1
|
Kala J, Joseph T, Pirovano M, Fenoglio R, Cosmai L. Acute Kidney Injury Associated with Anticancer Therapies: Small Molecules and Targeted Therapies. KIDNEY360 2024; 5:1750-1762. [PMID: 39186376 DOI: 10.34067/kid.0000000566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
Molecular targeted therapy has revolutionized cancer treatment by significantly improving patient survival compared with standard conventional chemotherapies. The use of these drugs targets specific molecules or targets, which block growth and spread of cancer cells. Many of these therapies have been approved for use with remarkable success in breast, blood, colorectal, lung, and ovarian cancers. The advantage over conventional chemotherapy is its ability to deliver drugs effectively with high specificity while being less toxic. Although known as "targeted," many of these agents lack specificity and selectivity, and they tend to inhibit multiple targets, including those in the kidneys. The side effects usually arise because of dysregulation of targets of the inhibited molecule in normal tissue. The off-target effects are caused by drug binding to unintended targets. The on-target effects are associated with inhibition toward the pathway reflecting inappropriate inhibition or activation of the intended drug target. Early detection and correct management of kidney toxicities is crucial to preserve kidney functions. The knowledge of these toxicities helps guide optimal and continued utilization of these potent therapies. This review summarizes the different types of molecular targeted therapies used in the treatment of cancer and the incidence, severity, and pattern of nephrotoxicity caused by them, with their plausible mechanism and proposed treatment recommendations.
Collapse
Affiliation(s)
- Jaya Kala
- Division of Nephrology, Department of Internal Medicine, University of Texas Health Science Center-McGovern Medical School, Houston, Texas
| | - Teresa Joseph
- Division of Nephrology, Department of Internal Medicine, University of Texas Health Science Center-McGovern Medical School, Houston, Texas
- Section of Nephrology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Marta Pirovano
- Department of Biomedical and Clinical Sciences, University of Milan, Milan, Italy
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Roberta Fenoglio
- University Center of Excellence on Nephrological, Rheumatological and Rare Diseases (ERK-net, ERN-Reconnect and RITA-ERN Member) including Nephrology and Dialysis Unit and Center of Immuno-Rheumatology and Rare Diseases (CMID), Turin, Italy
- Coordinating Center of the Interregional Network for Rare Diseases of Piedmont and Aosta Valley (North-West Italy), San Giovanni Bosco Hub Hospital, ASL Cittàdi Torino, Turin, Italy
- Department of Clinical and Biological Sciences of the University of Turin, Turin, Italy
| | - Laura Cosmai
- Onconephrology Outpatient Clinic, Nephrology and Dialysis Unit, ASST Fatebenefratelli Sacco, Milan, Italy
| |
Collapse
|
2
|
Loren P, Saavedra N, Saavedra K, Zambrano T, Moriel P, Salazar LA. Epigenetic Mechanisms Involved in Cisplatin-Induced Nephrotoxicity: An Update. Pharmaceuticals (Basel) 2021; 14:ph14060491. [PMID: 34063951 PMCID: PMC8223972 DOI: 10.3390/ph14060491] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023] Open
Abstract
Cisplatin is an antineoplastic drug used for the treatment of many solid tumors. Among its various side effects, nephrotoxicity is the most detrimental. In recent years, epigenetic regulation has emerged as a modulatory mechanism of cisplatin-induced nephrotoxicity, involving non-coding RNAs, DNA methylation and histone modifications. These epigenetic marks alter different signaling pathways leading to damage and cell death. In this review, we describe how different epigenetic modifications alter different pathways leading to cell death by apoptosis, autophagy, necroptosis, among others. The study of epigenetic regulation is still under development, and much research remains to fully determine the epigenetic mechanisms underlying cell death, which will allow leading new strategies for the diagnosis and therapy of this disease.
Collapse
Affiliation(s)
- Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (N.S.); (K.S.)
| | - Nicolás Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (N.S.); (K.S.)
| | - Kathleen Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (N.S.); (K.S.)
| | - Tomás Zambrano
- Department of Medical Technology, Faculty of Medicine, Universidad de Chile, Santiago 8380453, Chile;
| | - Patricia Moriel
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas 13083970, SP, Brazil;
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (N.S.); (K.S.)
- Correspondence: ; Tel.: +56-452-596-724
| |
Collapse
|
3
|
Chen J, Huang L, Zhu Q, Wang Z, Tang Z. MTSS1 hypermethylation is associated with prostate cancer progression. J Cell Physiol 2019; 235:2687-2697. [PMID: 31541465 DOI: 10.1002/jcp.29172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 08/23/2019] [Indexed: 01/08/2023]
Abstract
This study was conducted to evaluate the influence of DNA methylation of metastasis suppressor 1 (MTSS1) on prostate cancer (PCa) progression. Forty-nine paired PCa tissue samples and normal tissue samples from The Cancer Genome Atlas were analyzed. Methylome analysis, CpG island arrays and Hierarchical clustering were used to analyze methylation profiles of PCa tissues. MTSS1 methylation level was detected by methylation-specific PCR. Relative messenger RNA and the expression level of MTSS1 protein were identified by quantitative real-time PCR (qRT-PCR) and western blot analysis. The migration, invasion, proliferation, and cell cycle were detected separately by wound-healing assay, transwell chamber assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and flow cytometry. The roles of MTSS1 in PCa progression were demonstrated in vivo by tumor formation assays in nude mice. MTSS1 expression was decreased in PCa tissues in comparison with paired adjacent normal prostate tissues. Compared to the methylation of MTSS1 in normal prostate tissues based on the MethHC website, the MTSS1 in PCa tissues was hypermethylated. The expression of MTSS1 detected by qRT-PCR and western blot analysis was found to be downregulated in PCa cells and tissues. The reduced expression of MTSS1 by small interfering RNA-MTSS1 was recovered by 5-aza-2'-deoxycytidine treatment. Besides, MTSS1 demethylation inhibited migration, invasion, and proliferation of PCa cells, and induced cell cycle to be arrested at G0/G1 phase. Furthermore, it was shown by tumor xenograft assay that MTSS1 inhibited the growth of tumor in vivo. Hypermethylated MTSS1 promoted PCa cells migration, invasion, and proliferation, and suppressed cell cycle arrest at the G0/G1 phase.
Collapse
Affiliation(s)
- Junjie Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liang Huang
- Department of Urology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Quan Zhu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhao Wang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhengyan Tang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, Hunan, China.,Hunan Engineering Laboratory for Diagnosis and Treatment Technology of Urogenital Diseases, Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
4
|
Kolli RT, Glenn TC, Brown BT, Kaur SP, Barnett LM, Lash LH, Cummings BS. Bromate-induced Changes in p21 DNA Methylation and Histone Acetylation in Renal Cells. Toxicol Sci 2019; 168:460-473. [PMID: 30649504 PMCID: PMC6432867 DOI: 10.1093/toxsci/kfz016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Bromate (BrO3-) is a water disinfection byproduct (DBP) previously shown to induce nephrotoxicity in vitro and in vivo. We recently showed that inhibitors of DNA methyltransferase 5-aza-2'-deoxycytidine (5-Aza) and histone deacetylase trichostatin A (TSA) increased BrO3- nephrotoxicity whereas altering the expression of the cyclin-dependent kinase inhibitor p21. Human embryonic kidney cells (HEK293) and normal rat kidney (NRK) cells were sub-chronically exposed to BrO3- or epigenetic inhibitors for 18 days, followed by 9 days of withdrawal. DNA methylation was studied using a modification of bisulfite amplicon sequencing called targeted gene bisulfite sequencing. Basal promoter methylation in the human p21 promoter region was substantially lower than that of the rat DNA. Furthermore, 5-Aza decreased DNA methylation in HEK293 cells at the sis-inducible element at 3 distinct CpG sites located at 691, 855, and 895 bp upstream of transcription start site (TSS). 5-Aza also decreased methylation at the rat p21 promoter about 250 bp upstream of the p21 TSS. In contrast, sub-chronic BrO3- exposure failed to alter methylation in human or rat renal cells. BrO3- exposure altered histone acetylation in NRK cells at the p21 TSS, but not in HEK293 cells. Interestingly, changes in DNA methylation induced by 5-Aza persisted after its removal; however, TSA- and BrO3--induced histone hyperacetylation returned to basal levels after 3 days of withdrawal. These data demonstrate novel sites within the p21 gene that are epigenetically regulated and further show that significant differences exist in the epigenetic landscape between rat and human p21, especially with regards to toxicant-induced changes in histone acetylation.
Collapse
Affiliation(s)
- Ramya T Kolli
- Department of Pharmaceutical and Biomedical Sciences
- Interdisciplinary Toxicology Program
- National Institute of Environmental Health Sciences, Building 101, 111 TW Alexander Drive, Durham, NC 27709
| | - Travis C Glenn
- Interdisciplinary Toxicology Program
- Environmental Health Science
| | - Bradley T Brown
- College of Pharmacy, University of Georgia, Athens, Georgia 30602
| | | | - Lillie M Barnett
- Department of Pharmaceutical and Biomedical Sciences
- Interdisciplinary Toxicology Program
| | - Lawrence H Lash
- Department of Pharmacology, Wayne State University, Detroit, Michigan 48201
| | - Brian S Cummings
- Department of Pharmaceutical and Biomedical Sciences
- Interdisciplinary Toxicology Program
| |
Collapse
|
5
|
|
6
|
Theobald J, Ghanem A, Wallisch P, Banaeiyan AA, Andrade-Navarro MA, Taškova K, Haltmeier M, Kurtz A, Becker H, Reuter S, Mrowka R, Cheng X, Wölfl S. Liver-Kidney-on-Chip To Study Toxicity of Drug Metabolites. ACS Biomater Sci Eng 2017; 4:78-89. [DOI: 10.1021/acsbiomaterials.7b00417] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jannick Theobald
- Institute
of Pharmacy and Molecular Biotechnology, Pharmaceutical Biology, Heidelberg University, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Ali Ghanem
- Institute
of Pharmacy and Molecular Biotechnology, Pharmaceutical Biology, Heidelberg University, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Patrick Wallisch
- Institute
of Pharmacy and Molecular Biotechnology, Pharmaceutical Biology, Heidelberg University, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Amin A. Banaeiyan
- Biological
Physics, Department of Physics, Chalmers Campus, University of Gothenburg, Gothenburg SE-41296, Sweden
| | - Miguel A. Andrade-Navarro
- Computational
Biology and Data Mining Group, Institute for Molecular Biology, Johannes Gutenberg University Mainz, Ackermannweg 4, Mainz 55128, Germany
| | - Katerina Taškova
- Computational
Biology and Data Mining Group, Institute for Molecular Biology, Johannes Gutenberg University Mainz, Ackermannweg 4, Mainz 55128, Germany
| | | | - Andreas Kurtz
- Berlin-Brandenburg
Center for Regenerative Therapies, Charité - Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin 13353, Germany
| | - Holger Becker
- Microfluidic ChipShop GmbH, Stockholmer
Strasse 20, Jena 07747, Germany
| | - Stefanie Reuter
- Experimentelle
Nephrologie, KIM III, Universitätsklinikum Jena, Am Nonenplan 4, Jena 07747, Germany
| | - Ralf Mrowka
- Experimentelle
Nephrologie, KIM III, Universitätsklinikum Jena, Am Nonenplan 4, Jena 07747, Germany
| | - Xinlai Cheng
- Institute
of Pharmacy and Molecular Biotechnology, Pharmaceutical Biology, Heidelberg University, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| | - Stefan Wölfl
- Institute
of Pharmacy and Molecular Biotechnology, Pharmaceutical Biology, Heidelberg University, Im Neuenheimer Feld 364, D-69120 Heidelberg, Germany
| |
Collapse
|