1
|
Menna P, Salvatorelli E. Humans and Rodents: The Case of hOAT4 and mOat5. J Pharmacol Exp Ther 2024; 391:375-377. [PMID: 39562012 DOI: 10.1124/jpet.124.002307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 06/10/2024] [Indexed: 11/21/2024] Open
Affiliation(s)
- Pierantonio Menna
- Department of Science and Technology for Sustainable Development and One Health (P.M.), Fondazione Policlinico (P.M.), and Department of Medicine (E.S.), University Campus Bio-Medico, Rome, Italy
| | - Emanuela Salvatorelli
- Department of Science and Technology for Sustainable Development and One Health (P.M.), Fondazione Policlinico (P.M.), and Department of Medicine (E.S.), University Campus Bio-Medico, Rome, Italy
| |
Collapse
|
2
|
Chikasue A, Taguchi K, Iwatani R, Kimura K, Okuda S, Uesugi N, Fukami K. Three Cases of Red Yeast Rice-Containing Supplement-Induced Acute Kidney Injury and Fanconi Syndrome. Am J Kidney Dis 2024:S0272-6386(24)01031-X. [PMID: 39424254 DOI: 10.1053/j.ajkd.2024.08.007] [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: 04/11/2024] [Revised: 07/25/2024] [Accepted: 08/06/2024] [Indexed: 10/21/2024]
Abstract
Japan has recently faced a public health crisis owing to "Beni Koji Choleste Help," a product marketed as a dietary supplement. It is made from red yeast rice and has cholesterol-lowering effects. Following the identification of the first case of acute kidney injury (AKI) caused by the product, 76 possibly related deaths and 492 hospitalizations have been reported in Japan. We herein demonstrate 3 cases of AKI with renal tubular acidosis and Fanconi syndrome following consumption of the product for 2 weeks to 7 months. Kidney biopsy revealed diffuse tubular injury, tubular cell flattening, loss of brush border, sparse debris, and cast formation containing periodic acid-Schiff- and silver stain-positive fluffy material in the tubular lumens with interstitial cell infiltration, together with mild fibrosis. Electron microscopy revealed fluffy materials in the tubular lumen surrounded by necrotic tubular cell debris and intracellular contents. Discontinuation of the supplement and treatment with prednisolone improved kidney function and Fanconi syndrome. Longitudinal monitoring and surveys are required to detect and prevent the progression to chronic kidney disease in people who have consumed "Beni Koji Choleste Help."
Collapse
Affiliation(s)
- Ayako Chikasue
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Division of Nephrology, Koga Hospital 21, Kurume, Fukuoka, Japan
| | - Kensei Taguchi
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Research Institute of Medical Mass Spectrometry, Kurume University School of Medicine, Kurume, Japan.
| | - Ryuji Iwatani
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Division of Nephrology, Koga Hospital 21, Kurume, Fukuoka, Japan
| | - Koki Kimura
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Division of Nephrology, Koga Hospital 21, Kurume, Fukuoka, Japan
| | - Seiya Okuda
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan; Division of Nephrology, Koga Hospital 21, Kurume, Fukuoka, Japan
| | - Noriko Uesugi
- Department of Pathology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kei Fukami
- Division of Nephrology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
3
|
Moreno-Gordaliza E, González-Nicolás MÁ, Lázaro A, Barbas C, Gómez-Gómez MM, López-Gonzálvez Á. Untargeted metabolomics analysis of serum and urine unveils the protective effect of cilastatin on altered metabolic pathways during cisplatin-induced acute kidney injury. Biochem Pharmacol 2024; 227:116435. [PMID: 39025411 DOI: 10.1016/j.bcp.2024.116435] [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: 03/15/2024] [Revised: 06/26/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Acute kidney injury (AKI) is one of the most serious complications of cisplatin anticancer therapies. Cilastatin is a highly promising nephroprotective agent to eventually enter clinical use, but its biochemical mechanism is still not fully understood. We have employed an untargeted metabolomics approach based on capillary electrophoresis mass spectrometry (CE-MS) analysis of serum and urine from an in vivo rat model, to explore the metabolic pathways involved in cisplatin-induced AKI and cilastatin nephroprotection. A total of 155 and 76 identified metabolites were found to be significantly altered during cisplatin treatment in urine and serum, respectively. Most of these altered metabolites were either partially or totally recovered by cilastatin and cisplatin co-treatment. The main metabolic pathways disturbed by cisplatin during AKI involved diverse amino acids metabolism and biosynthesis, tricarboxylic acids (TCA) cycle, nicotinate and nicotinamide metabolism, among others. Cilastatin was proved to protect diverse cisplatin-altered pathways involving metabolites related to immunomodulation, inflammation, oxidative stress and amino acid metabolism in proximal tubules. However, cisplatin-altered mitochondrial metabolism (especially, the energy-producing TCA cycle) remained largely unprotected by cilastatin, suggesting an unresolved mitochondrial direct damage. Multivariate analysis allowed effective discrimination of cisplatin-induced AKI and cilastatin renoprotection based on metabolic features. A number of potential serum and urine biomarkers could also be foreseen for cisplatin-induced AKI detection and cilastatin nephroprotection.
Collapse
Affiliation(s)
- Estefanía Moreno-Gordaliza
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Computense s/n, 28040 Madrid, Spain.
| | - M Ángeles González-Nicolás
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain
| | - Alberto Lázaro
- Renal Physiopathology Laboratory, Department of Nephrology, Instituto de Investigación Sanitaria Gregorio Marañón, Hospital General Universitario Gregorio Marañón, 28007 Madrid, Spain; Department of Physiology, School of Medicine, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Complutense s/n, 28040 Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Faculty of Pharmacy, Universidad San Pablo-CEU (CEU Universities), Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| | - M Milagros Gómez-Gómez
- Department of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid (ROR 02p0gd045), Avda. Computense s/n, 28040 Madrid, Spain
| | - Ángeles López-Gonzálvez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Department of Chemistry and Biochemistry, Faculty of Pharmacy, Universidad San Pablo-CEU (CEU Universities), Urbanización Montepríncipe, 28660 Boadilla del Monte, Madrid, Spain
| |
Collapse
|
4
|
Pisapia F, O’Brien D, Tasinato E, Garner KL, Brown CDA. Development of a Highly Differentiated Human Primary Proximal Tubule MPS Model (aProximate MPS Flow). Bioengineering (Basel) 2023; 11:7. [PMID: 38275575 PMCID: PMC10813028 DOI: 10.3390/bioengineering11010007] [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: 11/20/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/27/2024] Open
Abstract
The kidney proximal tubule (PT) mediates renal drug elimination in vivo and is a major site of drug-induced toxicity. To reliably assess drug efficacy, it is crucial to construct a model in which PT functions are replicated. Current animal studies have proven poorly predictive of human outcome. To address this, we developed a physiologically relevant micro-physiological system (MPS) model of the human PT, the aProximate MPS Flow platform (Patent No: G001336.GB). In this model, primary human PT cells (hPTCs) are subjected to fluidic media flow and a shear stress of 0.01-0.2 Pa. We observe that these cells replicate the polarity of hPTCs and exhibit a higher expression of all the key transporters of SLC22A6 (OAT1), SLC22A8 (OAT3), SLC22A2 (OCT2), SLC47A1 (MATE1), SLC22A12 (URAT1), SLC2A9 (GLUT9), ABCB1 (MDR1), ABCC2 (MRP2), LRP2 (megalin), CUBN (cubilin), compared with cells grown under static conditions. Immunofluorescence microscopy confirmed an increase in OAT1, OAT3, and cilia protein expression. Increased sensitivity to nephrotoxic protein cisplatin was observed; creatinine and FITC-albumin uptake was significantly increased under fluidic shear stress conditions. Taken together, these data suggest that growing human PT cells under media flow significantly improves the phenotype and function of hPTC monolayers and has benefits to the utility and near-physiology of the model.
Collapse
Affiliation(s)
- Francesca Pisapia
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Donovan O’Brien
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Elena Tasinato
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
- Institute of Genetic Medicine, Newcastle University, International Centre for Life, Central Parkway, Newcastle upon Tyne NE1 3BZ, UK
| | - Kathryn L. Garner
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| | - Colin D. A. Brown
- Newcells Biotech Ltd., The Biosphere, Draymans Way, Newcastle Helix, Newcastle upon Tyne NE4 5BX, UK; (D.O.); (E.T.); (C.D.A.B.)
| |
Collapse
|
5
|
Zhang Z, Liang B, Jike W, Li R, Su X, Yu J, Liu T. The Protective Effect of Marsdenia tenacissima against Cisplatin-Induced Nephrotoxicity Mediated by Inhibiting Oxidative Stress, Inflammation, and Apoptosis. Molecules 2023; 28:7582. [PMID: 38005304 PMCID: PMC10674371 DOI: 10.3390/molecules28227582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/20/2023] [Accepted: 11/09/2023] [Indexed: 11/26/2023] Open
Abstract
Cisplatin (Cis) is considered to be one of the most effective drugs for killing cancer cells and remains a first-line chemotherapeutic agent. However, Cis's multiple toxicities (especially nephrotoxicity) have limited its clinical use. Marsdenia tenacissima (Roxb.) Wight et Arn. (MT), a traditional Chinese medicine (TCM) employed extensively in China, not only enhances the antitumor effect in combination with Cis, but is also used for its detoxifying effect, as it reduces the toxic side effects of chemotherapy drugs. The aim of this study was to explore the therapeutic effect of MT on Cis-induced nephrotoxicity, along with its underlying mechanisms. In this study, liquid-mass spectrometry was performed to identify the complex composition of the extracts of MT. In addition, we measured the renal function, antioxidant enzymes, and inflammatory cytokines in mice with Cis-induced nephrotoxicity and conducted renal histology evaluations to assess renal injury. The expressions of the proteins related to antioxidant, anti-inflammatory, and apoptotic markers in renal tissues was detected by Western blotting (WB). MT treatment improved the renal function, decreased the mRNA expression of the inflammatory factors, and increased the antioxidant enzyme activity in mice. A better renal histology was observed after MT treatment. Further, MT inhibited the expression of the phospho-NFκB p65 protein/NFκB p65 protein (p-p65)/p65, phospho-inhibitor of nuclear factor kappa B kinase beta subunit/inhibitor of nuclear factor kappa B kinase beta subunit (p-IKKβ/IKKβ), Bcl-2-associated X (Bax), and Cleaved Caspase 3/Caspase 3 proteins, while the expression of nuclear factor-erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), Recombinant NADH Dehydrogenase, Quinone 1 (NQO1), and B-cell lymphoma-2 (Bcl-2) was increased. The present study showed that MT ameliorated renal injury, which mainly occurs through the regulation of the Nrf2 pathway, the NF-κB pathway, and the suppression of renal tissue apoptosis. It also suggests that MT can be used as an adjuvant to mitigate the nephrotoxicity of Cis chemotherapy.
Collapse
Affiliation(s)
- Zhiguang Zhang
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| | - Boya Liang
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| | - Wugemo Jike
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| | - Runtian Li
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| | - Xinxin Su
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| | - Jie Yu
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| | - Tongxiang Liu
- School of Pharmacy, Minzu University of China, Beijing 100081, China
- Key Laboratory of Ethnomedicine, Ministry of Education, Minzu University of China, Beijing 100081, China
| |
Collapse
|
6
|
Su J, He T, You J, Cao J, Wang Q, Cao S, Mei Q, Zeng J, Liu L. Therapeutic effect and underlying mechanism of Shenkang injection against cisplatin-induced acute kidney injury in mice. JOURNAL OF ETHNOPHARMACOLOGY 2023; 301:115805. [PMID: 36216195 DOI: 10.1016/j.jep.2022.115805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 06/16/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Shenkang injection (SKI), a Chinese patent medicine injection, has been approved for the treatment of chronic kidney disease (CKD) due to its definite clinical therapeutic efficacy. However, the effect and associated underlying mechanism of Shenkang injection against cisplatin (CDDP)-induced acute kidney injury (AKI) has not yet been well elucidated. AIM OF THE STUDY This study aims to investigate the therapeutic effect and associated underlying mechanism of Shenkang injection against CDDP-induced AKI. MATERIALS AND METHODS We established a CDDP-induced AKI mouse model to evaluate renal function by biochemical markers measurement and to observe histopathological alterations by haemotoxylin and eosin (HE)-staining sections of renal. In addition, the distribution of representative components of SKI in the kidneys of mice was evaluated by liquid chromatography tandem mass spectrometry (LC-MS/MS). Furthermore, the degree of oxidative stress and inflammation were assessed by detecting the levels of inflammatory cytokines and oxidants, while the related mechanisms were elucidated by network pharmacology. RESULTS CDDP could induce excessive inflammation and severe injury to the kidneys of mice. However, SKI significantly ameliorated the kidney damages and improved the renal function by reducing the levels of renal function markers (SCr, BUN and urine protein), and inhibiting the production of inflammatory cytokines IL-34, IL-6 and TNF-α. SKI repaired oxidative balance through up-regulation of antioxidants SOD and GSH and down-regulated oxidants MDA. Moreover, 4 components from SKI were detected in the kidney by LC-MS/MS quantification. In addition, pharmacology network indicated the PI3K/AKT, TNF, MAPK, and p53 were the possible signaling pathways for the therapeutic effect of SKI against CDDP-induced AKI, which were related to inflammation, oxidative stress and apoptosis. CONCLUSION In the present study, we for the first time demonstrated that SKI alleviates CDDP-induced nephrotoxicity by antioxidant and anti-inflammation via regulating PI3K/AKT, MAPK, TNF, and p53 signaling pathways. The study may provide a scientific rationale for the clinical indication of SKI.
Collapse
Affiliation(s)
- Jiahan Su
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Luzhou New Drug Evaluation and Research Center, Luzhou, Sichuan, 646000, China
| | - Tingting He
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Luzhou New Drug Evaluation and Research Center, Luzhou, Sichuan, 646000, China
| | - Jing You
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; The People's Hospital of DaZhu, Dazhou, Sichuan, 635000, China
| | - Jingjie Cao
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qianru Wang
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shousong Cao
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qibing Mei
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China; Luzhou New Drug Evaluation and Research Center, Luzhou, Sichuan, 646000, China
| | - Jing Zeng
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Li Liu
- Department of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
7
|
Selim MS, Kassem AB, El-Bassiouny NA, Salahuddin A, Abu El-Ela RY, Hamza MS. Polymorphic renal transporters and cisplatin's toxicity in urinary bladder cancer patients: current perspectives and future directions. Med Oncol 2023; 40:80. [PMID: 36650399 PMCID: PMC9845168 DOI: 10.1007/s12032-022-01928-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/10/2022] [Indexed: 01/19/2023]
Abstract
Urinary bladder cancer (UBC) holds a potentially profound social burden and affects over 573,278 new cases annually. The disease's primary risk factors include occupational tobacco smoke exposure and inherited genetic susceptibility. Over the past 30 years, a number of treatment modalities have emerged, including cisplatin, a platinum molecule that has demonstrated effectiveness against UBC. Nevertheless, it has severe dose-limiting side effects, such as nephrotoxicity, among others. Since intracellular accumulation of platinum anticancer drugs is necessary for cytotoxicity, decreased uptake or enhanced efflux are the root causes of platinum resistance and response failure. Evidence suggests that genetic variations in any transporter involved in the entry or efflux of platinum drugs alter their kinetics and, to a significant extent, determine patients' responses to them. This review aims to consolidate and describe the major transporters and their polymorphic variants in relation to cisplatin-induced toxicities and resistance in UBC patients. We concluded that the efflux transporters ABCB1, ABCC2, SLC25A21, ATP7A, and the uptake transporter OCT2, as well as the organic anion uptake transporters OAT1 and OAT2, are linked to cisplatin accumulation, toxicity, and resistance in urinary bladder cancer patients. While suppressing the CTR1 gene's expression reduced cisplatin-induced nephrotoxicity and ototoxicity, inhibiting the expression of the MATE1 and MATE2-K genes has been shown to increase cisplatin's nephrotoxicity and resistance. The roles of ABCC5, ABCA8, ABCC10, ABCB10, ABCG1, ATP7B, ABCG2, and mitochondrial SLC25A10 in platinum-receiving urinary bladder cancer patients should be the subject of further investigation.
Collapse
Affiliation(s)
- Mohamed S Selim
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt.
| | - Amira B Kassem
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Noha A El-Bassiouny
- Clinical Pharmacy & Pharmacy Practice Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Ahmad Salahuddin
- Biochemistry Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
- Biochemistry Department, Scientific Research Center, Al-Ayen University, Thi-Qar, Iraq
| | - Raghda Y Abu El-Ela
- Medical Oncology Department, Faculty of Medicine, Fayoum University, Fayoum, Egypt
| | - Marwa Samir Hamza
- Clinical Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, Cairo, Egypt
| |
Collapse
|
8
|
Cisplatin nephrotoxicity: new insights and therapeutic implications. Nat Rev Nephrol 2023; 19:53-72. [PMID: 36229672 DOI: 10.1038/s41581-022-00631-7] [Citation(s) in RCA: 139] [Impact Index Per Article: 139.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2022] [Indexed: 11/08/2022]
Abstract
Cisplatin is an effective chemotherapeutic agent for various solid tumours, but its use is limited by adverse effects in normal tissues. In particular, cisplatin is nephrotoxic and can cause acute kidney injury and chronic kidney disease. Preclinical studies have provided insights into the cellular and molecular mechanisms of cisplatin nephrotoxicity, which involve intracellular stresses including DNA damage, mitochondrial pathology, oxidative stress and endoplasmic reticulum stress. Stress responses, including autophagy, cell-cycle arrest, senescence, apoptosis, programmed necrosis and inflammation have key roles in the pathogenesis of cisplatin nephrotoxicity. In addition, emerging evidence suggests a contribution of epigenetic changes to cisplatin-induced acute kidney injury and chronic kidney disease. Further research is needed to determine how these pathways are integrated and to identify the cell type-specific roles of critical molecules involved in regulated necrosis, inflammation and epigenetic modifications in cisplatin nephrotoxicity. A number of potential therapeutic targets for cisplatin nephrotoxicity have been identified. However, the effects of renoprotective strategies on the efficacy of cisplatin chemotherapy needs to be thoroughly evaluated. Further research using tumour-bearing animals, multi-omics and genome-wide association studies will enable a comprehensive understanding of the complex cellular and molecular mechanisms of cisplatin nephrotoxicity and potentially lead to the identification of specific targets to protect the kidney without compromising the chemotherapeutic efficacy of cisplatin.
Collapse
|
9
|
Chen Y, Lu S, Zhang Y, Chen B, Zhou H, Jiang H. Examination of the emerging role of transporters in the assessment of nephrotoxicity. Expert Opin Drug Metab Toxicol 2022; 18:787-804. [PMID: 36420583 DOI: 10.1080/17425255.2022.2151892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
INTRODUCTION The kidney is vulnerable to various injuries based on its function in the elimination of many xenobiotics, endogenous substances and metabolites. Since transporters are critical for the renal elimination of those substances, it is urgent to understand the emerging role of transporters in nephrotoxicity. AREAS COVERED This review summarizes the contribution of major renal transporters to nephrotoxicity induced by some drugs or toxins; addresses the role of transporter-mediated endogenous metabolic disturbances in nephrotoxicity; and discusses the advantages and disadvantages of in vitro models based on transporter expression and function. EXPERT OPINION Due to the crucial role of transporters in the renal disposition of xenobiotics and endogenous substances, it is necessary to further elucidate their renal transport mechanisms and pay more attention to the underlying relationship between the transport of endogenous substances and nephrotoxicity. Considering the species differences in the expression and function of transporters, and the low expression of transporters in general cell models, in vitro humanized models, such as humanized 3D organoids, shows significant promise in nephrotoxicity prediction and mechanism study.
Collapse
Affiliation(s)
- Yujia Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Shuanghui Lu
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Yingqiong Zhang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, P.R. China
| | - Binxin Chen
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China
| | - Hui Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Jinhua Institute of Zhejiang University, Jinhua, P.R. China
| | - Huidi Jiang
- Institute of Drug Metabolism and Pharmaceutical Analysis, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, P.R. China.,Jinhua Institute of Zhejiang University, Jinhua, P.R. China
| |
Collapse
|
10
|
Thind AS, Ashford B, Strbenac D, Mitchell J, Lee J, Mueller SA, Minaei E, Perry JR, Ch’ng S, Iyer NG, Clark JR, Gupta R, Ranson M. Whole genome analysis reveals the genomic complexity in metastatic cutaneous squamous cell carcinoma. Front Oncol 2022; 12:919118. [PMID: 35982973 PMCID: PMC9379253 DOI: 10.3389/fonc.2022.919118] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 06/27/2022] [Indexed: 12/13/2022] Open
Abstract
Metastatic cutaneous squamous cell carcinoma (CSCC) is a highly morbid disease requiring radical surgery and adjuvant therapy, which is associated with a poor prognosis. Yet, compared to other advanced malignancies, relatively little is known of the genomic landscape of metastatic CSCC. We have previously reported the mutational signatures and mutational patterns of CCCTC-binding factor (CTCF) regions in metastatic CSCC. However, many other genomic components (indel signatures, non-coding drivers, and structural variants) of metastatic CSCC have not been reported. To this end, we performed whole genome sequencing on lymph node metastases and blood DNA from 25 CSCC patients with regional metastases of the head and neck. We designed a multifaceted computational analysis at the whole genome level to provide a more comprehensive perspective of the genomic landscape of metastatic CSCC. In the non-coding genome, 3′ untranslated region (3′UTR) regions of EVC (48% of specimens), PPP1R1A (48% of specimens), and ABCA4 (20% of specimens) along with the tumor-suppressing long non-coding RNA (lncRNA) LINC01003 (64% of specimens) were significantly functionally altered (Q-value < 0.05) and represent potential non-coding biomarkers of CSCC. Recurrent copy number loss in the tumor suppressor gene PTPRD was observed. Gene amplification was much less frequent, and few genes were recurrently amplified. Single nucleotide variants driver analyses from three tools confirmed TP53 and CDKN2A as recurrently mutated genes but also identified C9 as a potential novel driver in this disease. Furthermore, indel signature analysis highlighted the dominance of ID signature 13 (ID13) followed by ID8 and ID9. ID9 has previously been shown to have no association with skin melanoma, unlike ID13 and ID8, suggesting a novel pattern of indel variation in metastatic CSCC. The enrichment analysis of various genetically altered candidates shows enrichment of “TGF-beta regulation of extracellular matrix” and “cell cycle G1 to S check points.” These enriched terms are associated with genetic instability, cell proliferation, and migration as mechanisms of genomic drivers of metastatic CSCC.
Collapse
Affiliation(s)
- Amarinder Singh Thind
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
| | - Bruce Ashford
- School of Medicine, University of Wollongong, Wollongong, NSW, Australia
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- Illawarra Shoalhaven Local Health District, Wollongong, NSW, Australia
- *Correspondence: Bruce Ashford,
| | - Dario Strbenac
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
| | - Jenny Mitchell
- Illawarra Shoalhaven Local Health District, Wollongong, NSW, Australia
| | - Jenny Lee
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
- Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Simon A. Mueller
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
- Department of Otorhinolaryngology, Head and Neck Surgery, Zurich University Hospital and University of Zurich, Zurich, Switzerland
| | - Elahe Minaei
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Jay R. Perry
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| | - Sydney Ch’ng
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
| | - N. Gopalakrishna Iyer
- Department of Head and Neck Surgery, National Cancer Center, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | - Jonathan R. Clark
- Sydney Medical School, Faculty of Medicine and Health, The University of Sydney, NSW, Australia
- Sydney Head and Neck Cancer Institute, Chris O’Brien Lifehouse, Sydney, NSW, Australia
- Royal Prince Alfred Institute of Academic Surgery, Sydney Local Health District, Sydney, NSW, Australia
| | - Ruta Gupta
- Anatomical Pathology, Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Marie Ranson
- Illawarra Health and Medical Research Institute, Wollongong, NSW, Australia
- School of Chemistry and Molecular Bioscience, University of Wollongong, Wollongong, NSW, Australia
| |
Collapse
|
11
|
Hafey MJ, Aleksunes LM, Bridges CC, Brouwer KR, Chien HC, Leslie EM, Hu S, Li Y, Shen J, Sparreboom A, Sprowl J, Tweedie D, Lai Y. Transporters and Toxicity: Insights from the International Transporter Consortium Workshop 4. Clin Pharmacol Ther 2022; 112:527-539. [PMID: 35546260 DOI: 10.1002/cpt.2638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 04/30/2022] [Indexed: 12/29/2022]
Abstract
Over the last decade, significant progress been made in elucidating the role of membrane transporters in altering drug disposition, with important toxicological consequences due to changes in localized concentrations of compounds. The topic of "Transporters and Toxicity" was recently highlighted as a scientific session at the International Transporter Consortium (ITC) Workshop 4 in 2021. The current white paper is not intended to be an extensive review on the topic of transporters and toxicity but an opportunity to highlight aspects of the role of transporters in various toxicities with clinically relevant implications as covered during the session. This includes a review of the role of solute carrier transporters in anticancer drug-induced organ injury, transporters as key players in organ barrier function, and the role of transporters in metal/metalloid toxicity.
Collapse
Affiliation(s)
- Michael J Hafey
- ADME and Discovery Toxicology, Merck & Co., Inc., Rahway, New Jersey, USA
| | - Lauren M Aleksunes
- Department of Pharmacology and Toxicology, Rutgers University, Piscataway, New Jersey, USA
| | - Christy C Bridges
- Department of Biomedical Sciences, Mercer University School of Medicine, Macon, Georgia, USA
| | | | - Huan-Chieh Chien
- Pharmacokinetics and Drug Metabolism, Amgen, Inc., South San Francisco, California, USA
| | - Elaine M Leslie
- Departments of Physiology and Lab Med and Path, Membrane Protein Disease Research Group, University of Alberta, Edmonton, Alberta, Canada
| | - Shuiying Hu
- Division of Outcomes and Translational Sciences, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Yang Li
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jinshan Shen
- Relay Therapeutics, Cambridge, Massachusetts, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, Ohio, USA
| | - Jason Sprowl
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University at Buffalo, Buffalo, New York, USA
| | | | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California, USA
| |
Collapse
|
12
|
Liu C, Zhou S, Bai W, Shi L, Li X. Protective effect of food derived nutrients on cisplatin nephrotoxicity and its mechanism. Food Funct 2022; 13:4839-4860. [PMID: 35416186 DOI: 10.1039/d1fo04391a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Platinum-based metal complexes, especially cisplatin (cis-diamminedichloroplatinum II, CDDP), possess strong anticancer properties and a broad anticancer spectrum. However, the clinical application of CDDP has been limited by its side effects including nephrotoxicity, ototoxicity, and neurotoxicity. Furthermore, the therapeutic effects of current clinical protocols are imperfect. Accordingly, it is essential to identify key targets and effective clinical protocols to restrict CDDP-induced nephrotoxicity. Herein, we first analyzed the relevant molecular mechanisms during the process of CDDP-induced nephrotoxicity including oxidative stress, apoptosis, and inflammation. Evidence from current studies was collected and potential targets and clinical protocols are summarized. The evidence indicates an efficacious role of nutrition-based substances in CDDP-induced renal injury.
Collapse
Affiliation(s)
- Chaofan Liu
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Sajin Zhou
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Weibin Bai
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Lei Shi
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| | - Xiaoling Li
- Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
13
|
Abstract
Medications are a common cause of AKI especially for patients admitted to hospital wards and the intensive care unit. Although drug-related kidney injury occurs through different mechanisms, this review will focus on three specific types of tubulointerstitial injury. Direct acute tubular injury develops from several medications, which are toxic to various cellular functions. Their excretory pathways through the proximal tubules contribute further to AKI. Drug-induced AKI may also develop through induction of inflammation within the tubulointerstitium. Medications can elicit a T cell-mediated immune response that promotes the development of acute interstitial nephritis leading to AKI. Although less common, a third pathway to kidney injury results from the insolubility of drugs in the urine leading to their precipitation as crystals within distal tubular lumens, causing a crystalline-related AKI. Intratubular obstruction, direct tubular injury, and localized inflammation lead to AKI. Clinicians should be familiar with the pathogenesis and clinical-pathologic manifestations of these forms of kidney injury. Prevention and treatment of AKI relies on understanding the pathogenesis and judiciously using these agents in settings where AKI risk is high.
Collapse
Affiliation(s)
- Mark A Perazella
- Section of Nephrology, Yale University School of Medicine, New Haven, Connecticut .,Veteran's Affairs Medical Center, West Haven, Connecticut
| | - Mitchell H Rosner
- Division of Nephrology, University of Virginia Health System, Charlottesville, Virginia
| |
Collapse
|
14
|
Kidney Transporters Drug Discovery, Development, and Safety. CURRENT OPINION IN TOXICOLOGY 2022. [DOI: 10.1016/j.cotox.2022.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
15
|
Jain A, Huang R, Lee J, Jawa N, Lim YJ, Guron M, Abish S, Boutros PC, Brudno M, Carleton B, Cuvelier GDE, Gunaratnam L, Ho C, Adeli K, Kuruvilla S, Lajoie G, Liu G, Nathan PC, Rod Rassekh S, Rieder M, Waikar SS, Welch SA, Weir MA, Winquist E, Wishart DS, Zorzi AP, Blydt-Hansen T, Zappitelli M, Urquhart B. A Canadian Study of Cisplatin Metabolomics and Nephrotoxicity (ACCENT): A Clinical Research Protocol. Can J Kidney Health Dis 2021; 8:20543581211057708. [PMID: 34820133 PMCID: PMC8606978 DOI: 10.1177/20543581211057708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 09/18/2021] [Indexed: 11/15/2022] Open
Abstract
Background: Cisplatin, a chemotherapy used to treat solid tumors, causes acute kidney injury (AKI), a known risk factor for chronic kidney disease and mortality. AKI diagnosis relies on biomarkers which are only measurable after kidney damage has occurred and functional impairment is apparent; this prevents timely AKI diagnosis and treatment. Metabolomics seeks to identify metabolite patterns involved in cell tissue metabolism related to disease or patient factors. The A Canadian study of Cisplatin mEtabolomics and NephroToxicity (ACCENT) team was established to harness the power of metabolomics to identify novel biomarkers that predict risk and discriminate for presence of cisplatin nephrotoxicity, so that early intervention strategies to mitigate onset and severity of AKI can be implemented. Objective: Describe the design and methods of the ACCENT study which aims to identify and validate metabolomic profiles in urine and serum associated with risk for cisplatin-mediated nephrotoxicity in children and adults. Design: Observational prospective cohort study. Setting: Six Canadian oncology centers (3 pediatric, 1 adult and 2 both). Patients: Three hundred adults and 300 children planned to receive cisplatin therapy. Measurements: During two cisplatin infusion cycles, serum and urine will be measured for creatinine and electrolytes to ascertain AKI. Many patient and disease variables will be collected prospectively at baseline and throughout therapy. Metabolomic analyses of serum and urine will be done using mass spectrometry. An untargeted metabolomics approach will be used to analyze serum and urine samples before and after cisplatin infusions to identify candidate biomarkers of cisplatin AKI. Candidate metabolites will be validated using an independent cohort. Methods: Patients will be recruited before their first cycle of cisplatin. Blood and urine will be collected at specified time points before and after cisplatin during the first infusion and an infusion later during cancer treatment. The primary outcome is AKI, defined using a traditional serum creatinine-based definition and an electrolyte abnormality-based definition. Chart review 3 months after cisplatin therapy end will be conducted to document kidney health and survival. Limitations: It may not be possible to adjust for all measured and unmeasured confounders when evaluating prediction of AKI using metabolite profiles. Collection of data across multiple sites will be a challenge. Conclusions: ACCENT is the largest study of children and adults treated with cisplatin and aims to reimagine the current model for AKI diagnoses using metabolomics. The identification of biomarkers predicting and detecting AKI in children and adults treated with cisplatin can greatly inform future clinical investigations and practices.
Collapse
Affiliation(s)
- Anshika Jain
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada.,Temerty Faculty of Medicine, University of Toronto, ON, Canada
| | - Ryan Huang
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
| | - Jasmine Lee
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada
| | - Natasha Jawa
- Division of Nephrology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Yong Jin Lim
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Mike Guron
- Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, Canada
| | - Sharon Abish
- Division of Hematology and Oncology, Montreal Children's Hospital, McGill University Health Centre, Montreal, QC, Canada
| | - Paul C Boutros
- Computational Biology Program, Ontario Institute for Cancer Research, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, ON, Canada
| | - Michael Brudno
- Department of Computer Science, University of Toronto, ON, Canada.,Canada Centre for Computational Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bruce Carleton
- Department of Pediatrics, The University of British Columbia, Vancouver, Canada.,Pharmaceutical Outcomes Programme, BC Children's Hospital, Vancouver, Canada.,BC Children's Hospital Research Institute, Vancouver, Canada
| | | | - Lakshman Gunaratnam
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Cheryl Ho
- Medical Oncology, BC Cancer, The University of British Columbia, Vancouver, Canada
| | - Khosrow Adeli
- Molecular Medicine, Research Institute, The Hospital for Sick Children, Toronto, ON, Canada.,University of Toronto, ON, Canada, Canada
| | - Sara Kuruvilla
- Division of Medical Oncology, Department of Oncology, Western University, London, ON, Canada
| | - Giles Lajoie
- Department of Biochemistry, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Paul C Nathan
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Shahrad Rod Rassekh
- Department of Pediatrics, Division of Hematology/Oncology/Bone Marrow Transplantation, BC Children's Hospital, The University of British Columbia, Vancouver, Canada
| | - Michael Rieder
- Department of Pediatrics, Western University, London, ON, Canada
| | - Sushrut S Waikar
- Section of Nephrology, Boston University School of Medicine, MA, USA.,Boston Medical Center, MA, USA
| | - Stephen A Welch
- Division of Medical Oncology, Department of Oncology, Western University, London, ON, Canada
| | - Matthew A Weir
- Division of Nephrology, Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Eric Winquist
- Division of Medical Oncology, Department of Oncology, Western University, London, ON, Canada
| | - David S Wishart
- Department of Biochemistry, University of Alberta, Edmonton, Canada
| | - Alexandra P Zorzi
- Division of Hematology/Oncology, Department of Pediatrics, Children's Hospital, Western University, London, ON, Canada
| | - Tom Blydt-Hansen
- Department of Pediatrics, BC Children's Hospital, The University of British Columbia, Vancouver, Canada
| | - Michael Zappitelli
- Child Health Evaluative Sciences, The Hospital for Sick Children, Toronto, ON, Canada.,Division of Nephrology, Department of Pediatrics, The Hospital for Sick Children, Toronto, ON, Canada
| | - Bradley Urquhart
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| |
Collapse
|
16
|
Targeting DNA Damage Response and Repair to Enhance Therapeutic Index in Cisplatin-Based Cancer Treatment. Int J Mol Sci 2021; 22:ijms22158199. [PMID: 34360968 PMCID: PMC8347825 DOI: 10.3390/ijms22158199] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 02/06/2023] Open
Abstract
Platinum-based chemotherapies, such as cisplatin, play a large role in cancer treatment. The development of resistance and treatment toxicity creates substantial barriers to disease control, yet. To enhance the therapeutic index of cisplatin-based chemotherapy, it is imperative to circumvent resistance and toxicity while optimizing tumor sensitization. One of the primary mechanisms by which cancer cells develop resistance to cisplatin is through upregulation of DNA repair pathways. In this review, we discuss the DNA damage response in the context of cisplatin-induced DNA damage. We describe the proteins involved in the pathways and their roles in resistance development. Common biomarkers for cisplatin resistance and their utilization to improve patient risk stratification and treatment personalization are addressed. Finally, we discuss some of the current treatments and future strategies to circumvent the development of cisplatin resistance.
Collapse
|
17
|
Pasquariello KZ, Dey JM, Sprowl JA. Current Understanding of Membrane Transporters as Regulators or Targets for Cisplatin-Induced Hearing Loss. Mol Pharmacol 2021; 100:348-355. [PMID: 34330821 DOI: 10.1124/molpharm.121.000274] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/22/2021] [Indexed: 11/22/2022] Open
Abstract
Cisplatin is a platinum-based drug which remains among the most efficacious anticancer treatment options. Unfortunately, use of cisplatin is hindered by dose-limiting toxicities, including irreversible hearing loss, which can grossly affect patient quality of life. Cisplatin-induced ototoxicity is the result of cochlear hair cell damage through a mechanism that is poorly understood. However, cisplatin cytotoxicity is reliant on intracellular accumulation, a process that is largely dependent on the presence of particular membrane transporters. This review will provide an update on our current understanding of the various transporters known to be involved in the disposition and cytotoxicity of platinum drugs or their metabolites, as well as their role in mediating cisplatin-induced hearing loss. We also provide a summary of the successes and opportunities in therapeutically targeting membrane transporters to alleviate platinum-induced hearing loss. Moreover, we describe how this approach could be used to reduce the severity or onset of other adverse events associated with exposure to various forms of platinum drugs, without diminishing anti-tumor efficacy. Significance Statement Cisplatin-induced hearing loss is a dose limiting and irreversible adverse event with no current preventative or curative treatment measures. Pharmacological targeting of membrane transporters that regulate platinum uptake into cochlear hair cells, if conducted appropriately, may alleviate this devastating side effect and could be applied to alleviate other platinum-induced toxicities.
Collapse
Key Words
- Uptake transporters (OATP, OAT, OCT, PEPT, MCT, NTCP, ASBT, etc.)
- cancer chemotherapy
- efflux transporters (P-gp, BCRP, MRP, MATE, BSEP, etc)
- ototoxicity
Collapse
Affiliation(s)
| | | | - Jason A Sprowl
- School of Pharmacy, University of Buffalo, United States
| |
Collapse
|
18
|
Lebel A, Chui H, McMahon KR, Lim YJ, Macri J, Wang S, Devarajan P, Blydt-Hansen TD, Zappitelli M, Urquhart BL. Association of Urine Platinum With Acute Kidney Injury in Children Treated With Cisplatin for Cancer. J Clin Pharmacol 2021; 61:871-880. [PMID: 33599997 PMCID: PMC8283690 DOI: 10.1002/jcph.1839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/15/2021] [Indexed: 01/19/2023]
Abstract
Cisplatin is a chemotherapeutic agent highly excreted in urine and known to cause acute kidney injury (AKI). As AKI diagnosis by serum creatinine (SCr) is usually delayed, endeavors for finding early AKI biomarkers continue. This study aims to determine if urine platinum (UP) concentration 24 hours after cisplatin infusion is associated with AKI, and to evaluate the association between urine platinum and tubular damage biomarkers: neutrophil gelatinase-associated lipocalin (NGAL) and kidney injury molecule-1 (KIM-1). Children treated with cisplatin in 12 Canadian centers (April 2013 to December 2017) were included. Urine from the morning after the first cisplatin infusion of the first or second cisplatin cycle was measured for urine platinum, NGAL, and KIM-1. SCr and serum electrolytes were used to detect AKI by either SCr elevation or urinary electrolyte wasting (potassium, magnesium, phosphate). The associations of urine platinum with AKI, NGAL, and KIM-1 were assessed. A total of 115 participants (54% boys, median age, 8.5 years; interquartile range, 4.0-13.4) were included, of which 29 (25%) and 105 (91%) developed AKI defined by SCr and electrolyte criteria, respectively. Higher urine platinum was associated with higher cisplatin dose (Spearman rho, 0.21) and with younger age (Spearman rho, -0.33). Urine platinum was not associated with postinfusion AKIor KIM-1, but was weakly associated with NGAL, particularly in participants without SCr AKI (Pearson's r, 0.22). Urine platinum may be a marker of mild tubular injury but is not likely to be a useful biomarker of clinically evident AKI.
Collapse
Affiliation(s)
- Asaf Lebel
- Department of Pediatrics, Division of Nephrology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Hayton Chui
- Department of Pediatrics, Division of Nephrology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Kelly R. McMahon
- Division of Nephrology, Department of Pediatrics, Montreal Children's Hospital, McGill University Health Centre, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Yong Jin Lim
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Joseph Macri
- Hamilton Regional Laboratory Medicine Program, Hamilton, Ontario, Canada
| | - Stella Wang
- Department of Pediatrics, Division of Nephrology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Prasad Devarajan
- Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Tom D. Blydt-Hansen
- British Columbia Children's Hospital, Division of Pediatric Nephrology, Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Michael Zappitelli
- Department of Pediatrics, Division of Nephrology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Bradley L. Urquhart
- Schulich School of Medicine and Dentistry, Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| |
Collapse
|
19
|
McSweeney KR, Gadanec LK, Qaradakhi T, Ali BA, Zulli A, Apostolopoulos V. Mechanisms of Cisplatin-Induced Acute Kidney Injury: Pathological Mechanisms, Pharmacological Interventions, and Genetic Mitigations. Cancers (Basel) 2021; 13:1572. [PMID: 33805488 PMCID: PMC8036620 DOI: 10.3390/cancers13071572] [Citation(s) in RCA: 164] [Impact Index Per Article: 54.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/19/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023] Open
Abstract
Administration of the chemotherapeutic agent cisplatin leads to acute kidney injury (AKI). Cisplatin-induced AKI (CIAKI) has a complex pathophysiological map, which has been linked to cellular uptake and efflux, apoptosis, vascular injury, oxidative and endoplasmic reticulum stress, and inflammation. Despite research efforts, pharmaceutical interventions, and clinical trials spanning over several decades, a consistent and stable pharmacological treatment option to reduce AKI in patients receiving cisplatin remains unavailable. This has been predominately linked to the incomplete understanding of CIAKI pathophysiology and molecular mechanisms involved. Herein, we detail the extensively known pathophysiology of cisplatin-induced nephrotoxicity that manifests and the variety of pharmacological and genetic alteration studies that target them.
Collapse
|
20
|
Naji-Talakar S, Sharma S, Martin LA, Barnhart D, Prasad B. Potential implications of DMET ontogeny on the disposition of commonly prescribed drugs in neonatal and pediatric intensive care units. Expert Opin Drug Metab Toxicol 2021; 17:273-289. [PMID: 33256492 PMCID: PMC8346204 DOI: 10.1080/17425255.2021.1858051] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/27/2020] [Indexed: 10/22/2022]
Abstract
Introduction: Pediatric patients, especially neonates and infants, are more susceptible to adverse drug events as compared to adults. In particular, immature small molecule drug metabolism and excretion can result in higher incidences of pediatric toxicity than adults if the pediatric dose is not adjusted.Area covered: We reviewed the top 29 small molecule drugs prescribed in neonatal and pediatric intensive care units and compiled the mechanisms of their metabolism and excretion. The ontogeny of Phase I and II drug metabolizing enzymes and transporters (DMETs), particularly relevant to these drugs, are summarized. The potential effects of DMET ontogeny on the metabolism and excretion of the top pediatric drugs were predicted. The current regulatory requirements and recommendations regarding safe and effective use of drugs in children are discussed. A few representative examples of the use of ontogeny-informed physiologically based pharmacokinetic (PBPK) models are highlighted.Expert opinion: Empirical prediction of pediatric drug dosing based on body weight or body-surface area from the adult parameters can be inaccurate because DMETs are not mature in children and the age-dependent maturation of these proteins is different. Ontogeny-informed-PBPK modeling provides a better alternative to predict the pharmacokinetics of drugs in children.
Collapse
Affiliation(s)
- Siavosh Naji-Talakar
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| | - Sheena Sharma
- Pediatrics and Neonatology, Providence Sacred Heart Medical Center and Children’s Hospital, Spokane, WA, USA
| | - Leslie A. Martin
- Pediatrics and Neonatology, Providence Sacred Heart Medical Center and Children’s Hospital, Spokane, WA, USA
| | - Derek Barnhart
- Pediatrics and Neonatology, Providence Sacred Heart Medical Center and Children’s Hospital, Spokane, WA, USA
| | - Bhagwat Prasad
- Department of Pharmaceutical Sciences, Washington State University, Spokane, WA, USA
| |
Collapse
|
21
|
The Role of Nucleotide Excision Repair in Cisplatin-Induced Peripheral Neuropathy: Mechanism, Prevention, and Treatment. Int J Mol Sci 2021; 22:ijms22041975. [PMID: 33671279 PMCID: PMC7921932 DOI: 10.3390/ijms22041975] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/06/2023] Open
Abstract
Platinum-based chemotherapy-induced peripheral neuropathy (CIPN) is one of the most common dose-limiting effects of cancer treatment and results in dose reduction and discontinuation of life-saving chemotherapy. Its debilitating effects are often permanent and lead to lifelong impairment of quality of life in cancer patients. While the mechanisms underlying the toxicity are not yet fully defined, dorsal root ganglia sensory neurons play an integral role in symptom development. DNA-platinum adducts accumulate in these cells and inhibit normal cellular function. Nucleotide excision repair (NER) is integral to the repair of platinum adducts, and proteins involved in its mechanism serve as potential targets for future therapeutics. This review aims to highlight NER’s role in cisplatin-induced peripheral neuropathy, summarize current clinical approaches to the toxicity, and discuss future perspectives for the prevention and treatment of CIPN.
Collapse
|
22
|
Uptake Transporters of the SLC21, SLC22A, and SLC15A Families in Anticancer Therapy-Modulators of Cellular Entry or Pharmacokinetics? Cancers (Basel) 2020; 12:cancers12082263. [PMID: 32806706 PMCID: PMC7464370 DOI: 10.3390/cancers12082263] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/16/2020] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Solute carrier transporters comprise a large family of uptake transporters involved in the transmembrane transport of a wide array of endogenous substrates such as hormones, nutrients, and metabolites as well as of clinically important drugs. Several cancer therapeutics, ranging from chemotherapeutics such as topoisomerase inhibitors, DNA-intercalating drugs, and microtubule binders to targeted therapeutics such as tyrosine kinase inhibitors are substrates of solute carrier (SLC) transporters. Given that SLC transporters are expressed both in organs pivotal to drug absorption, distribution, metabolism, and elimination and in tumors, these transporters constitute determinants of cellular drug accumulation influencing intracellular drug concentration required for efficacy of the cancer treatment in tumor cells. In this review, we explore the current understanding of members of three SLC families, namely SLC21 (organic anion transporting polypeptides, OATPs), SLC22A (organic cation transporters, OCTs; organic cation/carnitine transporters, OCTNs; and organic anion transporters OATs), and SLC15A (peptide transporters, PEPTs) in the etiology of cancer, in transport of chemotherapeutic drugs, and their influence on efficacy or toxicity of pharmacotherapy. We further explore the idea to exploit the function of SLC transporters to enhance cancer cell accumulation of chemotherapeutics, which would be expected to reduce toxic side effects in healthy tissue and to improve efficacy.
Collapse
|
23
|
Neamatallah T, El-Shitany N, Abbas A, Eid BG, Harakeh S, Ali S, Mousa S. Nano Ellagic Acid Counteracts Cisplatin-Induced Upregulation in OAT1 and OAT3: A Possible Nephroprotection Mechanism. Molecules 2020; 25:E3031. [PMID: 32630784 PMCID: PMC7411712 DOI: 10.3390/molecules25133031] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/29/2020] [Accepted: 07/01/2020] [Indexed: 02/07/2023] Open
Abstract
Cisplatin is an anticancer drug commonly used for solid tumors. However, it causes nephrotoxicity. OAT1 and OAT3 are organic anion transporters known to contribute to the uptake of cisplatin into renal tubular cells. The present study was designed to examine the protective role of ellagic acid nanoformulation (ellagic acid nano) on cisplatin-induced nephrotoxicity in rats, and the role of OAT1/OAT3 in this effect. Four groups of male Wistar rats were used (n = 6): (1) control, (2) cisplatin (7.5 mg/kg single dose, intraperitoneal), (3) cisplatin + ellagic acid nano (1 mg/kg), and (4) cisplatin + ellagic acid nano (2 mg/kg). Nephrotoxic rats treated with ellagic acid nano exhibited a significant reduction in elevated serum creatinine, urea, and oxidative stress marker, malondialdehyde (MDA). Additionally, ellagic acid nano restored renal glutathione (GSH), superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx). Ellagic acid nano improved the histopathological changes induced by cisplatin, such as tubular dilatation, necrosis, and degeneration. Interestingly, OAT1 and OAT3 showed significantly lower expression at both mRNA and protein levels following ellagic acid nano treatment relative to the cisplatin-exposed group. These findings reveal a potential inhibitory role of ellagic acid antioxidant on OAT1 and OAT3 expression and thus explains its nephroprotective effect against cisplatin nephrotoxicity.
Collapse
Affiliation(s)
- Thikryat Neamatallah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (T.N.); (B.G.E.)
| | - Nagla El-Shitany
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (T.N.); (B.G.E.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tanta University, Tanta 31511, Egypt
| | - Aymn Abbas
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.); (S.H.)
- Biotechnology Research Laboratories, Gastroenterology Surgery Center, Mansoura University, Mansoura 35511, Egypt
| | - Basma G. Eid
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (T.N.); (B.G.E.)
| | - Steve Harakeh
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (A.A.); (S.H.)
- Yousef Abdullatif Jameel Chair of Prophetic Medicine Application, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Soad Ali
- Anatomy Department of Cytology and Histology, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shaker Mousa
- Yousef Abdullatif Jameel Chair of Prophetic Medicine Application, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| |
Collapse
|
24
|
Scialis RJ, Ghanem CI, Manautou JE. The modulation of transcriptional expression and inhibition of multidrug resistance associated protein 4 (MRP4) by analgesics and their primary metabolites. Curr Res Toxicol 2020; 1:34-41. [PMID: 34345835 PMCID: PMC8320619 DOI: 10.1016/j.crtox.2020.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/02/2020] [Accepted: 04/24/2020] [Indexed: 11/17/2022] Open
Abstract
During the course of a toxic challenge, changes in gene expression can manifest such as induction of metabolizing enzymes as a compensatory detoxification response. We currently report that a single 400 mg/kg acetaminophen (APAP) dose to C57BL/6J mice led to an increase in multidrug resistance-associated (Mrp) 4 (Abcc4) mRNA 12 h after administration. Alanine aminotransferase, as a marker of liver injury, was also elevated indicating hepatotoxicity had occurred. Therefore, induction of Mrp4 mRNA was likely attributable to APAP-induced liver injury. Mrp4 has been shown to be upregulated during oxidative stress, and it is well-established that APAP overdose causes oxidative stress due to depletion of glutathione. Given the importance of Mrp4 upregulation as an adaptive response during cholestatic and oxidative liver injury, we next investigated the extent by which human MRP4 can be inhibited by the analgesics, APAP, diclofenac (DCF), and their metabolites. Using an in vitro assay with inside out human MRP4 vesicles, we determined that APAP-cysteine inhibited MRP4-mediated transport of leukotriene C4 with an apparent IC50 of 125 μM. APAP-glutathione also attenuated MRP4 activity though it achieved only 28% inhibition at 300 μM. Diclofenac acyl glucuronide (DCF-AG) inhibited MRP4 transport by 34% at 300 μM. The MRP4 in vitro inhibition occurs at APAP-cysteine and DCF-AG concentrations seen in vivo after toxic doses of APAP or DCF in mice, hence the findings are important given the role that Mrp4 serves as a compensatory response during oxidative stress following toxic challenge. Following 400 mg/kg APAP in mice, mean ALT 12 hours post-dose was 1,140 U/L A statistically significant increase in Mrp4 mRNA was observed 12 hours post-dose APAP-CYS inhibited human MRP4 transport of LTC4 with an IC50 = 125 μM (Ki = 122 μM) APAP-GSH decreased MRP4 transport by 29% inhibition at 300 μM APAP, APAP-GLU, APAP-NAC, and APAP-SUL did not exhibit significant MRP4 inhibition
Collapse
Key Words
- ALT, alanine aminotransferase
- AMP, adenosine monophosphate
- APAP, acetaminophen
- APAP-CYS, acetaminophen cysteine
- APAP-GLU, acetaminophen glucuronide
- APAP-NAC, acetaminophen N-acetylcysteine
- APAP-SUL, acetaminophen sulfate
- ATP, adenosine triphosphate
- Acetaminophen
- DCF, diclofenac
- DCF-AG, diclofenac acyl glucuronide
- Diclofenac
- Fmo, flavin containing monooxygenase
- IS, internal standard
- Inhibition
- LTC4, leukotriene C4
- MRP, multidrug resistance-associated protein
- MRP4
- Metabolite
- OH-DCF, 4′-hydroxy diclofenac
- PGE2, prostaglandin E2
Collapse
Affiliation(s)
| | | | - José E. Manautou
- Corresponding author at: University of Connecticut, Dept. of Pharmaceutical Sciences, 69 North Eagleville Road, Storrs, CT 06269-3092, USA.
| |
Collapse
|
25
|
Huo X, Meng Q, Wang C, Wu J, Zhu Y, Sun P, Ma X, Sun H, Liu K. Targeting renal OATs to develop renal protective agent from traditional Chinese medicines: Protective effect of Apigenin against Imipenem-induced nephrotoxicity. Phytother Res 2020; 34:2998-3010. [PMID: 32468621 DOI: 10.1002/ptr.6727] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 04/11/2020] [Accepted: 04/25/2020] [Indexed: 12/11/2022]
Abstract
Imipenem (Imp) is a widely used broad-spectrum antibiotic. However, renal adverse effects limit its clinical application. We previously reported that organic anion transporters (OATs) facilitated the renal transport of Imp and contributed its nephrotoxicity. Natural flavonoids exhibited renal protective effect. Here, we aimed to develop potent OAT inhibitors from traditional Chinese medicines (TCMs) and to evaluate its protective effect against Imp-induced nephrotoxicity. Among 50 TCMs, Tribuli Fructus, Platycladi Cacumen, and Lycopi Herba exhibited potent inhibition on OAT1/3. After screening their main components, Apigenin strongly inhibited Imp uptake by OAT1/3-HEK293 cells with IC50 values of 1.98 ± 0.36 μM (OAT1) and 2.29 ± 0.88 μM (OAT3). Moreover, Imp exhibited OAT1/3-dependent cytotoxicity, which was alleviated by Apigenin. Furthermore, Apigenin ameliorated Imp-induced nephrotoxicity in rabbits, and reduced the renal secretion of Imp. Apigenin inhibited intracellular accumulation of Imp and sequentially decreased the nephrocyte toxicity in rabbit primary proximal tubule cells (rPTCs). Apigenin, a flavone widely distributed in TCMs, was a potent OAT1/3 inhibitor. Through OAT inhibition, at least in part, Apigenin decreased the renal exposure of Imp and consequently protected against the nephrotoxicity of Imp. Apigenin can be used as a promising agent to reduce the renal adverse reaction of Imp in clinic.
Collapse
Affiliation(s)
- Xiaokui Huo
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,Provincial Key Laboratory for Pharmacokinetics and Transport Liaoning, Dalian Medical University, Dalian, China
| | - Qiang Meng
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,Provincial Key Laboratory for Pharmacokinetics and Transport Liaoning, Dalian Medical University, Dalian, China
| | - Changyuan Wang
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,Provincial Key Laboratory for Pharmacokinetics and Transport Liaoning, Dalian Medical University, Dalian, China
| | - Jingjing Wu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Yanna Zhu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Pengyuan Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Xiaodong Ma
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China
| | - Huijun Sun
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,Provincial Key Laboratory for Pharmacokinetics and Transport Liaoning, Dalian Medical University, Dalian, China
| | - Kexin Liu
- Department of Clinical Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, China.,College (Institute) of Integrative Medicine, Dalian Medical University, Dalian, China.,Provincial Key Laboratory for Pharmacokinetics and Transport Liaoning, Dalian Medical University, Dalian, China
| |
Collapse
|
26
|
Huang KM, Uddin ME, DiGiacomo D, Lustberg MB, Hu S, Sparreboom A. Role of SLC transporters in toxicity induced by anticancer drugs. Expert Opin Drug Metab Toxicol 2020; 16:493-506. [PMID: 32276560 DOI: 10.1080/17425255.2020.1755253] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION . Membrane transporters are integral to the maintenance of cellular integrity of all tissue and cell types. While transporters play an established role in the systemic pharmacokinetics of therapeutic drugs, tissue specific expression of uptake transporters can serve as an initiating mechanism that governs the accumulation and impact of cytotoxic drugs. AREAS COVERED . This review provides an overview of organic cation transporters as determinants of chemotherapy-induced toxicities. We also provide insights into the recently updated FDA guidelines for in vitro drug interaction studies, with a particular focus on the class of tyrosine kinase inhibitors as perpetrators of transporter-mediated drug interactions. EXPERT OPINION . Studies performed over the last few decades have highlighted the important role of basolateral uptake and apical efflux transporters in the pathophysiology of drug-induced organ damage. Increased understanding of the mechanisms that govern the accumulation of cytotoxic drugs has provided insights into the development of novel strategies to prevent debilitating toxicities. Furthermore, we argue that current regulatory guidelines provide inadequate recommendations for in vitro studies to identify substrates or inhibitors of drug transporters. Therefore, the translational and predictive power of FDA-approved drugs as modulators of transport function remains ambiguous and warrants further revision of the current guidelines.
Collapse
Affiliation(s)
- Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Muhammad Erfan Uddin
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Duncan DiGiacomo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Maryam B Lustberg
- Department of Medical Oncology, College of Medicine, the Ohio State University and Comprehensive Cancer Center , Columbus, OH, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, the Ohio State University , Columbus, OH, USA
| |
Collapse
|
27
|
Chow T, Wong FTM, Monetti C, Nagy A, Cox B, Rogers IM. Recapitulating kidney development in vitro by priming and differentiating mouse embryonic stem cells in monolayers. NPJ Regen Med 2020; 5:7. [PMID: 32351711 PMCID: PMC7171095 DOI: 10.1038/s41536-020-0092-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Accepted: 03/13/2020] [Indexed: 12/26/2022] Open
Abstract
In order to harness the potential of pluripotent stem cells, we need to understand how to differentiate them to our target cell types. Here, we developed a protocol to differentiate mouse embryonic stem cells (ESCs) to renal progenitors in a step-wise manner. Microarrays were used to track the transcriptional changes at each stage of differentiation and we observed that genes associated with metanephros, ureteric bud, and blood vessel development were significantly upregulated as the cells differentiated towards renal progenitors. Priming the ESCs and optimizing seeding cell density and growth factor concentrations helped improve differentiation efficiency. Organoids were used to determine the developmental potential of the renal progenitor cells. Aggregated renal progenitors gave rise to organoids consisting of LTL+/E-cadherin+ proximal tubules, cytokeratin+ ureteric bud-derived tubules, and extracellular matrix proteins secreted by the cells themselves. Over-expression of key kidney developmental genes, Pax2, Six1, Eya1, and Hox11 paralogs, during differentiation did not improve differentiation efficiency. Altogether, we developed a protocol to differentiate mouse ESCs in a manner that recapitulates embryonic kidney development and showed that precise gene regulation is essential for proper differentiation to occur.
Collapse
Affiliation(s)
- Theresa Chow
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada.,2Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Frances T M Wong
- 2Department of Physiology, University of Toronto, Toronto, ON Canada
| | - Claudio Monetti
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada
| | - Andras Nagy
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada.,3Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada.,4Institute of Medical Science, University of Toronto, Toronto, ON Canada
| | - Brian Cox
- 2Department of Physiology, University of Toronto, Toronto, ON Canada.,3Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada
| | - Ian M Rogers
- 1Lunenfeld Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON Canada.,2Department of Physiology, University of Toronto, Toronto, ON Canada.,3Department of Obstetrics and Gynaecology, University of Toronto, Toronto, ON Canada
| |
Collapse
|
28
|
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
|
29
|
Abstract
The mercapturic acid pathway is a major route for the biotransformation of xenobiotic and endobiotic electrophilic compounds and their metabolites. Mercapturic acids (N-acetyl-l-cysteine S-conjugates) are formed by the sequential action of the glutathione transferases, γ-glutamyltransferases, dipeptidases, and cysteine S-conjugate N-acetyltransferase to yield glutathione S-conjugates, l-cysteinylglycine S-conjugates, l-cysteine S-conjugates, and mercapturic acids; these metabolites constitute a "mercapturomic" profile. Aminoacylases catalyze the hydrolysis of mercapturic acids to form cysteine S-conjugates. Several renal transport systems facilitate the urinary elimination of mercapturic acids; urinary mercapturic acids may serve as biomarkers for exposure to chemicals. Although mercapturic acid formation and elimination is a detoxication reaction, l-cysteine S-conjugates may undergo bioactivation by cysteine S-conjugate β-lyase. Moreover, some l-cysteine S-conjugates, particularly l-cysteinyl-leukotrienes, exert significant pathophysiological effects. Finally, some enzymes of the mercapturic acid pathway are described as the so-called "moonlighting proteins," catalytic proteins that exert multiple biochemical or biophysical functions apart from catalysis.
Collapse
Affiliation(s)
- Patrick E Hanna
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN, USA
| | - M W Anders
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
30
|
Anderson JT, Huang KM, Lustberg MB, Sparreboom A, Hu S. Solute Carrier Transportome in Chemotherapy-Induced Adverse Drug Reactions. Rev Physiol Biochem Pharmacol 2020; 183:177-215. [PMID: 32761456 DOI: 10.1007/112_2020_30] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Members of the solute carrier (SLC) family of transporters are responsible for the cellular influx of a broad range of endogenous compounds and xenobiotics. These proteins are highly expressed in the gastrointestinal tract and eliminating organs such as the liver and kidney, and are considered to be of particular importance in governing drug absorption and elimination. Many of the same transporters are also expressed in a wide variety of organs targeted by clinically important anticancer drugs, directly affect cellular sensitivity to these agents, and indirectly influence treatment-related side effects. Furthermore, targeted intervention strategies involving the use of transport inhibitors have been recently developed, and have provided promising lead candidates for combinatorial therapies associated with decreased toxicity. Gaining a better understanding of the complex interplay between transporter-mediated on-target and off-target drug disposition will help guide the further development of these novel treatment strategies to prevent drug accumulation in toxicity-associated organs, and improve the safety of currently available treatment modalities. In this report, we provide an update on this rapidly emerging field with particular emphasis on anticancer drugs belonging to the classes of taxanes, platinum derivatives, nucleoside analogs, and anthracyclines.
Collapse
Affiliation(s)
- Jason T Anderson
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Kevin M Huang
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Maryam B Lustberg
- Department of Medical Oncology, The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
| | - Alex Sparreboom
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Shuiying Hu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
31
|
Qi L, Luo Q, Zhang Y, Jia F, Zhao Y, Wang F. Advances in Toxicological Research of the Anticancer Drug Cisplatin. Chem Res Toxicol 2019; 32:1469-1486. [PMID: 31353895 DOI: 10.1021/acs.chemrestox.9b00204] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cisplatin is one of the most widely used chemotherapeutic agents for various solid tumors in the clinic due to its high efficacy and broad spectrum. The antineoplastic activity of cisplatin is mainly due to its ability to cross-link with DNA, thus blocking transcription and replication. Unfortunately, the clinical use of cisplatin is limited by its severe, dose-dependent toxic side effects. There are approximately 40 specific toxicities of cisplatin, among which nephrotoxicity is the most common one. Other common side effects include ototoxicity, neurotoxicity, gastrointestinal toxicity, hematological toxicity, cardiotoxicity, and hepatotoxicity. These side effects together reduce the life quality of patients and require lowering the dosage of the drug, even stopping administration, thus weakening the treatment effect. Few effective measures exist clinically against these side effects because the exact mechanisms of various side effects from cisplatin remain still unclear. Therefore, substantial effort has been made to explore the complicated biochemical processes involved in the toxicology of cisplatin, aiming to identify effective ways to reduce or eradicate its toxicity. This review summarizes and reviews the updated advances in the toxicological research of cisplatin. We anticipate to provide insights into the understanding of the mechanisms underlying the side effects of cisplatin and designing comprehensive therapeutic strategies involving cisplatin.
Collapse
Affiliation(s)
- Luyu Qi
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Qun Luo
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China
| | - Yanyan Zhang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Feifei Jia
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Yao Zhao
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China
| | - Fuyi Wang
- Beijing National Laboratory for Molecular Sciences; National Centre for Mass Spectrometry in Beijing; CAS Key Laboratory of Analytical Chemistry for Living Biosystems , Institute of Chemistry, Chinese Academy of Sciences , Beijing 100190 , P.R. China.,University of Chinese Academy of Sciences , Beijing 100049 , P.R. China.,Basic Medical College , Shandong University of Chinese Traditional Medicine , Jinan 250355 , P.R. China
| |
Collapse
|
32
|
Lee HS, Shin HJ, Cho M, Lee SH, Oh DS. Inhibitory effects of Kampo medicines, Keishibukuryogan and Shakuyakukanzoto, on the substrate uptake activities of solute carrier organic anion transporters. J Pharmacol Sci 2018; 138:279-283. [PMID: 30424926 DOI: 10.1016/j.jphs.2018.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to assess the effects of Keishibukuryogan (K-06) and Shakuyakukanzoto (TJ-68), commercial herbal medicines, on the substrate uptake activities of renal organic anion transporters. We performed transporter uptake and cell viability assays in Xenopus oocytes and HEK293 human kidney embryonic cells treated with K-06 or TJ-68. K-06 and TJ-68 markedly inhibited the substrate uptake activities of URAT1, OAT1, and OAT3, while they did not exhibit non-cytotoxic effects. Our findings demonstrated that K-06 and TJ-68 inhibited the substrate uptake activities of renal transporters, suggesting their mechanism of action as nephroprotective agents.
Collapse
Affiliation(s)
- Ho-Sung Lee
- The K-herb Research Center, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Ho Jung Shin
- Department of Pharmacology and PharmacoGenomics Research Center, College of Medicine, Inje University, Busan 47392, Republic of Korea; SPMED Co., Ltd., Busan 46508, Republic of Korea
| | - Munju Cho
- Department of Pharmacology and PharmacoGenomics Research Center, College of Medicine, Inje University, Busan 47392, Republic of Korea
| | - Seung Hoon Lee
- The K-herb Research Center, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea
| | - Dal-Seok Oh
- The K-herb Research Center, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon 34054, Republic of Korea.
| |
Collapse
|
33
|
Hu S, Sprowl JA. Strategies to Reduce Solute Carrier-Mediated Toxicity. Clin Pharmacol Ther 2018; 104:799-802. [DOI: 10.1002/cpt.1185] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/30/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Shuiying Hu
- Division of Pharmaceutics and Pharmaceutical Chemistry; College of Pharmacy and Comprehensive Cancer Center; The Ohio State University; Columbus Ohio USA
| | - Jason A. Sprowl
- Department of Pharmaceutical Sciences; School of Pharmacy and Pharmaceutical Sciences, University at Buffalo; State University of New York; Buffalo NY USA
| |
Collapse
|
34
|
Mohamadi Yarijani Z, Godini A, Madani SH, Najafi H. Reduction of cisplatin-induced renal and hepatic side effects in rat through antioxidative and anti-inflammatory properties of Malva sylvestris L. extract. Biomed Pharmacother 2018; 106:1767-1774. [PMID: 30119252 DOI: 10.1016/j.biopha.2018.07.115] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Cisplatin is widely used in the chemotherapy of solid organ cancers. However, its application is associated with serious side effects in various organs including the kidneys and liver. OBJECTIVES The aim of this study was to investigate the effects of mallow extract on the side effects of cisplatin in the kidneys and liver. METHODS Hydroalcoholic extract of mallow, at doses of 200, 400, and 600 mg/kg BW, was administered to the animals for seven days intraperitoneally (ip). Animals in the Cis + Mallow group received a dose of cisplatin (8 mg/kg, ip) on the third day. Renal and hepatic functional disturbances were evaluated by measuring concentrations of creatinine, urea-nitrogen, aspartate aminotransferase (AST), and alanine aminotransferase (ALT) in the plasma. In order to assess oxidative stress, malondialdehyde (MDA) and ferric reducing antioxidant power (FRAP) levels were measured in the kidney tissue. Then, degree of mRNA expressions of TNF-α and ICAM-1 were measured to examine renal inflammation. Hematoxylin and Eosin (H & E) staining of kidney and liver tissues was performed to study tissue damage and leukocyte infiltration. RESULTS Cisplatin increased levels of plasma creatinine, urea-nitrogen, AST, and ALT; levels of tissue damage and leukocytes infiltration in the kidneys and liver; and MDA level and expression of pro-inflammatory factors in the kidney tissue. Meanwhile, it decreased FRAP level in the kidney tissue. Pretreatment by mallow extract resulted in significant improvement in all measured variables although 200-mg and 400-mg doses yielded better results. CONCLUSION Results showed that mallow supplement protects the kidneys and liver against side effects of cisplatin, and reduces the resultant oxidative stress and inflammation.
Collapse
Affiliation(s)
| | - Aliashraf Godini
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Hamid Madani
- Department of Pathology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Houshang Najafi
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
35
|
Soo JYC, Jansen J, Masereeuw R, Little MH. Advances in predictive in vitro models of drug-induced nephrotoxicity. Nat Rev Nephrol 2018; 14:378-393. [PMID: 29626199 PMCID: PMC6013592 DOI: 10.1038/s41581-018-0003-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In vitro screens for nephrotoxicity are currently poorly predictive of toxicity in humans. Although the functional proteins that are expressed by nephron tubules and mediate drug susceptibility are well known, current in vitro cellular models poorly replicate both the morphology and the function of kidney tubules and therefore fail to demonstrate injury responses to drugs that would be nephrotoxic in vivo. Advances in protocols to enable the directed differentiation of pluripotent stem cells into multiple renal cell types and the development of microfluidic and 3D culture systems have opened a range of potential new platforms for evaluating drug nephrotoxicity. Many of the new in vitro culture systems have been characterized by the expression and function of transporters, enzymes, and other functional proteins that are expressed by the kidney and have been implicated in drug-induced renal injury. In vitro platforms that express these proteins and exhibit molecular biomarkers that have been used as readouts of injury demonstrate improved functional maturity compared with static 2D cultures and represent an opportunity to model injury to renal cell types that have hitherto received little attention. As nephrotoxicity screening platforms become more physiologically relevant, they will facilitate the development of safer drugs and improved clinical management of nephrotoxicants.
Collapse
Affiliation(s)
- Joanne Y-C Soo
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia
- Murdoch Children's Research Institute, Parkville, Victoria, Australia
| | - Jitske Jansen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Rosalinde Masereeuw
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, Netherlands
| | - Melissa H Little
- Department of Paediatrics, The University of Melbourne, Parkville, Victoria, Australia.
- Murdoch Children's Research Institute, Parkville, Victoria, Australia.
- Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
36
|
Organic solute carrier 22 (SLC22) family: Potential for interactions with food, herbal/dietary supplements, endogenous compounds, and drugs. J Food Drug Anal 2018; 26:S45-S60. [PMID: 29703386 PMCID: PMC9326878 DOI: 10.1016/j.jfda.2018.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/07/2023] Open
|
37
|
Nieskens TTG, Peters JGP, Dabaghie D, Korte D, Jansen K, Van Asbeck AH, Tavraz NN, Friedrich T, Russel FGM, Masereeuw R, Wilmer MJ. Expression of Organic Anion Transporter 1 or 3 in Human Kidney Proximal Tubule Cells Reduces Cisplatin Sensitivity. Drug Metab Dispos 2018. [PMID: 29514829 DOI: 10.1124/dmd.117.079384] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cisplatin is a cytostatic drug used for treatment of solid organ tumors. The main adverse effect is organic cation transporter 2 (OCT2)-mediated nephrotoxicity, observed in 30% of patients. The contribution of other renal drug transporters is elusive. Here, cisplatin-induced toxicity was evaluated in human-derived conditionally immortalized proximal tubule epithelial cells (ciPTEC) expressing renal drug transporters, including OCT2 and organic anion transporters 1 (OAT1) or 3 (OAT3). Parent ciPTEC demonstrated OCT2-dependent cisplatin toxicity (TC50 34 ± 1 μM after 24-hour exposure), as determined by cell viability. Overexpression of OAT1 and OAT3 resulted in reduced sensitivity to cisplatin (TC50 45 ± 6 and 64 ± 11 μM after 24-hour exposure, respectively). This effect was independent of OAT-mediated transport, as the OAT substrates probenecid and diclofenac did not influence cytotoxicity. Decreased cisplatin sensitivity in OAT-expressing cells was associated directly with a trend toward reduced intracellular cisplatin accumulation, explained by reduced OCT2 gene expression and activity. This was evaluated by Vmax of the OCT2-model substrate ASP+ (23.5 ± 0.1, 13.1 ± 0.3, and 21.6 ± 0.6 minutes-1 in ciPTEC-parent, ciPTEC-OAT1, and ciPTEC-OAT3, respectively). Although gene expression of cisplatin efflux transporter multidrug and toxin extrusion 1 (MATE1) was 16.2 ± 0.3-fold upregulated in ciPTEC-OAT1 and 6.1 ± 0.7-fold in ciPTEC-OAT3, toxicity was unaffected by the MATE substrate pyrimethamine, suggesting that MATE1 does not play a role in the current experimental set-up. In conclusion, OAT expression results in reduced cisplatin sensitivity in renal proximal tubule cells, explained by reduced OCT2-mediated uptake capacity. In vitro drug-induced toxicity studies should consider models that express both OCT and OAT drug transporters.
Collapse
Affiliation(s)
- Tom T G Nieskens
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Janny G P Peters
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Dina Dabaghie
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Daphne Korte
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Katja Jansen
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Alexander H Van Asbeck
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Neslihan N Tavraz
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Thomas Friedrich
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Frans G M Russel
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Rosalinde Masereeuw
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| | - Martijn J Wilmer
- Department of Pharmacology and Toxicology, Radboud Institute for Molecular Life Sciences, Radboudumc, Nijmegen, The Netherlands (T.T.G.N., J.G.P.P., D.D., D.K., K.J., A.H.V.A., F.G.M.R., M.J.W.); Department of Physical Chemistry/Bioenergetics, Institute of Chemistry PC14, Technical University of Berlin, Berlin, Germany (N.N.T., T.F.); and Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht, The Netherlands (K.J., R.M.)
| |
Collapse
|