1
|
Fonoudi H, Jouni M, Cejas RB, Magdy T, Blancard M, Ge N, Shah DA, Lyra-Leite DM, Neupane A, Gharib M, Jiang Z, Sapkota Y, Burridge PW. Functional Validation of Doxorubicin-Induced Cardiotoxicity-Related Genes. JACC CardioOncol 2024; 6:38-50. [PMID: 38510289 PMCID: PMC10950437 DOI: 10.1016/j.jaccao.2023.11.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 11/16/2023] [Accepted: 11/21/2023] [Indexed: 03/22/2024] Open
Abstract
Background Genome-wide association studies and candidate gene association studies have identified more than 180 genetic variants statistically associated with anthracycline-induced cardiotoxicity (AIC). However, the lack of functional validation has hindered the clinical translation of these findings. Objectives The aim of this study was to functionally validate all genes associated with AIC using human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Methods Through a systemic literature search, 80 genes containing variants significantly associated with AIC were identified. Additionally, 3 more genes with potential roles in AIC (GSTM1, CBR1, and ERBB2) were included. Of these, 38 genes exhibited expression in human fetal heart, adult heart, and hiPSC-CMs. Using clustered regularly interspaced short palindromic repeats/Cas9-based genome editing, each of these 38 genes was systematically knocked out in control hiPSC-CMs, and the resulting doxorubicin-induced cardiotoxicity (DIC) phenotype was assessed using hiPSC-CMs. Subsequently, functional assays were conducted for each gene knockout on the basis of hypothesized mechanistic implications in DIC. Results Knockout of 26 genes increased the susceptibility of hiPSC-CMs to DIC. Notable genes included efflux transporters (ABCC10, ABCC2, ABCB4, ABCC5, and ABCC9), well-established DIC-associated genes (CBR1, CBR3, and RAC2), and genome-wide association study-discovered genes (RARG and CELF4). Conversely, knockout of ATP2B1, HNMT, POR, CYBA, WDR4, and COL1A2 had no significant effect on the in vitro DIC phenotype of hiPSC-CMs. Furthermore, knockout of the uptake transporters (SLC28A3, SLC22A17, and SLC28A1) demonstrated a protective effect against DIC. Conclusions The present findings establish a comprehensive platform for the functional validation of DIC-associated genes, providing insights for future studies in DIC variant associations and potential mechanistic targets for the development of cardioprotective drugs.
Collapse
Affiliation(s)
- Hananeh Fonoudi
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mariam Jouni
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Romina B. Cejas
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Tarek Magdy
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Malorie Blancard
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ning Ge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Disheet A. Shah
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Davi M. Lyra-Leite
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Achal Neupane
- Department of Epidemiology and Cancer Control, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Mennat Gharib
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Zhengxin Jiang
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Yadav Sapkota
- Department of Epidemiology and Cancer Control, St. Jude Children’s Hospital, Memphis, Tennessee, USA
| | - Paul W. Burridge
- Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Center for Pharmacogenomics, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
2
|
Virgili Manrique AC, Salazar J, Arranz MJ, Bagué S, Orellana R, López-Pousa A, Cerdà P, Gracia I, Majercakova K, Peiró A, Trullols L, Fernández M, Valverde S, Quintana MJ, Bell O, Artigas-Baleri A, Sebio A. Pharmacogenetic Profiling in High-Risk Soft Tissue Sarcomas Treated with Neoadjuvant Chemotherapy. J Pers Med 2022; 12:jpm12040618. [PMID: 35455734 PMCID: PMC9024670 DOI: 10.3390/jpm12040618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/07/2022] [Indexed: 12/10/2022] Open
Abstract
Neoadjuvant chemotherapy based on anthracyclines and ifosfamide for high-risk soft tissue sarcomas (STS) of the extremities and trunk is a controversial treatment option. There are substantial interindividual differences in clinical outcomes in patients treated with neoadjuvant chemotherapy. The aim of this study was to evaluate, as biomarkers, polymorphisms in genes encoding drug-metabolizing enzymes, drug transporters, or drug targets and their association with toxicity and survival in STS patients treated with neoadjuvant chemotherapy. We analysed variants in genes involved in anthracycline metabolism (ABCB1, ABCC2, NQO1, CBR3, and SLC22A16) and in ifosfamide catabolism (ALDH1A1) in 79 treated patients. Two genes showed significant association after adjusted multivariate analysis: ABCC2 and ALDH1A1. In patients treated with anthracyclines, ABCC2 rs3740066 was associated with risk of febrile neutropenia (p = 0.031), and with decreased overall survival (OS) (p = 0.024). ABCC2 rs2273697 was associated with recurrence-free survival (RFS) (p = 0.024). In patients treated with ifosfamide, ALDH1A1 rs3764435 was associated with RFS (p = 0.046). Our pharmacogenetic study shows for the first time that variants in genes regulating the metabolism of neoadjuvant chemotherapy may be helpful to predict toxicity and survival benefit in high-risk STS treated with neoadjuvant chemotherapy. Further validation studies are needed to establish their clinical utility.
Collapse
Affiliation(s)
- Anna C. Virgili Manrique
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.M.); (A.L.-P.); (P.C.)
- Department of Medicine, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Juliana Salazar
- Medical Translational Oncology Laboratory, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
- Correspondence: (J.S.); (A.S.)
| | | | - Silvia Bagué
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (S.B.); (R.O.)
| | - Ruth Orellana
- Department of Pathology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (S.B.); (R.O.)
| | - Antonio López-Pousa
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.M.); (A.L.-P.); (P.C.)
| | - Paula Cerdà
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.M.); (A.L.-P.); (P.C.)
| | - Isidre Gracia
- Orthopaedics and Trauma Surgery, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (I.G.); (A.P.); (L.T.)
| | - Katarina Majercakova
- Radiation Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Ana Peiró
- Orthopaedics and Trauma Surgery, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (I.G.); (A.P.); (L.T.)
| | - Laura Trullols
- Orthopaedics and Trauma Surgery, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (I.G.); (A.P.); (L.T.)
| | - Manuel Fernández
- Plastic and Reconstructive Surgery, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Sandra Valverde
- Radiology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - María Jesús Quintana
- Epidemiology Department, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Olga Bell
- Medical Translational Oncology Laboratory, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Alícia Artigas-Baleri
- Genetics Department, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain;
| | - Ana Sebio
- Department of Medical Oncology, Hospital de la Santa Creu i Sant Pau, 08041 Barcelona, Spain; (A.C.V.M.); (A.L.-P.); (P.C.)
- Correspondence: (J.S.); (A.S.)
| |
Collapse
|
3
|
van Eijk M, Pluim D, Dorlo TPC, Marchetti S, Huitema ADR, Beijnen JH. Investigating the influence of relevant pharmacogenetic variants on the pharmacokinetics and pharmacodynamics of orally administered docetaxel combined with ritonavir. THE PHARMACOGENOMICS JOURNAL 2021; 21:336-345. [PMID: 33649517 DOI: 10.1038/s41397-021-00213-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 01/12/2021] [Accepted: 01/20/2021] [Indexed: 11/09/2022]
Abstract
The anticancer drug docetaxel exhibits large interpatient pharmacokinetic and pharmacodynamic variability. In this study, we aimed to assess the functional significance of 14 polymorphisms in the CYP3A, CYP1B1, ABCB1, ABCC2, and SLCO1B3 genes for the pharmacokinetics and pharmacodynamics of oral docetaxel, co-administered with ritonavir. None of the tested CYP3A, ABCB1, ABCC2, and SLCO1B3 genotypes and diplotypes showed a significant relation with an altered bioavailability or clearance of either docetaxel or ritonavir. Similarly, no clear effect of CYP1B1 genotype on clinical outcomes was observed in a subgroup of non-small cell lung cancer (NSCLC) patients. Our post hoc power analysis indicated that our pharmacogenetic-pharmacokinetic analysis was only powered for relatively high effect sizes, which were to be expected given the high interpatient variability. This makes it unlikely that future studies will explain the high observed interpatient variability in oral docetaxel pharmacokinetics as a result of any of these separate polymorphisms and diplotypes.
Collapse
Affiliation(s)
- Maarten van Eijk
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands.
| | - Dick Pluim
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Thomas P C Dorlo
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Serena Marchetti
- Division of Clinical Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands
| | - Alwin D R Huitema
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Department of Clinical Pharmacy, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Pharmacology, Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Jos H Beijnen
- Department of Pharmacy & Pharmacology, The Netherlands Cancer Institute-Antoni van Leeuwenhoek, Amsterdam, The Netherlands.,Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
4
|
Gao S, Bell EC, Zhang Y, Liang D. Racial Disparity in Drug Disposition in the Digestive Tract. Int J Mol Sci 2021; 22:1038. [PMID: 33494365 PMCID: PMC7865938 DOI: 10.3390/ijms22031038] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
The major determinants of drug or, al bioavailability are absorption and metabolism in the digestive tract. Genetic variations can cause significant differences in transporter and enzyme protein expression and function. The racial distribution of selected efflux transporter (i.e., Pgp, BCRP, MRP2) and metabolism enzyme (i.e., UGT1A1, UGT1A8) single nucleotide polymorphisms (SNPs) that are highly expressed in the digestive tract are reviewed in this paper with emphasis on the allele frequency and the impact on drug absorption, metabolism, and in vivo drug exposure. Additionally, preclinical and clinical models used to study the impact of transporter/enzyme SNPs on protein expression and function are also reviewed. The results showed that allele frequency of the major drug efflux transporters and the major intestinal metabolic enzymes are highly different in different races, leading to different drug disposition and exposure. The conclusion is that genetic polymorphism is frequently observed in different races and the related protein expression and drug absorption/metabolism function and drug in vivo exposure can be significantly affected, resulting in variations in drug response. Basic research on race-dependent drug absorption/metabolism is expected, and FDA regulations of drug dosing adjustment based on racial disparity are suggested.
Collapse
Affiliation(s)
- Song Gao
- Department of Pharmaceutical Science, College of Pharmacy and Health Sciences, Texas Southern University, 3100 Cleburne Street, Houston, TX 77004, USA; (E.C.B.); (Y.Z.); (D.L.)
| | | | | | | |
Collapse
|
5
|
Yamazoe T, Mori T, Yoshio S, Kanto T. Hepatocyte ploidy and pathological mutations in hepatocellular carcinoma: impact on oncogenesis and therapeutics. Glob Health Med 2020; 2:273-281. [PMID: 33330821 DOI: 10.35772/ghm.2020.01089] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 12/23/2022]
Abstract
Hepatocellular carcinoma (HCC) occurs in the chronic liver inflammation such as viral hepatitis, alcoholic and non-alcoholic steatohepatitis. While anti-viral treatment has been significantly improved, the prevalence of HCC remains high and treatment is still challenging. The continuation of hepatocyte death, inflammation, and fibrosis leads to the accumulation of gene alterations, which may trigger carcinogenesis. Hepatocytes are a unique cell type having more than one complete set of 23 chromosomes, termed polyploidy. Due to gene redundancy, hepatocytes may tolerate lethal mutations. Next generation sequencing technology has revealed gene alterations in HCC related to telomere maintenance, Wnt/β-catenin pathway, p53 cell-cycle pathway, epigenetic modifiers, oxidative stress pathway, PI3K/AKT/mTOR, and RAS/RAF/MAPK pathway with or without a chromosomal instability. Some type of driver gene mutations accumulates in hepatocytes and breaks the orchestration of excessive copies of chromosomes, which may lead to unfavorable gene expressions and fuel tumorigenesis. Recently, molecular targeted drugs, developed with the aim of interfering with these signaling pathways, are being used for HCC patients in the clinics. Therefore, a deeper understanding of hepatocyte ploidy and genetic or epigenetic alterations is indispensable for the establishment of novel therapeutic strategies against HCC.
Collapse
Affiliation(s)
- Taiji Yamazoe
- Department of Liver Disease, Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| | - Taizo Mori
- Department of Liver Disease, Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| | - Sachiyo Yoshio
- Department of Liver Disease, Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| | - Tatsuya Kanto
- Department of Liver Disease, Research Center for Hepatitis and Immunology, National Center for Global Health and Medicine, Ichikawa, Chiba, Japan
| |
Collapse
|
6
|
Cabral LKD, Tiribelli C, Sukowati CHC. Sorafenib Resistance in Hepatocellular Carcinoma: The Relevance of Genetic Heterogeneity. Cancers (Basel) 2020; 12:E1576. [PMID: 32549224 PMCID: PMC7352671 DOI: 10.3390/cancers12061576] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
Despite advances in biomedicine, the incidence and the mortality of hepatocellular carcinoma (HCC) remain high. The majority of HCC cases are diagnosed in later stages leading to the less than optimal outcome of the treatments. Molecular targeted therapy with sorafenib, a dual-target inhibitor targeting the serine-threonine kinase Raf and the tyrosine kinases VEGFR/PDGFR, is at present the main treatment for advanced-stage HCC, either in a single or combinatory regimen. However, it was observed in a large number of patients that its effectiveness is hampered by drug resistance. HCC is highly heterogeneous, within the tumor and among individuals, and this influences disease progression, classification, prognosis, and naturally cellular susceptibility to drug resistance. This review aims to provide an insight on how HCC heterogeneity influences the different primary mechanisms of chemoresistance against sorafenib including reduced drug intake, enhanced drug efflux, intracellular drug metabolism, alteration of molecular targets, activation/inactivation of signaling pathways, changes in the DNA repair machinery, and negative balance between apoptosis and survival of the cancer cells. The diverse variants, mutations, and polymorphisms in molecules and their association with drug response can be a helpful tool in treatment decision making. Accordingly, the existence of heterogeneous biomarkers in the tumor must be considered to strengthen multi-target strategies in patient-tailored treatment.
Collapse
Affiliation(s)
| | | | - Caecilia H. C. Sukowati
- Fondazione Italiana Fegato (Italian Liver Foundation), AREA Science Park, Basovizza, 34149 Trieste, Italy; (L.K.D.C.); (C.T.)
| |
Collapse
|