oxaliplatin adducts, factors other than DNA binding, or both determine the unique features of the mechanism of action of oxaliplatin.
Collapse
Affiliation(s)
- J M Woynarowski
- Cancer Therapy and Research Center, Institute for Drug Development, San Antonio, Texas 78245, USA.
| | | | | | | | | |
Collapse
|
50
|
|