51
|
Pan Y, Omori K, Ali I, Tachikawa M, Terasaki T, Brouwer KLR, Nicolazzo JA. Altered Expression of Small Intestinal Drug Transporters and Hepatic Metabolic Enzymes in a Mouse Model of Familial Alzheimer's Disease. Mol Pharm 2018; 15:4073-4083. [PMID: 30074800 DOI: 10.1021/acs.molpharmaceut.8b00500] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Drug transporter expression and function at the blood-brain barrier is altered in Alzheimer's disease (AD). However, the impact of AD on the expression of transporters and metabolizing enzymes in peripheral tissues has received little attention. The current study evaluated the expression of drug transporters and metabolizing enzymes in the small intestine and liver from 8- to 9-month-old female wild-type (WT) and APPswe/PSEN 1dE9 (APP/PS1) transgenic mice, a widely used AD model, using a quantitative targeted absolute proteomics (QTAP) approach. Furthermore, the general morphological appearance of the liver was assessed by immunohistochemistry, and lipid content was visualized using Oil Red O staining. The small intestines of APP/PS1 mice exhibited a significant 2.3-fold increase in multidrug resistance-associated protein 2 (Mrp2), a 1.9-fold decrease in monocarboxylate transporter 1 (Mct1), and a 3.6-fold increase in UDP-glucuronosyltransferase (Ugt) 2b5 relative to those from WT mice based on QTAP analysis. While the liver from APP/PS1 mice exhibited no changes in drug transporter expression, there was a 1.3-fold elevation in cytochrome P450 (Cyp) 51a1 and a 1.2-fold reduction in Cyp2c29 protein expression, and this was associated with morphological alterations including accumulation of hepatocyte lipids. These studies are the first to demonstrate that the protein expression of transporters and metabolizing enzymes important in oral drug absorption are modified in a mouse model of familial AD, which may lead to altered disposition of some orally administered drugs in AD.
Collapse
Affiliation(s)
- Yijun Pan
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , 399 Royal Parade , Parkville , Victoria 3052 , Australia
| | - Kotaro Omori
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , 6-3 Aoba , Aramaki, Aoba-ku, Sendai 980-8578 , Japan
| | - Izna Ali
- UNC Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , 301 Pharmacy Lane , Chapel Hill , North Carolina 27599 , United States
| | - Masanori Tachikawa
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , 6-3 Aoba , Aramaki, Aoba-ku, Sendai 980-8578 , Japan
| | - Tetsuya Terasaki
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences , Tohoku University , 6-3 Aoba , Aramaki, Aoba-ku, Sendai 980-8578 , Japan
| | - Kim L R Brouwer
- UNC Eshelman School of Pharmacy , University of North Carolina at Chapel Hill , 301 Pharmacy Lane , Chapel Hill , North Carolina 27599 , United States
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences , Monash University , 399 Royal Parade , Parkville , Victoria 3052 , Australia
| |
Collapse
|
52
|
Sanchez-Dominguez CN, Gallardo-Blanco HL, Salinas-Santander MA, Ortiz-Lopez R. Uridine 5'-diphospho-glucronosyltrasferase: Its role in pharmacogenomics and human disease. Exp Ther Med 2018; 16:3-11. [PMID: 29896223 DOI: 10.3892/etm.2018.6184] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/17/2017] [Indexed: 12/18/2022] Open
Abstract
Biotransformation is an enzyme-catalyzed process in which the body converts endogenous compounds, xenobiotics and toxic substances into harmless or easily excreted metabolites. The biotransformation reactions are classified as phase I and II reactions. Uridine 5'-diphospho (UDP)-glucuronosyltransferases (UGTs) are a superfamily of phase II enzymes which have roles in the conjugation of xenobiotics or endogenous compounds, including drugs and bilirubin, with glucuronic acid to make them easier to excrete. The method the human body uses to achieve glucuronidation may be affected by a large interindividual variation due to changes in the sequences of the genes encoding these enzymes. In the last five years, the study of the genetic variants of the UGTs at a molecular level has become important due to its association with several diseases and the ability to predict adverse events due to drug metabolism. In the present review, the structure and the prominent genetic variants of the UGT1A subfamily and their metabolic and clinical implications are described.
Collapse
Affiliation(s)
- Celia N Sanchez-Dominguez
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon 64460, Mexico
| | - Hugo L Gallardo-Blanco
- Department of Genetics, Faculty of Medicine, Autonomous University of Nuevo Leon, Monterrey, Nuevo Leon 64460, Mexico
| | | | - Rocio Ortiz-Lopez
- Tecnologico de Monterrey, Medical School and Health Sciences, Monterrey, Nuevo Leon 64710, Mexico
| |
Collapse
|
53
|
Belkhir L, Seguin-Devaux C, Elens L, Pauly C, Gengler N, Schneider S, Ruelle J, Haufroid V, Vandercam B. Impact of UGT1A1 polymorphisms on Raltegravir and its glucuronide plasma concentrations in a cohort of HIV-1 infected patients. Sci Rep 2018; 8:7359. [PMID: 29743555 PMCID: PMC5943329 DOI: 10.1038/s41598-018-25803-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 04/27/2018] [Indexed: 01/01/2023] Open
Abstract
The aim of this study was to evaluate the effect of UGT1A1 polymorphisms on Raltegravir (RAL) and its metabolite RAL-glucuronide trough plasma concentrations ([RAL]plasma and [RAL-glu]plasma) and on the metabolic ratio (MR): [RAL-glu]plasma/[RAL]plasma. UGT1A1 genotyping was performed on 96 patients. 44% (n = 42) were homozygous UGT1A1*1/*1 while 50% (n = 48) and 6% (n = 6) were UGT1A1*28 and UGT1A1*36 carriers, respectively. The median concentration and interquartile range (IQR) of [RAL]plasma were 88.5 ng/ml (41.0-236), 168 ng/ml (85.8-318) and 92.5 ng/ml (36.4-316) for UGT1A1*1/*1, UGT1A1*28 and UGT1A1*36 carriers, respectively. Only the difference between UGT1A1*1/*1 and *28 carriers was statistically significant (p = 0.022). The median MR (IQR) were 5.8 (3-10), 2.9 (1.6-5.3) and 3.2 (1.7-5.9) for UGT1A1*1/*1, UGT1A1*28 and UGT1A1*36 carriers, respectively. Only the difference between UGT1A1*1/*1 and *28 carriers was statistically significant (p = 0.004) with an allele-dependent effect: UGT1A1*28 homozygous having lower MR than heterozygous carriers who show lower MR compared to *1/*1. Except for the sensation of fatigue, this PK effect did not correlate with clinical adverse events or biological abnormalities. In Conclusion, we demonstrate that UGT1A1*28 polymorphism has a significant impact on RAL metabolism: UGT1A1*28 carriers being characterized by higher [RAL]plasma and lower MR.
Collapse
Affiliation(s)
- Leïla Belkhir
- AIDS Reference center, Cliniques Universitaires Saint-Luc, 1200, Brussels, Belgium. .,Louvain centre for Toxicology and Applied Pharmacology (LTAP), Institut de recherche expérimentale et clinique (IREC), Université catholique de Louvain (UCL), 1200, Brussels, Belgium.
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Laure Elens
- Louvain centre for Toxicology and Applied Pharmacology (LTAP), Institut de recherche expérimentale et clinique (IREC), Université catholique de Louvain (UCL), 1200, Brussels, Belgium.,Louvain Drug Research institute, UCL, 1200, Brussels, Belgium
| | - Caroline Pauly
- Department of toxicology, Laboratoire National de Santé, 3555, Dudelange, Luxembourg
| | - Nicolas Gengler
- Department of toxicology, Laboratoire National de Santé, 3555, Dudelange, Luxembourg
| | - Serge Schneider
- Department of toxicology, Laboratoire National de Santé, 3555, Dudelange, Luxembourg
| | - Jean Ruelle
- AIDS reference laboratory, IREC, UCL, 1200, Brussels, Belgium
| | - Vincent Haufroid
- Louvain centre for Toxicology and Applied Pharmacology (LTAP), Institut de recherche expérimentale et clinique (IREC), Université catholique de Louvain (UCL), 1200, Brussels, Belgium.,Department of Clinical Chemistry, Cliniques Universitaires Saint-Luc, 1200, Brussels, Belgium
| | - Bernard Vandercam
- AIDS Reference center, Cliniques Universitaires Saint-Luc, 1200, Brussels, Belgium
| |
Collapse
|
54
|
Naidoo A, Ramsuran V, Chirehwa M, Denti P, McIlleron H, Naidoo K, Yende-Zuma N, Singh R, Ngcapu S, Chaudhry M, Pepper MS, Padayatchi N. Effect of genetic variation in UGT1A and ABCB1 on moxifloxacin pharmacokinetics in South African patients with tuberculosis. Pharmacogenomics 2018; 19:17-29. [PMID: 29210323 PMCID: PMC5753622 DOI: 10.2217/pgs-2017-0144] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 10/10/2017] [Indexed: 01/10/2023] Open
Abstract
AIM We assessed the effect of genetic variability in UGT1A and ABCB1 genes on moxifloxacin pharmacokinetics. METHODS Genotypes for selected UGT1A and ABCB1 SNPs were determined using a TaqMan® Genotyping OpenArray™ and high-resolution melt analysis for rs8175347. A nonlinear mixed-effects model was used to describe moxifloxacin pharmacokinetics. RESULTS Genotypes of UGT1A SNPs, rs8175347 and rs3755319 (20.6% lower and 11.6% increased clearance, respectively) and ABCB1 SNP rs2032582 (40% reduced bioavailability in one individual) were significantly associated with changes in moxifloxacin pharmacokinetic parameters. CONCLUSION Genetic variation in UGT1A as represented by rs8175347 to a lesser extent rs3755319 and the ABCB1 rs2032582 SNP is modestly associated with the interindividual variability reported in moxifloxacin pharmacokinetics and exposure. Clinical relevance of the effects of genetic variation on moxifloxacin pharmacokinetic requires further investigation.
Collapse
Affiliation(s)
- Anushka Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Veron Ramsuran
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
- School of Laboratory Medicine & Medical Sciences, University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Maxwell Chirehwa
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Western Cape, South Africa
| | - Paolo Denti
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Western Cape, South Africa
| | - Helen McIlleron
- Department of Medicine, Division of Clinical Pharmacology, University of Cape Town, Western Cape, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
- MRC-CAPRISA HIV-TB Pathogenesis & Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, KwaZulu-Natal, South Africa
| | - Nonhlanhla Yende-Zuma
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Ravesh Singh
- Department of Microbiology, National Health Laboratory Services, KZN Academic Complex, Inkosi Albert Luthuli Central Hospital, Durban, KwaZulu-Natal, South Africa
| | - Sinaye Ngcapu
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
| | - Mamoonah Chaudhry
- Department of Immunology & the Institute for Cellular & Molecular Medicine; South African Medical Research Council Extramural Unit for Stem Cell Research & Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Michael S Pepper
- Department of Immunology & the Institute for Cellular & Molecular Medicine; South African Medical Research Council Extramural Unit for Stem Cell Research & Therapy, Faculty of Health Sciences, University of Pretoria, Pretoria, Gauteng, South Africa
| | - Nesri Padayatchi
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, KwaZulu-Natal, South Africa
- MRC-CAPRISA HIV-TB Pathogenesis & Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, KwaZulu-Natal, South Africa
| |
Collapse
|
55
|
Howell S, Marshall K, Reid M, McFarlane-Anderson N, McKenzie C. A cross-sectional clinic-based study exploring whether variants within the glutathione S-transferase, haptoglobin and uridine 5'-diphospho-glucuronosyltransferase 1A1 genes are associated with interindividual phenotypic variation in sickle cell anaemia in Jamaica. Eur J Haematol 2017; 100:147-153. [PMID: 29114966 DOI: 10.1111/ejh.12993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2017] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To explore putative associations between specific variants in either the glutathione S-transferase (GST), haptoglobin (HP) or uridine 5'-diphospho-glucuronosyltransferase 1A1 (UGT1A1) genes and clinically important phenotypes in sickle cell anaemia (HbSS). METHODS 371 HbSS participants were recruited from the Sickle Cell Clinic of the Sickle Cell Unit at the University of the West Indies, Kingston, Jamaica. Markers within four GST superfamily genes, the HP gene and the UGT1A1 gene were analysed using PCR-based assays. RESULTS Multivariable regression revealed statistically significant associations between the GSTP1 Ile105Val heterozygote and HbA2 levels (P = .016), HbF percentage (P = .001), MCH concentration (P = .028) and reticulocyte count (P = .032), while the GSTM3 D/D homozygote was significantly associated with HbA2 levels (P = .032). The UGT1A1 (TA)6 /(TA)8 heterozygote showed statistically significant associations with HbA2 levels (P = .019), HbF percentage (P < .001), haemoglobin levels (P = .008), PCV values (P = .007) and RBC counts (P = .041). CONCLUSION This exploratory cross-sectional study has generated novel and informative genotype-phenotype estimates of association, but larger studies are needed to determine whether these specific variants within the GST, UGT1A1 and HP genes are related to interindividual phenotypic variability in HbSS.
Collapse
Affiliation(s)
- Sharon Howell
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research (formerly Tropical Medicine Research Institute), The University of the West Indies, Mona, Jamaica
| | - Kwesi Marshall
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research (formerly Tropical Medicine Research Institute), The University of the West Indies, Mona, Jamaica
| | - Marvin Reid
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research (formerly Tropical Medicine Research Institute), The University of the West Indies, Mona, Jamaica
| | | | - Colin McKenzie
- Tropical Metabolism Research Unit, Caribbean Institute for Health Research (formerly Tropical Medicine Research Institute), The University of the West Indies, Mona, Jamaica
| |
Collapse
|
56
|
Labriet A, De Mattia E, Cecchin E, Lévesque É, Jonker D, Couture F, Buonadonna A, D'Andrea M, Villeneuve L, Toffoli G, Guillemette C. Improved Progression-Free Survival in Irinotecan-Treated Metastatic Colorectal Cancer Patients Carrying the HNF1A Coding Variant p.I27L. Front Pharmacol 2017; 8:712. [PMID: 29066969 PMCID: PMC5641335 DOI: 10.3389/fphar.2017.00712] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 09/22/2017] [Indexed: 01/15/2023] Open
Abstract
Hepatocyte nuclear factor 1-alpha (HNF1A) is a liver-enriched transcription factor that plays a key role in many aspects of hepatic functions including detoxification processes. We examined whether HNF1A polymorphisms are associated with clinical outcomes in two independent cohorts combining 417 European ancestry patients with metastatic colorectal cancer (mCRC) treated with irinotecan-based chemotherapy. The intronic rs2244608A>G marker was predictive of an improved progression-free survival with a trend in the Canadian cohort and reaching significance in the Italian cohort, with hazard ratios (HR) of 0.74 and 0.72, P = 0.076 and 0.038, respectively. A strong association between rs2244608A>G and improved PFS was found in the combined analysis of both cohorts (HR = 0.72; P = 0.002). Consistent with an altered HNF1A function, mCRC carriers of the rs2244608G minor allele displayed enhanced drug exposure by 45% (P = 0.032) compared to non-carriers. In Caucasians, rs2244608A>G is in strong linkage with the coding variant rs1169288c.79A>C (HNF1A p.I27L). In healthy donors, we observed an altered hepatic (ABCC1, P = 0.009, ABCC2, P = 0.048 and CYP3A5, P = 0.001; n = 89) and intestinal (TOP1, P = 0.004; n = 75) gene expression associated with the rs1169288C allele. In addition, the rs1169288C polymorphism could significantly increase the ABCC1 promoter activity by 27% (P = 0.008) and 15% (P = 0.041) in the human kidney HEK293 and the human liver HepG2 cell lines, respectively. Our findings suggest that the HNF1A rs2244608, or the tightly linked functional coding variant p.I27L, might be a potential prognostic marker with irinotecan-based regimens.
Collapse
Affiliation(s)
- Adrien Labriet
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Elena De Mattia
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Erika Cecchin
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Éric Lévesque
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Derek Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, ON, Canada
| | - Félix Couture
- Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - Angela Buonadonna
- Medical Oncology Unit, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Mario D'Andrea
- Medical Oncology Unit, San Filippo Neri Hospital, Rome, Italy
| | - Lyne Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - Giuseppe Toffoli
- Clinical and Experimental Pharmacology, IRCCS National Cancer Institute 'Centro di Riferimento Oncologico', Aviano, Italy
| | - Chantal Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| |
Collapse
|
57
|
Marques D, Ferreira-Costa LR, Ferreira-Costa LL, Correa RDS, Borges AMP, Ito FR, Ramos CCDO, Bortolin RH, Luchessi AD, Ribeiro-dos-Santos Â, Santos S, Silbiger VN. Association of insertion-deletions polymorphisms with colorectal cancer risk and clinical features. World J Gastroenterol 2017; 23:6854-6867. [PMID: 29085228 PMCID: PMC5645618 DOI: 10.3748/wjg.v23.i37.6854] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 06/24/2017] [Accepted: 08/15/2017] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the association between 16 insertion-deletions (INDEL) polymorphisms, colorectal cancer (CRC) risk and clinical features in an admixed population.
METHODS One hundred and forty patients with CRC and 140 cancer-free subjects were examined. Genomic DNA was extracted from peripheral blood samples. Polymorphisms and genomic ancestry distribution were assayed by Multiplex-PCR reaction, separated by capillary electrophoresis on the ABI 3130 Genetic Analyzer instrument and analyzed on GeneMapper ID v3.2. Clinicopathological data were obtained by consulting the patients’ clinical charts, intra-operative documentation, and pathology scoring.
RESULTS Logistic regression analysis showed that polymorphism variations in IL4 gene was associated with increased CRC risk, while TYMS and UCP2 genes were associated with decreased risk. Reference to anatomical localization of tumor Del allele of NFKB1 and CASP8 were associated with more colon related incidents than rectosigmoid. In relation to the INDEL association with tumor node metastasis (TNM) stage risk, the Ins alleles of ACE, HLAG and TP53 (6 bp INDEL) were associated with higher TNM stage. Furthermore, regarding INDEL association with relapse risk, the Ins alleles of ACE, HLAG, and UGT1A1 were associated with early relapse risk, as well as the Del allele of TYMS. Regarding INDEL association with death risk before 10 years, the Ins allele of SGSM3 and UGT1A1 were associated with death risk.
CONCLUSION The INDEL variations in ACE, UCP2, TYMS, IL4, NFKB1, CASP8, TP53, HLAG, UGT1A1, and SGSM3 were associated with CRC risk and clinical features in an admixed population. These data suggest that this cancer panel might be useful as a complementary tool for better clinical management, and more studies need to be conducted to confirm these findings.
Collapse
Affiliation(s)
- Diego Marques
- Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Programa de Pós-graduação em Ciências Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66055-080, Pará, Brazil
| | - Layse Raynara Ferreira-Costa
- Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
| | - Lorenna Larissa Ferreira-Costa
- Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
| | - Romualdo da Silva Correa
- Departamento de Cirurgia Oncológica, Liga Norte Riograndense Contra o Câncer, Natal 59040-000, Rio Grande do Norte, Brazil
| | - Aline Maciel Pinheiro Borges
- Departamento de Cirurgia Oncológica, Liga Norte Riograndense Contra o Câncer, Natal 59040-000, Rio Grande do Norte, Brazil
| | - Fernanda Ribeiro Ito
- Departamento de Cirurgia Oncológica, Liga Norte Riograndense Contra o Câncer, Natal 59040-000, Rio Grande do Norte, Brazil
| | - Carlos Cesar de Oliveira Ramos
- Laboratório de Patologia e Citopatologia, Liga Norte Riograndense Contra o Câncer, Natal 59040-000, Rio Grande do Norte, Brazil
| | - Raul Hernandes Bortolin
- Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Programa de Pós-graduação em Ciências Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
| | - André Ducati Luchessi
- Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Programa de Pós-graduação em Ciências Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
| | - Ândrea Ribeiro-dos-Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66055-080, Pará, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil
| | - Sidney Santos
- Laboratório de Genética Humana e Médica, Universidade Federal do Pará, Belém 66055-080, Pará, Brazil
- Núcleo de Pesquisas em Oncologia, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil
| | - Vivian Nogueira Silbiger
- Laboratório de Bioanálise e Biotecnologia Molecular, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Departamento de Análises Clínicas e Toxicológicas, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
- Programa de Pós-graduação em Ciências Farmacêutica, Universidade Federal do Rio Grande do Norte, Natal 59012-570, Rio Grande do Norte, Brazil
| |
Collapse
|
58
|
Characterisation of the HLA-DRB1*07:01 biomarker for lapatinib-induced liver toxicity during treatment of early-stage breast cancer patients with lapatinib in combination with trastuzumab and/or taxanes. THE PHARMACOGENOMICS JOURNAL 2017; 18:480-486. [PMID: 28786423 DOI: 10.1038/tpj.2017.39] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 05/18/2017] [Accepted: 06/07/2017] [Indexed: 01/11/2023]
Abstract
HLA-DRB1*07:01 allele carriage was characterised as a risk biomarker for lapatinib-induced liver injury in a large global study evaluating lapatinib, alone and in combination with trastuzumab and taxanes, as adjuvant therapy for advanced breast cancer (adjuvant lapatinib and/or trastuzumab treatment optimisation). HLA-DRB1*07:01 carriage was associated with serum alanine aminotransferase (ALT) elevations in lapatinib-treated patients (odds ratio 6.5, P=3 × 10-26, n=4482) and the risk and severity of ALT elevation for lapatinib-treated patients was higher in homozygous than heterozygous HLA-DRB1*07:01 genotype carriers. A higher ALT case incidence plus weaker HLA association observed during concurrent administration of lapatinib and taxane suggested a subset of liver injury in this combination group that was HLA-DRB1*07:01 independent. Furthermore, the incidence of ALT elevation demonstrated an expected correlation with geographic HLA-DRB1*07:01 carriage frequency. Robust ALT elevation risk estimates for HLA-DRB1*07:01 may support causality discrimination and safety risk management during the use of lapatinib combination therapy for the treatment of metastatic breast cancer.
Collapse
|
59
|
Naidoo A, Naidoo K, McIlleron H, Essack S, Padayatchi N. A Review of Moxifloxacin for the Treatment of Drug-Susceptible Tuberculosis. J Clin Pharmacol 2017; 57:1369-1386. [PMID: 28741299 DOI: 10.1002/jcph.968] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 05/21/2017] [Indexed: 11/08/2022]
Abstract
Moxifloxacin, an 8-methoxy quinolone, is an important drug in the treatment of multidrug-resistant tuberculosis and is being investigated in novel drug regimens with pretomanid, bedaquiline, and pyrazinamide, or rifapentine, for the treatment of drug-susceptible tuberculosis. Early results of these studies are promising. Although current evidence does not support the use of moxifloxacin in treatment-shortening regimens for drug-susceptible tuberculosis, it may be recommended in patients unable to tolerate standard first-line drug regimens or for isoniazid monoresistance. Evidence suggests that the standard 400-mg dose of moxifloxacin used in the treatment of tuberculosis may be suboptimal in some patients, leading to worse tuberculosis treatment outcomes and emergence of drug resistance. Furthermore, a drug interaction with the rifamycins results in up to 31% reduced plasma concentrations of moxifloxacin when these are combined for treatment of drug-susceptible tuberculosis, although the clinical relevance of this interaction is unclear. Moxifloxacin exhibits extensive interindividual pharmacokinetic variability. Higher doses of moxifloxacin may be needed to achieve drug exposures required for improved clinical outcomes. Further study is, however, needed to determine the safety of proposed higher doses and clinically validated targets for drug exposure to moxifloxacin associated with improved tuberculosis treatment outcomes. We discuss in this review the evidence for the use of moxifloxacin in drug-susceptible tuberculosis and explore the role of moxifloxacin pharmacokinetics, pharmacodynamics, and drug interactions with rifamycins, on tuberculosis treatment outcomes when used in first-line tuberculosis drug regimens.
Collapse
Affiliation(s)
- Anushka Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa
| | - Kogieleum Naidoo
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| | - Helen McIlleron
- Division of Clinical Pharmacology, Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Sabiha Essack
- Antimicrobial Research Unit, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Nesri Padayatchi
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), University of KwaZulu-Natal, Durban, South Africa.,MRC-CAPRISA HIV-TB Pathogenesis and Treatment Research Unit, Doris Duke Medical Research Institute, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
60
|
Del Re M, Latiano T, Fidilio L, Restante G, Morelli F, Maiello E, Danesi R. Unusual gastrointestinal and cutaneous toxicities by bleomycin, etoposide, and cisplatin: a case report with pharmacogenetic analysis to personalize treatment. EPMA J 2017; 8:69-73. [PMID: 28620445 DOI: 10.1007/s13167-017-0080-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/24/2017] [Indexed: 11/26/2022]
Abstract
The standard treatment of testicular germ cell tumors is based on the combination of bleomycin, etoposide, and cisplatin (PEB). However, this treatment may be associated with severe adverse reactions, such as hematological and non-hematological toxicities. Here, we report a case of a patient suffering from severe PEB-related toxicities, to whom pharmacogenetic analyses were performed, comprising a panel of genes involved in PEB metabolism. The analysis revealed the presence of a complex pattern of polymorphisms in GSTP1, UGT1A1 (TA)6/7, UGT1A7, and ABCB1. The present case shows that a pharmacogenetic approach can help in the management of adverse drug reactions in order to predict, prevent, and personalize treatments.
Collapse
Affiliation(s)
- Marzia Del Re
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 55, Via Roma, 56126 Pisa, Italy
| | - Tiziana Latiano
- Medical Oncology Unit, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Foggia Italy
| | - Leonardo Fidilio
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 55, Via Roma, 56126 Pisa, Italy
| | - Giuliana Restante
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 55, Via Roma, 56126 Pisa, Italy
| | - Franco Morelli
- Medical Oncology Unit, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Foggia Italy
| | - Evaristo Maiello
- Medical Oncology Unit, Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Foggia Italy
| | - Romano Danesi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, 55, Via Roma, 56126 Pisa, Italy
| |
Collapse
|
61
|
Characteristics of the heme catabolic pathway in mild unconjugated hyperbilirubinemia and their associations with inflammation and disease prevention. Sci Rep 2017; 7:755. [PMID: 28389660 PMCID: PMC5429724 DOI: 10.1038/s41598-017-00933-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 03/17/2017] [Indexed: 01/03/2023] Open
Abstract
Heme catabolism exerts physiological functions that impact health through depressing inflammation. Upon reactive pathway progression, as in Gilbert’s Syndrome (GS; UGT1A1*28 polymorphism), aggravated health effects have been determined. Based on lower inflammation and improved metabolic health reported for GS, inter-group differences in heme catabolism were explored. Therefore, a case-control study including 120 fasted, healthy, age- and gender matched subjects with/without GS, was conducted. Genetic expressions of HMOX-1 and BLVRA were measured. Additionally participants were genotyped for those polymorphisms that are known (UGT1A1*28) or likely (HMOX-1 microsatellites) to impact bilirubinemia. Intracellular interleukins (IL-6, IL-1β, TNFα), circulatory C-reactive protein (CRP), serum amyloid A (SAA) and haptoglobin (Hpt) were analysed as inflammatory markers. To assess intracellular heme oxygenase 1 (HO-1) isolated PBMCs were used. In GS vs. C, inflammation markers were significantly decreased. This was supported by an altered heme catabolism, indirectly reflecting in elevated unconjugated bilirubin (UCB; main phenotypic feature of GS) and iron, decreased hemopexin (Hpx) and Hpt and in up-regulated biliverdin reductase (BLVRA) gene expressions. Moreover, HMOX (GT)n short alleles were non-significantly more prominent in female GS individuals. Herewith, we propose a concept to elucidate why GS individuals encounter lower inflammation, and are thus less prone to oxidative-stress mediated diseases.
Collapse
|
62
|
Abstract
Cancer treatment is becoming more and more individually based as a result of the large inter-individual differences that exist in treatment outcome and toxicity when patients are treated using population-based drug doses. Polymorphisms in genes encoding drug-metabolizing enzymes and transporters can significantly influence uptake, metabolism, and elimination of anticancer drugs. As a result, the altered pharmacokinetics can greatly influence drug efficacy and toxicity. Pharmacogenetic screening and/or drug-specific phenotyping of cancer patients eligible for treatment with chemotherapeutic drugs, prior to the start of anticancer treatment, can identify patients with tumors that are likely to be responsive or resistant to the proposed drugs. Similarly, the identification of patients with an increased risk of developing toxicity would allow either dose adaptation or the application of other targeted therapies. This review focuses on the role of genetic polymorphisms significantly altering the pharmacokinetics of anticancer drugs. Polymorphisms in DPYD, TPMT, and UGT1A1 have been described that have a major impact on the pharmacokinetics of 5-fluorouracil, mercaptopurine, and irinotecan, respectively. For other drugs, however, the association of polymorphisms with pharmacokinetics is less clear. To date, the influence of genetic variations on the pharmacokinetics of the increasingly used monoclonal antibodies has hardly been investigated. Some studies indicate that genes encoding the Fcγ-receptor family are of interest, but more research is needed to establish if screening before the start of therapy is beneficial. Considering the profound impact of polymorphisms in drug transporters and drug-metabolizing enzymes on the pharmacokinetics of chemotherapeutic drugs and hence, their toxicity and efficacy, pharmacogenetic and pharmacokinetic profiling should become the standard of care.
Collapse
Affiliation(s)
| | | | - André B P van Kuilenburg
- Department of Clinical Chemistry, Laboratory Genetic Metabolic Diseases, Academic Medical Center, University of Amsterdam, Emma Children's Hospital, F0-220, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
63
|
Abstract
It is well established that variations in genes can alter the pharmacokinetic and pharmacodynamic profile of a drug and immunological responses to it. Early advances in pharmacogenetics were made with traditional genetic techniques such as functional cloning of genes using knowledge gained from purified proteins, and candidate gene analysis. Over the past decade, techniques for analysing the human genome have accelerated greatly as knowledge and technological capabilities have grown. These techniques were initially focussed on understanding genetic factors of disease, but increasingly they are helping to clarify the genetic basis of variable drug responses and adverse drug reactions (ADRs). We examine genetic methods that have been applied to the understanding of ADRs, review the current state of knowledge of genetic factors that influence ADR development, and discuss how the application of genome-wide association studies and next-generation sequencing approaches is supporting and extending existing knowledge of pharmacogenetic processes leading to ADRs. Such approaches have identified single genes that are major contributing genetic risk factors for an ADR, (such as flucloxacillin and drug-induced liver disease), making pre-treatment testing a possibility. They have contributed to the identification of multiple genetic determinants of a single ADR, some involving both pharmacologic and immunological processes (such as phenytoin and severe cutaneous adverse reactions). They have indicated that rare genetic variants, often not previously reported, are likely to have more influence on the phenotype than common variants that have been traditionally tested for. The problem of genotype/phenotype discordance affecting the interpretation of pharmacogenetic screening and the future of genome-based testing applied to ADRs are also discussed.
Collapse
|
64
|
Van Cutsem E, Cervantes A, Adam R, Sobrero A, Van Krieken JH, Aderka D, Aranda Aguilar E, Bardelli A, Benson A, Bodoky G, Ciardiello F, D'Hoore A, Diaz-Rubio E, Douillard JY, Ducreux M, Falcone A, Grothey A, Gruenberger T, Haustermans K, Heinemann V, Hoff P, Köhne CH, Labianca R, Laurent-Puig P, Ma B, Maughan T, Muro K, Normanno N, Österlund P, Oyen WJG, Papamichael D, Pentheroudakis G, Pfeiffer P, Price TJ, Punt C, Ricke J, Roth A, Salazar R, Scheithauer W, Schmoll HJ, Tabernero J, Taïeb J, Tejpar S, Wasan H, Yoshino T, Zaanan A, Arnold D. ESMO consensus guidelines for the management of patients with metastatic colorectal cancer. Ann Oncol 2016; 27:1386-422. [PMID: 27380959 DOI: 10.1093/annonc/mdw235] [Citation(s) in RCA: 2256] [Impact Index Per Article: 282.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 05/31/2016] [Indexed: 02/11/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignancies in Western countries. Over the last 20 years, and the last decade in particular, the clinical outcome for patients with metastatic CRC (mCRC) has improved greatly due not only to an increase in the number of patients being referred for and undergoing surgical resection of their localised metastatic disease but also to a more strategic approach to the delivery of systemic therapy and an expansion in the use of ablative techniques. This reflects the increase in the number of patients that are being managed within a multidisciplinary team environment and specialist cancer centres, and the emergence over the same time period not only of improved imaging techniques but also prognostic and predictive molecular markers. Treatment decisions for patients with mCRC must be evidence-based. Thus, these ESMO consensus guidelines have been developed based on the current available evidence to provide a series of evidence-based recommendations to assist in the treatment and management of patients with mCRC in this rapidly evolving treatment setting.
Collapse
Affiliation(s)
- E Van Cutsem
- Digestive Oncology, University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| | - A Cervantes
- Medical Oncology Department, INCLIVA University of Valencia, Valencia, Spain
| | - R Adam
- Hepato-Biliary Centre, Paul Brousse Hospital, Villejuif, France
| | - A Sobrero
- Medical Oncology, IRCCS San Martino Hospital, Genova, Italy
| | - J H Van Krieken
- Research Institute for Oncology, Radboud University Nijmegen Medical Center, Nijmegen, The Netherlands
| | - D Aderka
- Division of Oncology, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - E Aranda Aguilar
- Medical Oncology Department, University Hospital Reina Sofia, Cordoba, Spain
| | - A Bardelli
- School of Medicine, University of Turin, Turin, Italy
| | - A Benson
- Division of Hematology/Oncology, Northwestern Medical Group, Chicago, USA
| | - G Bodoky
- Department of Oncology, St László Hospital, Budapest, Hungary
| | - F Ciardiello
- Division of Medical Oncology, Seconda Università di Napoli, Naples, Italy
| | - A D'Hoore
- Abdominal Surgery, University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| | - E Diaz-Rubio
- Medical Oncology Department, Hospital Clínico San Carlos, Madrid, Spain
| | - J-Y Douillard
- Medical Oncology, Institut de Cancérologie de l'Ouest (ICO), St Herblain
| | - M Ducreux
- Department of Medical Oncology, Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - A Falcone
- Department of Medical Oncology, University of Pisa, Pisa, Italy Division of Medical Oncology, Department of Oncology, University Hospital 'S. Chiara', Istituto Toscano Tumori, Pisa, Italy
| | - A Grothey
- Division of Medical Oncology, Mayo Clinic, Rochester, USA
| | - T Gruenberger
- Department of Surgery I, Rudolfstiftung Hospital, Vienna, Austria
| | - K Haustermans
- Department of Radiation Oncology, University Hospitals Gasthuisberg and KU Leuven, Leuven, Belgium
| | - V Heinemann
- Comprehensive Cancer Center, University Clinic Munich, Munich, Germany
| | - P Hoff
- Instituto do Câncer do Estado de São Paulo, University of São Paulo, São Paulo, Brazil
| | - C-H Köhne
- Northwest German Cancer Center, University Campus Klinikum Oldenburg, Oldenburg, Germany
| | - R Labianca
- Cancer Center, Ospedale Giovanni XXIII, Bergamo, Italy
| | - P Laurent-Puig
- Digestive Oncology Department, European Hospital Georges Pompidou, Paris, France
| | - B Ma
- Department of Clinical Oncology, Prince of Wales Hospital, State Key Laboratory in Oncology in South China, Chinese University of Hong Kong, Shatin, Hong Kong
| | - T Maughan
- CRUK/MRC Oxford Institute for Radiation Oncology, Gray Laboratories, University of Oxford, Oxford, UK
| | - K Muro
- Department of Clinical Oncology and Outpatient Treatment Center, Aichi Cancer Center Hospital, Nagoya, Japan
| | - N Normanno
- Cell Biology and Biotherapy Unit, I.N.T. Fondazione G. Pascale, Napoli, Italy
| | - P Österlund
- Helsinki University Central Hospital, Comprehensive Cancer Center, Helsinki, Finland Department of Oncology, University of Helsinki, Helsinki, Finland
| | - W J G Oyen
- The Institute of Cancer Research and The Royal Marsden Hospital, London, UK
| | - D Papamichael
- Department of Medical Oncology, Bank of Cyprus Oncology Centre, Nicosia, Cyprus
| | - G Pentheroudakis
- Department of Medical Oncology, University of Ioannina, Ioannina, Greece
| | - P Pfeiffer
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - T J Price
- Haematology and Medical Oncology Unit, Queen Elizabeth Hospital, Woodville, Australia
| | - C Punt
- Department of Medical Oncology, Academic Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - J Ricke
- Department of Radiology and Nuclear Medicine, University Clinic Magdeburg, Magdeburg, Germany
| | - A Roth
- Digestive Tumors Unit, Geneva University Hospitals (HUG), Geneva, Switzerland
| | - R Salazar
- Catalan Institute of Oncology (ICO), Barcelona, Spain
| | - W Scheithauer
- Department of Internal Medicine I and Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - H J Schmoll
- Department of Internal Medicine IV, University Clinic Halle, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - J Tabernero
- Medical Oncology Department, Vall d' Hebron University Hospital, Vall d'Hebron Institute of Oncology (V.H.I.O.), Barcelona, Spain
| | - J Taïeb
- Digestive Oncology Department, European Hospital Georges Pompidou, Paris, France
| | - S Tejpar
- Digestive Oncology, University Hospitals Gasthuisberg Leuven and KU Leuven, Leuven, Belgium
| | - H Wasan
- Department of Cancer Medicine, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - T Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - A Zaanan
- Digestive Oncology Department, European Hospital Georges Pompidou, Paris, France
| | - D Arnold
- Instituto CUF de Oncologia (ICO), Lisbon, Portugal
| |
Collapse
|
65
|
Thomas S, Wolstencroft K, de Bono B, Hunter PJ. A physiome interoperability roadmap for personalized drug development. Interface Focus 2016; 6:20150094. [PMID: 27051513 DOI: 10.1098/rsfs.2015.0094] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The goal of developing therapies and dosage regimes for characterized subgroups of the general population can be facilitated by the use of simulation models able to incorporate information about inter-individual variability in drug disposition (pharmacokinetics), toxicity and response effect (pharmacodynamics). Such observed variability can have multiple causes at various scales, ranging from gross anatomical differences to differences in genome sequence. Relevant data for many of these aspects, particularly related to molecular assays (known as '-omics'), are available in online resources, but identification and assignment to appropriate model variables and parameters is a significant bottleneck in the model development process. Through its efforts to standardize annotation with consequent increase in data usability, the human physiome project has a vital role in improving productivity in model development and, thus, the development of personalized therapy regimes. Here, we review the current status of personalized medicine in clinical practice, outline some of the challenges that must be overcome in order to expand its applicability, and discuss the relevance of personalized medicine to the more widespread challenges being faced in drug discovery and development. We then review some of (i) the key data resources available for use in model development and (ii) the potential areas where advances made within the physiome modelling community could contribute to physiologically based pharmacokinetic and physiologically based pharmacokinetic/pharmacodynamic modelling in support of personalized drug development. We conclude by proposing a roadmap to further guide the physiome community in its on-going efforts to improve data usability, and integration with modelling efforts in the support of personalized medicine development.
Collapse
Affiliation(s)
- Simon Thomas
- Cyprotex Discovery Ltd , 15 Beech Lane, Macclesfield SK10 2DR , UK
| | - Katherine Wolstencroft
- Leiden Institute of Advanced Computer Science , Leiden University , 111 Snellius, Niels Bohrweg 1, 2333 CA Leiden , The Netherlands
| | - Bernard de Bono
- Farr Institute, University College London, London NW1 2DA, UK; Auckland Bioengineering Institute, The University of Auckland, Auckland 1010, New Zealand
| | - Peter J Hunter
- Auckland Bioengineering Institute , The University of Auckland , Auckland 1010 , New Zealand
| |
Collapse
|
66
|
Gammal RS, Court MH, Haidar CE, Iwuchukwu OF, Gaur AH, Alvarellos M, Guillemette C, Lennox JL, Whirl‐Carrillo M, Brummel SS, Ratain MJ, Klein TE, Schackman BR, Caudle KE, Haas DW. Clinical Pharmacogenetics Implementation Consortium (CPIC) Guideline for UGT1A1 and Atazanavir Prescribing. Clin Pharmacol Ther 2016; 99:363-9. [PMID: 26417955 PMCID: PMC4785051 DOI: 10.1002/cpt.269] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 09/24/2015] [Indexed: 01/09/2023]
Abstract
The antiretroviral protease inhibitor atazanavir inhibits hepatic uridine diphosphate glucuronosyltransferase (UGT) 1A1, thereby preventing the glucuronidation and elimination of bilirubin. Resultant indirect hyperbilirubinemia with jaundice can cause premature discontinuation of atazanavir. Risk for bilirubin-related discontinuation is highest among individuals who carry two UGT1A1 decreased function alleles (UGT1A1*28 or *37). We summarize published literature that supports this association and provide recommendations for atazanavir prescribing when UGT1A1 genotype is known (updates at www.pharmgkb.org).
Collapse
Affiliation(s)
- RS Gammal
- Department of Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - MH Court
- Individualized Medicine Program, Department of Veterinary Clinical SciencesWashington State University College of Veterinary MedicinePullmanWashingtonUSA
| | - CE Haidar
- Department of Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - OF Iwuchukwu
- Division of Pharmaceutical SciencesFairleigh Dickinson University School of PharmacyFlorham ParkNew JerseyUSA
- Division of Clinical Pharmacology, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - AH Gaur
- Department of Infectious DiseasesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - M Alvarellos
- Department of GeneticsStanford UniversityStanfordCaliforniaUSA
| | - C Guillemette
- Laval University CHU de Québec Research CenterQuebecQuebecCanada
| | - JL Lennox
- Division of Infectious DiseaseEmory University School of MedicineAtlantaGeorgiaUSA
| | | | - SS Brummel
- Center for Biostatistics in AIDS ResearchHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - MJ Ratain
- Center for Personalized Therapeutics, Comprehensive Cancer CenterThe University of ChicagoChicagoIllinoisUSA
| | - TE Klein
- Department of GeneticsStanford UniversityStanfordCaliforniaUSA
| | - BR Schackman
- Department of Healthcare Policy and ResearchWeill Cornell Medical CollegeNew YorkNew YorkUSA
| | - KE Caudle
- Department of Pharmaceutical SciencesSt. Jude Children's Research HospitalMemphisTennesseeUSA
| | - DW Haas
- Departments of Medicine, Pharmacology, Pathology, Microbiology & ImmunologyVanderbilt University School of MedicineNashvilleTennesseeUSA
| |
Collapse
|
67
|
Claudio-Campos K, Duconge J, Cadilla CL, Ruaño G. Pharmacogenetics of drug-metabolizing enzymes in US Hispanics. Drug Metab Pers Ther 2016; 30:87-105. [PMID: 25431893 DOI: 10.1515/dmdi-2014-0023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 10/02/2014] [Indexed: 12/19/2022]
Abstract
Although the Hispanic population is continuously growing in the United States, they are underrepresented in pharmacogenetic studies. This review addresses the need for compiling available pharmacogenetic data in US Hispanics, discussing the prevalence of clinically relevant polymorphisms in pharmacogenes encoding for drug-metabolizing enzymes. CYP3A5*3 (0.245-0.867) showed the largest frequency in a US Hispanic population. A higher prevalence of CYP2C9*3, CYP2C19*4, and UGT2B7 IVS1+985 A>G was observed in US Hispanic vs. non-Hispanic populations. We found interethnic and intraethnic variability in frequencies of genetic polymorphisms for metabolizing enzymes, which highlights the need to define the ancestries of participants in pharmacogenetic studies. New approaches should be integrated in experimental designs to gain knowledge about the clinical relevance of the unique combination of genetic variants occurring in this admixed population. Ethnic subgroups in the US Hispanic population may harbor variants that might be part of multiple causative loci or in linkage-disequilibrium with functional variants. Pharmacogenetic studies in Hispanics should not be limited to ascertain commonly studied polymorphisms that were originally identified in their parental populations. The success of the Personalized Medicine paradigm will depend on recognizing genetic diversity between and within US Hispanics and the uniqueness of their genetic backgrounds.
Collapse
|
68
|
Abstract
Certain genetic polymorphisms of UDP glucuronosyltransferase 1 family, polypeptide A1 (UGT1A1) can reduce gene expression (*28, *60, *93) or activity (*6), thereby altering the pharmacokinetics, pharmacodynamics, and the risk of toxicities of UGT1A1 substrates, of which irinotecan is a widely-described example. This review presents an overview of the clinical effects of UGT1A1 polymorphisms on the pharmacology of UGT1A1 substrates, with a special focus on the novel histone deacetylase inhibitor belinostat. Belinostat, approved for the treatment of peripheral T-cell lymphoma, is primarily glucuronidated by UGT1A1. Recent preclinical and clinical data showed that UGT1A1*28 was associated with reduced glucuronidation in human liver microsomes, while in a retrospective analysis of a Phase I trial with patients receiving belinostat UGT1A1*60 was predominantly associated with increased belinostat plasma concentrations. Furthermore, both UGT1A1*28 and *60 variants were associated with increased incidence of thrombocytopenia and neutropenia. Using population pharmacokinetic analysis a 33% dose reduction has been proposed for patients carrying UGT1A1 variant alleles. Clinical effects of this genotype-based dosing recommendation is currently prospectively being investigated. Overall, the data suggest that UGT1A1 genotyping is useful for improving belinostat therapy.
Collapse
Affiliation(s)
- Andrew K L Goey
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - William D Figg
- Clinical Pharmacology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
69
|
ABCC5 and ABCG1 polymorphisms predict irinotecan-induced severe toxicity in metastatic colorectal cancer patients. Pharmacogenet Genomics 2015; 25:573-83. [DOI: 10.1097/fpc.0000000000000168] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
70
|
Syn NLX, Yong WP, Lee SC, Goh BC. Genetic factors affecting drug disposition in Asian cancer patients. Expert Opin Drug Metab Toxicol 2015; 11:1879-92. [PMID: 26548636 DOI: 10.1517/17425255.2015.1108964] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION In the era of genomic medicine, it is increasingly recognized that ethnogeographic differences in drug pharmacology exist between Asian and other populations. This is particularly pertinent to oncology, where drugs forming the backbone of chemotherapy often have narrow therapeutic windows and are frequently dosed close to maximally tolerable levels. AREAS COVERED At the population level, ancestry is important because historical-biogeographical confluences have shaped population genetics and pharmacoethnicity in the Asian race through allelic differentiation and interethnic differences in inheritance patterns of linkage disequilibrium. At the individual level, cis- and trans-acting germline polymorphisms and somatic mutations in genes encoding drug-metabolizing enzymes and transporters act in a multifactorial manner to determine drug disposition phenotype and clinical response in Asian cancer patients. A growing body of evidence also finds that complex genetic interactions and regulation, including a multiplicity of gene control mechanisms, are increasingly implicated in genotype-phenotype correlates than has hitherto been appreciated--potentially serving as the mechanistic links between hits in non-coding regions of genome-wide association studies and drug toxicity. Together, these genetic factors contribute to the clinical heterogeneity of drug disposition in Asian cancer patients. EXPERT OPINION This topic has broad relevance for the optimization and individualization of anticancer strategies in Asians.
Collapse
Affiliation(s)
- Nicholas Li-Xun Syn
- a Department of Haematology-Oncology , National University Cancer Institute , Singapore 119228
| | - Wei-Peng Yong
- a Department of Haematology-Oncology , National University Cancer Institute , Singapore 119228.,b Cancer Science Institute of Singapore , National University of Singapore, Centre for Translational Medicine , Singapore 117599
| | - Soo-Chin Lee
- a Department of Haematology-Oncology , National University Cancer Institute , Singapore 119228.,b Cancer Science Institute of Singapore , National University of Singapore, Centre for Translational Medicine , Singapore 117599
| | - Boon-Cher Goh
- a Department of Haematology-Oncology , National University Cancer Institute , Singapore 119228.,b Cancer Science Institute of Singapore , National University of Singapore, Centre for Translational Medicine , Singapore 117599.,c Department of Pharmacology, Yong Loo Lin School of Medicine , National University of Singapore , Singapore 119077
| |
Collapse
|
71
|
Taburet AM, Sauvageon H, Grinsztejn B, Assuied A, Veloso V, Pilotto JH, De Castro N, Grondin C, Fagard C, Molina JM. Pharmacokinetics of Raltegravir in HIV-Infected Patients on Rifampicin-Based Antitubercular Therapy. Clin Infect Dis 2015; 61:1328-35. [PMID: 26105170 DOI: 10.1093/cid/civ477] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/08/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Rifampicin (RIF) induces UGT1A1, an enzyme involved in raltegravir (RAL) elimination, thereby potentially lowering RAL exposure. We examined the pharmacokinetics of RAL in human immunodeficiency virus (HIV)-infected patients on RIF-based antitubercular therapy in the French National Agency for HIV/AIDS and Viral Hepatitis Research 12 180 Reflate Tuberculosis trial. METHODS Patients started RAL in combination with tenofovir disoproxil fumarate and lamivudine after initiation of RIF (10 mg/kg/day). In arm 1 (n = 21), they received 400 mg RAL twice daily; in arm 2 (n = 16), they received RAL 800 mg twice daily initially then 400 mg twice daily 4 weeks after RIF discontinuation. Pharmacokinetic sampling was performed over 12-hour periods, 4 weeks after initiation of RAL together with RIF (period 1), 4 weeks after RIF discontinuation (period 2), and after the RAL dose reduction in arm 2 (period 3). RESULTS In arm 1, the geometric mean ratio (GMR) between period 1 and period 2 was 0.94 (90% confidence interval [CI], .64-1.37) for the 12-hour area under the time-concentration curve (AUC0-12), and 0.69 (90% CI, .42-1.13) for the concentration at 12 hours (C12). In arm 2, the corresponding GMRs were 0.75 (90% CI, .48-1.17) and 1.10 (90% CI, .61-2.00) for period 1 vs period 2, and 1.10 (90% CI, .78-1.55) and 1.68 (90% CI, .88-3.23) for period 1 vs period 3. CONCLUSIONS The double dose of RAL overcompensated for RIF induction, but the standard dose was associated with only small decreases in AUC0-12 and C12 during RIF coadministration, warranting further evaluation in patients with HIV/tuberculosis coinfection. CLINICAL TRIALS REGISTRATION NCT0082231.
Collapse
Affiliation(s)
- Anne-Marie Taburet
- Hospital Bicetre, Assistance Publique-Hôpitaux de Paris, DHU Hepatinov; INSERM U1184, Center for Immunology of Viral Infections and Autoimmune Diseases, Université Paris-Sud, Kremlin Bicêtre
| | - Hélène Sauvageon
- Hospital Saint-Louis, Assistance Publique-Hôpitaux de Paris, France
| | - Beatriz Grinsztejn
- Instituto de Pesquisa Clinica Evandro Chagas, Fundaçao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - Alex Assuied
- Centre INSERM U897-Epidémiologie-Biostatistiques, Université de Bordeaux, Bordeaux, France
| | - Valdilea Veloso
- Instituto de Pesquisa Clinica Evandro Chagas, Fundaçao Oswaldo Cruz, Rio de Janeiro, Brazil
| | - José Henrique Pilotto
- Hospital General de Nova Iguaçu and Laboratório de AIDS e Imunologia Molecular, Rio de Janeiro, Brazil
| | | | - Carine Grondin
- Centre INSERM U897-Epidémiologie-Biostatistiques, Université de Bordeaux, Bordeaux, France
| | - Catherine Fagard
- Centre INSERM U897-Epidémiologie-Biostatistiques, Université de Bordeaux, Bordeaux, France
| | - Jean-Michel Molina
- Hospital Saint-Louis, Assistance Publique-Hôpitaux de Paris, France University Paris Diderot INSERM U941, Paris, France
| |
Collapse
|
72
|
Falvella FS, Cheli S, Martinetti A, Mazzali C, Iacovelli R, Maggi C, Gariboldi M, Pierotti MA, Di Bartolomeo M, Sottotetti E, Mennitto R, Bossi I, de Braud F, Clementi E, Pietrantonio F. DPD and UGT1A1 deficiency in colorectal cancer patients receiving triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan. Br J Clin Pharmacol 2015; 80:581-8. [PMID: 25782327 DOI: 10.1111/bcp.12631] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/22/2022] Open
Abstract
AIMS Triplet chemotherapy with fluoropyrimidines, oxaliplatin and irinotecan is a standard therapy for metastatic colorectal cancer (CRC). Single nucleotide polymorphisms (SNPs) in DPYD and UGT1A1 influence fluoropyrimdines and irinotecan adverse events (AEs). Low frequency DPYD variants (c.1905 + 1G > A, c.1679 T > G, c.2846A > T) are validated but more frequent ones (c.496A > G, c.1129-5923C > G and c.1896 T > C) are not. rs895819 T > C polymorphism in hsa-mir-27a is associated with reduced DPD activity. In this study, we evaluated the clinical usefulness of a pharmacogenetic panel for patients receiving triplet combinations. METHODS Germline DNA was available from 64 CRC patients enrolled between 2008 and 2013 in two phase II trials of capecitabine, oxaliplatin and irinotecan plus bevacizumab or cetuximab. SNPs were determined by Real-Time PCR. We evaluated the functional variants in DPYD (rare: c.1905 + 1G > A, c.1679 T > G, c.2846A > T; most common: c.496A > G, c.1129-5923C > G, c.1896 T > C), hsa-mir-27a (rs895819) and UGT1A1 (*28) genes to assess their association with grade 3-4 AEs. RESULTS None of the patients carried rare DPYD variants. We found DPYD c.496A > G, c.1129-5923C > G, c.1896 T > C in heterozygosity in 19%, 5% and 8%, respectively, homozygous rs895819 in hsa-mir-27a in 9% and homozygous UGT1A1*28 in 8%. Grade 3-4 AEs were observed in 36% patients and were associated with DPYD c.496A > G (odds ratio (OR) 4.93, 95% CI 1.29, 18.87; P = 0.021) and homozygous rs895819 in hsa-mir-27a (OR 11.11, 95% CI 1.21, 102.09; P = 0.020). Carriers of DPYD c.1896 T > C and homozygous UGT1A1*28 showed an OR of 8.42 (95% CI 0.88, 80.56; P = 0.052). Multivariate analysis confirmed an independent value for DPYD c.496A > G and c.1896 T > C. CONCLUSIONS Concomitant assessment of DPYD variants and the UGT1A1*28 allele is a promising strategy needing further validation for dose personalization.
Collapse
Affiliation(s)
- Felicia Stefania Falvella
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital 'Luigi Sacco', Università di Milano, Milan, Italy
| | - Stefania Cheli
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital 'Luigi Sacco', Università di Milano, Milan, Italy
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Cristina Mazzali
- Department of Biomedical and Clinical Sciences, University Hospital 'Luigi Sacco', Università di Milano, Milan, Italy.,Department of Management, Economics and Industrial Engineering, Politecnico di Milano, Milan, Italy
| | - Roberto Iacovelli
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Claudia Maggi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Manuela Gariboldi
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy.,FIRC Institute of Molecular Oncology Foundation, Milan, Italy
| | | | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Roberta Mennitto
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Ilaria Bossi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Emilio Clementi
- Scientific Institute, IRCCS E. Medea, Bosisio Parini, Lecco, Italy.,Unit of Clinical Pharmacology, Department of Biomedical, Clinical Sciences, Consiglio Nazionale delle Ricerche Institute of Neuroscience, University Hospital 'Luigi Sacco', Università di Milano, Milan, Italy
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
73
|
Etienne-Grimaldi MC, Boyer JC, Thomas F, Quaranta S, Picard N, Loriot MA, Narjoz C, Poncet D, Gagnieu MC, Ged C, Broly F, Le Morvan V, Bouquié R, Gaub MP, Philibert L, Ghiringhelli F, Le Guellec C. UGT1A1genotype and irinotecan therapy: general review and implementation in routine practice. Fundam Clin Pharmacol 2015; 29:219-37. [DOI: 10.1111/fcp.12117] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 02/05/2015] [Accepted: 02/24/2015] [Indexed: 12/19/2022]
Affiliation(s)
| | - Jean-Christophe Boyer
- Unité de Toxicologie; Laboratoire de Biochimie; CHU Carémeau, Place du Pr Debré; 30029 Nîmes Cedex France
| | - Fabienne Thomas
- Institut Claudius Regaud; 1, avenue Irène Joliot-Curie 31059 Toulouse France
| | - Sylvie Quaranta
- Service de Pharmacocinétique et Toxicologie; Laboratoire de Biologie Médicale; Hôpital de la Timone; Bât F; 264 rue Saint Pierre 13385 Marseille Cedex 05 France
| | - Nicolas Picard
- Service Pharmacologie; Toxicologie et Pharmacovigilance; CHU Limoges, Bâtiment CBRS; 2 avenue Martin Luther King 87042 Limoges France
| | - Marie-Anne Loriot
- Hôpital Européen Georges Pompidou; SERVICE BIOCHIMIE; 20 Rue Leblanc 75015 Paris France
| | - Céline Narjoz
- Hôpital Européen Georges Pompidou; SERVICE BIOCHIMIE; 20 Rue Leblanc 75015 Paris France
| | - Delphine Poncet
- Equipe Signalisation Métabolisme et Progression Tumorale; UMR 1052-5286; Centre Léon Bérard; 28 rue Laennec 69373 Lyon Cedex 08 69008 Lyon France
| | - Marie-Claude Gagnieu
- Fédération de Biochimie; UF Pharmacologie Spécialisée; Hôpital E. Herriot; 5 place d'Arsonval 69437 Lyon Cedex 03 France
| | - Cécile Ged
- Plateau Technique de Biologie Moléculaire; Pôle de Biologie et Pathologie; CHU de Bordeaux; 1, place A Raba Leon 33 000 Bordeaux France
| | - Franck Broly
- Service de Toxicologie et Génopathies; Centre de Biologie Pathologie Génétique; Centre Hospitalier Régional et Universitaire de Lille; 59037 Lille Cedex France
| | - Valérie Le Morvan
- Institut Bergonié; Unité Inserm VINCO; 229 cours de l'Argonne 33076 Bordeaux Cedex France
| | - Régis Bouquié
- Laboratoire de Pharmacologie clinique; Institut de Biologie - CHU Nantes; 9, quai Moncousu 44093 Nantes Cedex 1 France
| | - Marie-Pierre Gaub
- EA3430; FMTS Université de Strasbourg; Laboratoire de Biochimie- Biologie Moléculaire; Hôpital de hautepierre; Avenue Molière 67098 Strasbourg France
| | - Laurent Philibert
- Unité de Biopathologie et pharmacogénétique; Laboratoire d'oncopharmacologie; Institut régional du Cancer Montpellier - Val d'Aurelle; 208 Avenue des Apothicaires 34298 Montpellier Cedex 5 France
| | - François Ghiringhelli
- Département de biopathologie; Centre Georges Francois Leclerc; 1 rue du professeur Marion 21000 Dijon France
| | - Chantal Le Guellec
- Unité de pharmacogénétique; Laboratoire de biochimie et biologie moléculaire; CHU Bretonneau; 2 bis boulevard Tonnellé 37044 Tours France
| | | |
Collapse
|
74
|
Falvella FS, Caporale M, Cheli S, Martinetti A, Berenato R, Maggi C, Niger M, Ricchini F, Bossi I, Di Bartolomeo M, Sottotetti E, Bernardi FF, de Braud F, Clementi E, Pietrantonio F. Undetected toxicity risk in pharmacogenetic testing for dihydropyrimidine dehydrogenase. Int J Mol Sci 2015; 16:8884-95. [PMID: 25906475 PMCID: PMC4425114 DOI: 10.3390/ijms16048884] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 03/30/2015] [Accepted: 04/13/2015] [Indexed: 12/22/2022] Open
Abstract
Fluoropyrimidines, the mainstay agents for the treatment of colorectal cancer, alone or as a part of combination therapies, cause severe adverse reactions in about 10%–30% of patients. Dihydropyrimidine dehydrogenase (DPD), a key enzyme in the catabolism of 5-fluorouracil, has been intensively investigated in relation to fluoropyrimidine toxicity, and several DPD gene (DPYD) polymorphisms are associated with decreased enzyme activity and increased risk of fluoropyrimidine-related toxicity. In patients carrying non-functional DPYD variants (c.1905+1G>A, c.1679T>G, c.2846A>T), fluoropyrimidines should be avoided or reduced according to the patients’ homozygous or heterozygous status, respectively. For other common DPYD variants (c.496A>G, c.1129-5923C>G, c.1896T>C), conflicting data are reported and their use in clinical practice still needs to be validated. The high frequency of DPYD polymorphism and the lack of large prospective trials may explain differences in studies’ results. The epigenetic regulation of DPD expression has been recently investigated to explain the variable activity of the enzyme. DPYD promoter methylation and its regulation by microRNAs may affect the toxicity risk of fluoropyrimidines. The studies we reviewed indicate that pharmacogenetic testing is promising to direct personalised dosing of fluoropyrimidines, although further investigations are needed to establish the role of DPD in severe toxicity in patients treated for colorectal cancer.
Collapse
Affiliation(s)
- Felicia Stefania Falvella
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan 20157, Italy.
| | - Marta Caporale
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Stefania Cheli
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, University Hospital "Luigi Sacco", Università di Milano, Milan 20157, Italy.
| | - Antonia Martinetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Rosa Berenato
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Claudia Maggi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Monica Niger
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Francesca Ricchini
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Ilaria Bossi
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Maria Di Bartolomeo
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Elisa Sottotetti
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Francesca Futura Bernardi
- Department of Experimental Medicine, Section of Pharmacology "L. Donatelli", Faculty of Medicine and Surgery, Second University of Naples, Naples 80138, Italy.
| | - Filippo de Braud
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| | - Emilio Clementi
- Scientific Institute, IRCCS E. Medea, Bosisio Parini, Lecco 23842, Italy.
- Unit of Clinical Pharmacology, Department of Biomedical and Clinical Sciences, Consiglio Nazionale delle Ricerche Institute of Neuroscience, University Hospital "Luigi Sacco", Università di Milano, Milan 20157, Italy.
| | - Filippo Pietrantonio
- Medical Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian, Milan 20133, Italy.
| |
Collapse
|
75
|
Tourancheau A, Margaillan G, Rouleau M, Gilbert I, Villeneuve L, Lévesque E, Droit A, Guillemette C. Unravelling the transcriptomic landscape of the major phase II UDP-glucuronosyltransferase drug metabolizing pathway using targeted RNA sequencing. THE PHARMACOGENOMICS JOURNAL 2015; 16:60-70. [PMID: 25869014 DOI: 10.1038/tpj.2015.20] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/21/2014] [Accepted: 02/09/2015] [Indexed: 02/04/2023]
Abstract
A comprehensive view of the human UDP-glucuronosyltransferase (UGT) transcriptome is a prerequisite to the establishment of an individual's UGT metabolic glucuronidation signature. Here, we uncover the transcriptome landscape of the 10 human UGT gene loci in normal and tumoral metabolic tissues by targeted RNA next-generation sequencing. Alignment on the human hg19 reference genome identifies 234 novel exon-exon junctions. We recover all previously known UGT1 and UGT2 enzyme-coding transcripts and identify over 130 structurally and functionally diverse novel UGT variants. We further expose a revised genomic structure of UGT loci and provide a comprehensive repertoire of transcripts for each UGT gene. Data also uncover a remodelling of the UGT transcriptome occurring in a tissue- and tumor-specific manner. The complex alternative splicing program regulating UGT expression and protein functions is likely critical in determining detoxification capacity of an organ and stress-related responses, with significant impact on drug responses and diseases.
Collapse
Affiliation(s)
- A Tourancheau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - G Margaillan
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - M Rouleau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - I Gilbert
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - L Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada
| | - E Lévesque
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Medicine, Laval University, Québec, QC, Canada
| | - A Droit
- Faculty of Medicine, Laval University, Québec, QC, Canada
| | - C Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire (CHU) de Québec Research Center, Québec, QC, Canada.,Faculty of Pharmacy, Laval University, Québec, QC, Canada.,Canada Research Chair in Pharmacogenomics, Pharmacogenomics Laboratory, CHU de Quebec Research Center, Quebec, QC, Canada
| |
Collapse
|
76
|
Chen S, Laverdiere I, Tourancheau A, Jonker D, Couture F, Cecchin E, Villeneuve L, Harvey M, Court MH, Innocenti F, Toffoli G, Lévesque E, Guillemette C. A novel UGT1 marker associated with better tolerance against irinotecan-induced severe neutropenia in metastatic colorectal cancer patients. THE PHARMACOGENOMICS JOURNAL 2015; 15:513-20. [PMID: 25778466 DOI: 10.1038/tpj.2015.12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 12/03/2014] [Accepted: 01/28/2015] [Indexed: 12/13/2022]
Abstract
The risk of severe irinotecan-induced neutropenia has been shown to be related to the UGT1 variant UGT1A1*28, which increases exposure to the potent metabolite SN-38. Our goal was to identify a novel UGT1 marker(s) using 28 haplotype-tagged single nucleotide polymorphisms genotyped by mass spectrometry. By characterizing the UGT1 sequence from a cohort of 167 Canadian metastatic colorectal cancer (mCRC) patients and a validation cohort of 250 Italian mCRC patients, we found rs11563250G, located in the intergenic region downstream of UGT1, to be significantly associated with reduced risk of severe neutropenia (odds ratio (OR)=0.21; P=0.043 and OR=0.27; P=0.036, respectively, and OR=0.31 when combined; P=0.001), which remained significant upon correction for multiple testing in the combined cohort (P=0.041). For the two-marker haplotype rs11563250G and UGT1A1*1 (rs8175347 TA6), the OR was of 0.17 (P=0.0004). Genetic testing of this marker may identify patients who might benefit from increased irinotecan dosing.
Collapse
Affiliation(s)
- S Chen
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada
| | - I Laverdiere
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada
| | - A Tourancheau
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada
| | - D Jonker
- Division of Medical Oncology, Department of Medicine, Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - F Couture
- Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec, Québec, Canada
| | - E Cecchin
- Division of Experimental and Clinical Pharmacology, Department of Molecular Biology and Translational Research, National Cancer Institute and Cancer for Molecular Biomedicine, Aviano, Italy
| | - L Villeneuve
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada
| | - M Harvey
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada
| | - M H Court
- Individualized Medicine Program, Department of Veterinary Clinical Sciences, Washington State University College of Veterinary Medicine, Pullman, WA, USA
| | - F Innocenti
- Division of Pharmacotherapy & Experimental Therapeutics, Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC, USA
| | - G Toffoli
- Division of Experimental and Clinical Pharmacology, Department of Molecular Biology and Translational Research, National Cancer Institute and Cancer for Molecular Biomedicine, Aviano, Italy
| | - E Lévesque
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada.,Centre Hospitalier Universitaire de Québec Research Center and Faculty of Medicine, Laval University, Québec, Québec, Canada
| | - C Guillemette
- Pharmacogenomics Laboratory, Centre Hospitalier Universitaire de Québec Research Center and Faculty of Pharmacy, Laval University, Québec, Québec, Canada
| |
Collapse
|
77
|
Germline oncopharmacogenetics, a promising field in cancer therapy. Cell Oncol (Dordr) 2015; 38:65-89. [PMID: 25573079 DOI: 10.1007/s13402-014-0214-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/15/2014] [Indexed: 12/14/2022] Open
Abstract
Pharmacogenetics (PGx) is the study of the relationship between inter-individual genetic variation and drug responses. Germline variants of genes involved in drug metabolism, drug transport, and drug targets can affect individual response to medications. Cancer therapies are characterized by an intrinsically high toxicity; therefore, the application of pharmacogenetics to cancer patients is a particularly promising method for avoiding the use of inefficacious drugs and preventing the associated adverse effects. However, despite continuing efforts in this field, very few labels include information about germline genetic variants associated with drug responses. DPYD, TPMT, UGT1A1, G6PD, CYP2D6, and HLA are the sole loci for which the European Medicines Agency (EMA) and the US Food and Drug Administration (FDA) report specific information. This review highlights the germline PGx variants that have been approved to date for anticancer treatments, and also provides some insights about other germline variants with potential clinical applications. The continuous and rapid evolution of next-generation sequencing applications, together with the development of computational methods, should help to refine the implementation of personalized medicine. One day, clinicians may be able to prescribe the best treatment and the correct drug dosage based on each patient's genotype. This approach would improve treatment efficacy, reduce toxicity, and predict non-responders, thereby decreasing chemotherapy-associated morbidity and improving health benefits.
Collapse
|
78
|
Abul-Husn NS, Owusu Obeng A, Sanderson SC, Gottesman O, Scott SA. Implementation and utilization of genetic testing in personalized medicine. PHARMACOGENOMICS & PERSONALIZED MEDICINE 2014; 7:227-40. [PMID: 25206309 PMCID: PMC4157398 DOI: 10.2147/pgpm.s48887] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Clinical genetic testing began over 30 years ago with the availability of mutation detection for sickle cell disease diagnosis. Since then, the field has dramatically transformed to include gene sequencing, high-throughput targeted genotyping, prenatal mutation detection, preimplantation genetic diagnosis, population-based carrier screening, and now genome-wide analyses using microarrays and next-generation sequencing. Despite these significant advances in molecular technologies and testing capabilities, clinical genetics laboratories historically have been centered on mutation detection for Mendelian disorders. However, the ongoing identification of deoxyribonucleic acid (DNA) sequence variants associated with common diseases prompted the availability of testing for personal disease risk estimation, and created commercial opportunities for direct-to-consumer genetic testing companies that assay these variants. This germline genetic risk, in conjunction with other clinical, family, and demographic variables, are the key components of the personalized medicine paradigm, which aims to apply personal genomic and other relevant data into a patient’s clinical assessment to more precisely guide medical management. However, genetic testing for disease risk estimation is an ongoing topic of debate, largely due to inconsistencies in the results, concerns over clinical validity and utility, and the variable mode of delivery when returning genetic results to patients in the absence of traditional counseling. A related class of genetic testing with analogous issues of clinical utility and acceptance is pharmacogenetic testing, which interrogates sequence variants implicated in interindividual drug response variability. Although clinical pharmacogenetic testing has not previously been widely adopted, advances in rapid turnaround time genetic testing technology and the recent implementation of preemptive genotyping programs at selected medical centers suggest that personalized medicine through pharmacogenetics is now a reality. This review aims to summarize the current state of implementing genetic testing for personalized medicine, with an emphasis on clinical pharmacogenetic testing.
Collapse
Affiliation(s)
- Noura S Abul-Husn
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aniwaa Owusu Obeng
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA ; Department of Pharmacy, Mount Sinai Hospital, New York, NY, USA
| | - Saskia C Sanderson
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Omri Gottesman
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Stuart A Scott
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
79
|
Pharmacogenomics of human uridine diphospho-glucuronosyltransferases and clinical implications. Clin Pharmacol Ther 2014; 96:324-39. [PMID: 24922307 DOI: 10.1038/clpt.2014.126] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/07/2014] [Indexed: 12/12/2022]
Abstract
Glucuronidation by uridine diphospho-glucuronosyltransferase enzymes (UGTs) is a major phase II biotransformation pathway and, complementary to phase I metabolism and membrane transport, one of the most important cellular defense mechanisms responsible for the inactivation of therapeutic drugs, other xenobiotics, and endogenous molecules. Interindividual variability in UGT pathways is significant and may have profound pharmacological and toxicological implications. Several genetic and genomic processes underlie this variability and are discussed in relation to drug metabolism and diseases such as cancer.
Collapse
|