1
|
Bouhniz OE, Kenani A. Potential role of genetic polymorphisms in neoadjuvant chemotherapy response in breast cancer. J Chemother 2025; 37:97-111. [PMID: 38511398 DOI: 10.1080/1120009x.2024.2330241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 02/09/2024] [Accepted: 03/11/2024] [Indexed: 03/22/2024]
Abstract
Chemoresistance leads to treatment failure, which can arise through different mechanisms including patients' characteristics. Searching for genetic profiles as a predictor for drug response and toxicity has been extensively studied in pharmacogenomics, thus contributing to personalized medicine and providing alternative treatments. Numerous studies have demonstrated significant evidence of association between genetic polymorphisms and response to neoadjuvant chemotherapy (NAC) in breast cancer. In this review, we explored the potential impact of genetic polymorphisms in NAC primary resistance through selecting a specific clinical profile. The genetic variability within pharmacokinetics, pharmacodynamics, DNA synthesis and repair, and oncogenic signaling pathways genes could be predictive or prognostic markers for NAC resistance. The clinical implication of these results can help provide individualized treatment plans in the early stages of breast cancer treatment. Further studies are needed to determine the genetic hosts of primary chemoresistance mechanisms in order to further emphasize the implementation of genotypic approaches in personalized medicine.
Collapse
Affiliation(s)
- Om Elez Bouhniz
- Research Laboratory "Environment, Inflammation, Signaling and Pathologies" (LR18ES40), Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| | - Abderraouf Kenani
- Research Laboratory "Environment, Inflammation, Signaling and Pathologies" (LR18ES40), Faculty of Medicine of Monastir, University of Monastir, Monastir, Tunisia
| |
Collapse
|
2
|
van der Perk MEM, Broer L, Yasui Y, Robison LL, Hudson MM, Laven JSE, van der Pal HJ, Tissing WJE, Versluys B, Bresters D, Kaspers GJL, de Vries ACH, Lambalk CB, Overbeek A, Loonen JJ, Beerendonk CCM, Byrne J, Berger C, Clemens E, Dirksen U, Falck Winther J, Fosså SD, Grabow D, Muraca M, Kaiser M, Kepák T, Kruseova J, Modan-Moses D, Spix C, Zolk O, Kaatsch P, Krijthe JH, Kremer LCM, Brooke RJ, Baedke JL, van Schaik RHN, van den Anker JN, Uitterlinden AG, Bos AME, van Leeuwen FE, van Dulmen-den Broeder E, van der Kooi ALLF, van den Heuvel-Eibrink MM, on behalf of the PanCareLIFE Consortium. Effect of Genetic Variation in CYP450 on Gonadal Impairment in a European Cohort of Female Childhood Cancer Survivors, Based on a Candidate Gene Approach: Results from the PanCareLIFE Study. Cancers (Basel) 2021; 13:4598. [PMID: 34572825 PMCID: PMC8470074 DOI: 10.3390/cancers13184598] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Female childhood cancer survivors (CCSs) carry a risk of therapy-related gonadal dysfunction. Alkylating agents (AA) are well-established risk factors, yet inter-individual variability in ovarian function is observed. Polymorphisms in CYP450 enzymes may explain this variability in AA-induced ovarian damage. We aimed to evaluate associations between previously identified genetic polymorphisms in CYP450 enzymes and AA-related ovarian function among adult CCSs. METHODS Anti-Müllerian hormone (AMH) levels served as a proxy for ovarian function in a discovery cohort of adult female CCSs, from the pan-European PanCareLIFE cohort (n = 743; age (years): median 25.8, interquartile range (IQR) 22.1-30.6). Using two additive genetic models in linear and logistic regression, nine genetic variants in three CYP450 enzymes were analyzed in relation to cyclophosphamide equivalent dose (CED) score and their impact on AMH levels. The main model evaluated the effect of the variant on AMH and the interaction model evaluated the modifying effect of the variant on the impact of CED score on log-transformed AMH levels. Results were validated, and meta-analysis performed, using the USA-based St. Jude Lifetime Cohort (n = 391; age (years): median 31.3, IQR 26.6-37.4). RESULTS CYP3A4*3 was significantly associated with AMH levels in the discovery and replication cohort. Meta-analysis revealed a significant main deleterious effect (Beta (95% CI): -0.706 (-1.11--0.298), p-value = 7 × 10-4) of CYP3A4*3 (rs4986910) on log-transformed AMH levels. CYP2B6*2 (rs8192709) showed a significant protective interaction effect (Beta (95% CI): 0.527 (0.126-0.928), p-value = 0.01) on log-transformed AMH levels in CCSs receiving more than 8000 mg/m2 CED. CONCLUSIONS Female CCSs CYP3A4*3 carriers had significantly lower AMH levels, and CYP2B6*2 may have a protective effect on AMH levels. Identification of risk-contributing variants may improve individualized counselling regarding the treatment-related risk of infertility and fertility preservation options.
Collapse
Affiliation(s)
- M. E. Madeleine van der Perk
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Linda Broer
- Department of Internal Medicine, Rotterdam, ErasmusMC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (L.B.); (A.G.U.)
| | - Yutaka Yasui
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Y.); (L.L.R.); (M.M.H.); (R.J.B.); (J.L.B.)
| | - Leslie L. Robison
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Y.); (L.L.R.); (M.M.H.); (R.J.B.); (J.L.B.)
| | - Melissa M. Hudson
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Y.); (L.L.R.); (M.M.H.); (R.J.B.); (J.L.B.)
- Department of Oncology, Division of Survivorship, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Joop S. E. Laven
- Department of Obstetrics and Gynecology, Erasmus MC–University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Helena J. van der Pal
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Wim J. E. Tissing
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Birgitta Versluys
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Dorine Bresters
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Gertjan J. L. Kaspers
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
- Department of Pediatric Oncology-Haematology, Emma Children’s Hospital, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Andrica C. H. de Vries
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Cornelis B. Lambalk
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (C.B.L.); (A.O.)
| | - Annelies Overbeek
- Department of Obstetrics and Gynaecology, Amsterdam UMC, Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (C.B.L.); (A.O.)
| | - Jacqueline J. Loonen
- Department of Haematology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands;
| | - Catharina C. M. Beerendonk
- Department of Obstetrics and Gynaecology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands;
| | - Julianne Byrne
- Boyne Research Institute, 5 Bolton Square, East, Drogheda, A92 RY6K Co. Louth, Ireland;
| | - Claire Berger
- Department of Paediatric Oncology, University Hospital, 42 055 Saint-Etienne, France;
- Lyon University, Jean Monnet University, INSERM, U 1059, Sainbiose, 42023 Saint-Etienne, France
| | - Eva Clemens
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Uta Dirksen
- University Hospital Essen, Pediatrics III, West German Cancer Centre, 45147 Essen, Germany;
- German Cancer Research Centre, DKTK, Site Essen, 45147 Essen, Germany
| | - Jeanette Falck Winther
- Childhood Cancer Research Group, Danish Cancer Society Research Center, 2100 Copenhagen, Denmark;
- Department of Clinical Medicine, Faculty of Health, Aarhus University and University Hospital, 8200 Aarhus, Denmark
| | - Sophie D. Fosså
- Department of Oncology, Oslo University Hospital, 0372 Oslo, Norway;
| | - Desiree Grabow
- Division of Childhood Cancer Epidemiology, German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.G.); (M.K.); (C.S.); (P.K.)
| | - Monica Muraca
- Epidemiology and Biostatistics Unit and DOPO Clinic, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Melanie Kaiser
- Division of Childhood Cancer Epidemiology, German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.G.); (M.K.); (C.S.); (P.K.)
| | - Tomáš Kepák
- University Hospital Brno, International Clinical Research Center (FNUSA-ICRC), Masaryk University, 656 91 Brno, Czech Republic;
| | | | - Dalit Modan-Moses
- The Edmond and Lily Safra Children’s Hospital, Chaim Sheba Medical Center, Tel Hashomer, and the Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel;
| | - Claudia Spix
- Division of Childhood Cancer Epidemiology, German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.G.); (M.K.); (C.S.); (P.K.)
| | - Oliver Zolk
- Institute of Clinical Pharmacology, Brandenburg Medical School Theodor Fontane, Immanuel Klinik Rüdersdorf, 16816 Neuruppin, Germany;
| | - Peter Kaatsch
- Division of Childhood Cancer Epidemiology, German Childhood Cancer Registry, Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany; (D.G.); (M.K.); (C.S.); (P.K.)
| | - Jesse H. Krijthe
- Department of Intelligent Systems, Delft University of Technology, 2628 BL Delft, The Netherlands;
| | - Leontien C. M. Kremer
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Russell J. Brooke
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Y.); (L.L.R.); (M.M.H.); (R.J.B.); (J.L.B.)
| | - Jessica L. Baedke
- Department of Epidemiology and Cancer Control, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA; (Y.Y.); (L.L.R.); (M.M.H.); (R.J.B.); (J.L.B.)
| | - Ron H. N. van Schaik
- Department of clinical chemistry, Erasmus MC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands;
| | - John N. van den Anker
- Division of Clinical Pharmacology, Children’s National Hospital, Washington, DC 20010, USA;
| | - André G. Uitterlinden
- Department of Internal Medicine, Rotterdam, ErasmusMC University Medical Center Rotterdam, 3015 GD Rotterdam, The Netherlands; (L.B.); (A.G.U.)
| | - Annelies M. E. Bos
- Department of Reproductive Medicine and Gynecology, University Medical Center Utrecht, 3584 CS Utrecht, The Netherlands;
| | - Flora E. van Leeuwen
- Department of Epidemiology, Netherlands Cancer Institute, 1066 CX Amsterdam, The Netherlands;
| | - Eline van Dulmen-den Broeder
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | - Anne-Lotte L. F. van der Kooi
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
- Department of Obstetrics and Gynecology, Erasmus MC–University Medical Center, 3015 GD Rotterdam, The Netherlands;
| | - Marry M. van den Heuvel-Eibrink
- Princess Máxima Center for Pediatric Oncology, 3584 CS Utrecht, The Netherlands; (H.J.v.d.P.); (W.J.E.T.); (B.V.); (D.B.); (G.J.L.K.); (A.C.H.d.V.); (E.C.); (L.C.M.K.); (E.v.D.-d.B.); (A.-L.L.F.v.d.K.); (M.M.v.d.H.-E.)
| | | |
Collapse
|
3
|
Wang K, Lin Z, Zhang H, Zhang X, Zheng X, Zhao L, Yang Q, Ahima J, Boateng NAS. Investigating proteome and transcriptome response of Cryptococcus podzolicus Y3 to citrinin and the mechanisms involved in its degradation. Food Chem 2019; 283:345-352. [PMID: 30722882 DOI: 10.1016/j.foodchem.2019.01.052] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 12/16/2018] [Accepted: 01/13/2019] [Indexed: 11/28/2022]
Abstract
Citrinin (CIT) contamination has been reported in agricultural foods and is known to be nephrotoxic to human and animals. In the present study, the proteomes and transcriptomes of C. podzolicus Y3 treated with or without 10 μg/mL CIT were compared by two-dimensional electrophoresis (2-DE) and RNA sequencing, respectively. The proteomics results showed that there were 23 differentially expressed proteins (DEPs), 8 DEPs were up-regulated and 15 DEPs were significantly down-regulated. Transcriptomic analysis showed that 1208 genes were differentially expressed, 551 (43.05%) DEGs were up regulated and 657 (56.95%) were down-regulated. These results showed that the CIT treatment caused DNA damage, oxidative stress and cell apoptosis in C. podzolicus Y3. CIT treatment also activated the defense response (DNA repair and drug resistance biological process, antioxidative activity and TCA cycle) as well as drug metabolism (synthesize the CIT-degrading enzymes) in yeast cells to respond to CIT stress and degrade CIT.
Collapse
Affiliation(s)
- Kaili Wang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Zhen Lin
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Hongyin Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China.
| | - Xiaoyun Zhang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Xiangfeng Zheng
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Lina Zhao
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Qiya Yang
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Joseph Ahima
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| | - Nana Adwoa Serwah Boateng
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, Jiangsu, People's Republic of China
| |
Collapse
|
4
|
Ghadban T, Schmidt-Yang M, Uzunoglu FG, Perez DR, El Gammal AT, Miro JT, Wellner U, Pantel K, Izbicki JR, Vashist YK. Evaluation of the germline single nucleotide polymorphism rs583522 in the TNFAIP3 gene as a prognostic marker in esophageal cancer. Cancer Genet 2015; 208:595-601. [PMID: 26598072 DOI: 10.1016/j.cancergen.2015.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2015] [Revised: 09/21/2015] [Accepted: 09/22/2015] [Indexed: 12/20/2022]
Abstract
Most esophageal cancer patients die because of disease relapse, hence an accurate prognosis of disease relapse and survival is essential. Genetic variations in cancer patients may serve as important indicators. Three genotypes (GG, AG, and AA) are displayed by the single nucleotide polymorphism (SNP) rs583522, which maps to the TNFAIP3 gene on chromosome 6. Evaluation of the potential prognostic value of the TNFAIP3-SNP in esophageal cancer (EC) was the aim of this study. A total of 158 patients underwent complete surgical resection of the esophagus for EC. None of them received any neoadjuvant or adjuvant treatment. Peripheral blood was sampled, and genomic DNA was extracted from leukocytes before each operation. Clinicopathologic parameters, tumor cell dissemination in bone marrow, and clinical outcome were correlated with the TNFAIP3-SNP. A-allele carriers showed advanced tumor stages compared with those of homozygous G-allele carriers (P<0.001). Patients with an A-allele genotype (AA or AG) were significantly more likely to experience a relapse (P=0.003). Survival analysis (log-rank test) revealed a significant difference in overall survival between the three groups (P=0.039); however, none of the genotypes was identified as a disease stage-independent prognostic marker. In conclusion, TNFAIP3-SNP stratifies patients into different risk groups; however, it could not be identified as an independent prognostic marker.
Collapse
Affiliation(s)
- Tarik Ghadban
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Magdalena Schmidt-Yang
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Faik G Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniel R Perez
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander T El Gammal
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jameel T Miro
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulrich Wellner
- Clinic for Surgery, University Clinic of Schleswig-Holstein, Campus Luebeck, Luebeck, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob R Izbicki
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yogesh K Vashist
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
5
|
An A/C germline single-nucleotide polymorphism in the TNFAIP3 gene is associated with advanced disease stage and survival in only surgically treated esophageal cancer. J Hum Genet 2014; 59:661-6. [PMID: 25354935 DOI: 10.1038/jhg.2014.90] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 08/31/2014] [Accepted: 09/11/2014] [Indexed: 12/20/2022]
Abstract
Prognostication of disease relapse and survival is essential for cancer patients and genetic variations in cancer patients may serve as important indicators. A single-nucleotide polymorphism (SNP) mapping to the tumor necrosis factor, alpha-induced protein 3 (TNFAIP3) gene at position 138241110 displays three genotypes (AA, AC and CC). The aim of this study was to evaluate the potential prognostic value of the TNFAIP3-SNP in esophageal cancer (EC). Genomic DNA was extracted from peripheral blood leukocytes of 173 patients who underwent complete surgical resection for EC and did not receive any neoadjuvant or adjuvant therapy. For SNP detection, a 260- bp fragment was PCR amplified, purified and sequenced with tested primers. The product was analyzed by automatic DNA sequencer.The TNFAIP3 genotypes were correlated with clinico-pathological parameters, tumor cell dissemination in bone marrow and clinical outcome. The C-allele carrier presented with higher disease stage (P<0.001). This was predominantly because of the presence of lymph node metastasis (P<0.001). The recurrence rate was higher in C-allele carriers (AC and CC genotype; P=0.004). Kaplan-Meier plots for disease-free (P=0.017) and overall survival (P<0.001) displayed a gene dosage-associated outcome with AA genotype patients presenting the longest and CC genotype patients the poorest survival. In disease stage-adjusted multivariate analysis the TNFAIP3-SNP was identified as an independent prognostic factor for survival (hazard ratio 1.9; P=0.008). The TNFAIP3-SNP allows risk stratification of EC patients and may be a useful tool to identify patients eligible for multimodal therapy concepts.
Collapse
|
6
|
Kang HW, Song PH, Ha YS, Kim WT, Kim YJ, Yun SJ, Lee SC, Choi YH, Moon SK, Kim WJ. Glutathione S-transferase M1 and T1 polymorphisms: susceptibility and outcomes in muscle invasive bladder cancer patients. Eur J Cancer 2013; 49:3010-9. [PMID: 23810248 DOI: 10.1016/j.ejca.2013.05.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 04/08/2013] [Accepted: 05/23/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND We investigated whether genetic polymorphisms in the glutathione S transferase mu (GSTM1) and theta (GSTT1) genes modulated risk, disease progression and survival in primary muscle invasive bladder cancer (MIBC). METHODS GSTM1 and GSTT1 polymorphisms were analysed by multiplex polymerase chain reaction (PCR) using blood genomic DNA in 110 MIBC patients and 220 gender- and age-matched healthy controls. The influence of the genetic polymorphisms on patient survival was evaluated by Kaplan-Meier survival curves and Cox Proportional Hazard models. We also evaluated whether cigarette smoking and treatment modality modified the association between genotype and prognosis. RESULTS GSTM1-null individuals exhibited increased risk for MIBC and an association with cigarette smoking. GSTT1-null subjects showed significant disease progression and cancer-specific death. In the combined analysis, GSTT1-null genotype was an independent risk factor for disease progression and cancer specific death regardless of GSTM1 genotype. Significant differences in progression-free survival (PFS) and cancer-specific survival (CSS) were seen based on GSTT1 genotype. The survival impact of the GSTT1 genotype was only valid for smokers. The GSTT1-null genotype was an independent prognostic factor for shorter PFS in patients who received chemotherapy and those who did not undergo radical cystectomy. By multivariate Cox regression analysis, GSTT1-null genotype was a predictive factor for disease progression and cancer specific survival regardless of treatment modality. CONCLUSIONS The GSTM1-null genotype plays an important role in genetic susceptibility to MIBC and the GSTT1-null genotype is associated with disease progression and shorter survival in MIBC.
Collapse
Affiliation(s)
- Ho Won Kang
- Department of Urology, College of Medicine, Chungbuk National University, Cheongju, South Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Uzunoglu FG, Heumann A, Musici S, Kutup A, Koenig A, Roch N, Thomssen A, Dohrmann T, Tsui TY, Mann O, Izbicki JR, Vashist YK. The T393C polymorphism of GNAS1 is a predictor for relapse and survival in resectable non-small cell lung cancer. Lung Cancer 2012. [PMID: 23201296 DOI: 10.1016/j.lungcan.2012.11.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
INTRODUCTION The GNAS1 T393C single nucleotide polymorphism (T393C-SNP) correlates with Gαs mRNA stability and protein expression and augmented apoptosis. Genetic germ line variations as stable and reproducible markers potentially serve as prognostic marker in oncology. The aim of this study was to evaluate the potential prognostic value of T393C-SNP in complete resected non-small cell lung cancer (NSCLC). PATIENTS AND METHODS In total 163 Caucasian patients, who had been surgically treated for NSCLC between 1998 and 2010, were included in this study. Genotyping of peripheral blood cells was performed by polymerase chain reaction and digestion using the restriction enzyme FokI. The T393C-SNP was correlated with clinic-pathological parameters and survival. Chi-square test, Kaplan-Meier estimator and cox regression hazard model were used to assess the prognostic value of the T393C-SNP. RESULTS C-allele carriers had a higher recurrence rate (p=0.018) and a shorter disease-free survival compared to homozygous T-allele carriers (12.26 months vs. 44.65 months, p=0.009). The overall survival in homozygous C allele carriers was shorter (19.10 months vs. 53.95 months, p=0.019). Multivariate Cox regression identified the CC genotype as a negative independent prognostic factor for recurrence (hazard ratio 2.36, p=0.007) and survival (hazard ratio 2.51, p=0.008). CONCLUSION Determination of T393C-SNP preoperatively potentially allows allocation of NSCLC patients into different risk profiles and may influence the therapeutic strategy.
Collapse
Affiliation(s)
- Faik Güntac Uzunoglu
- Department of General, Visceral and Thoracic Surgery, University Medical Centre of Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
8
|
Association of cytochrome P450 genetic polymorphisms with neoadjuvant chemotherapy efficacy in breast cancer patients. BMC MEDICAL GENETICS 2012; 13:45. [PMID: 22702493 PMCID: PMC3458973 DOI: 10.1186/1471-2350-13-45] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 05/22/2012] [Indexed: 01/29/2023]
Abstract
Background The enzymes of the cytochrome P450 family (CYPs) play an important role in the metabolism of a great variety of anticancer agents; therefore, polymorphisms in genes encoding for metabolizing enzymes and drugs transporters can affect drug efficacy and toxicity. Methods The genetic polymorphisms of cytochrome P450 were studied in 395 patients with breast cancer by RLFP analysis. Results Here, we studied the association of functionally significant variant alleles of CYP3A4, CYP3A5, CYP2B6, CYP2C8, CYP2C9 and CYP2C19 with the clinical response to neoadjuvant chemotherapy in breast cancer patients. A significant correlation was observed between the CYP2C9*2 polymorphism and chemotherapy resistance (OR = 4.64; CI 95% = 1.01 – 20.91), as well as between CYP2C9*2 heterozygotes and chemotherapy resistance in women with nodal forms of breast cancer and a cancer hereditary load (OR = 15.50; CI 95% = 1.08 – 826.12) when the potential combined effects were examined. No significant association between chemotherapy resistance and the other examined genotypes and the potential combined clinical and tumour-related parameters were discovered. Conclusion In conclusion, CYP2C9*2 was associated with neoadjuvant chemotherapy resistance (OR = 4.64; CI 95% = 1.01 – 20.91) in the population of interest.
Collapse
|
9
|
Engebraaten O, Edvardsen H, Løkkevik E, Naume B, Kristensen V, Ottestad L, Natarajan V. Gefitinib in Combination with Weekly Docetaxel in Patients with Metastatic Breast Cancer Caused Unexpected Toxicity: Results from a Randomized Phase II Clinical Trial. ISRN ONCOLOGY 2012; 2012:176789. [PMID: 22666610 PMCID: PMC3361199 DOI: 10.5402/2012/176789] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 02/29/2012] [Indexed: 11/30/2022]
Abstract
In patients with metastatic breast cancer, taxane treatment demonstrates activity but is not curative. Targeted treatment modalities are therefore necessary in order to improve outcomes in this group. A randomized placebo-controlled phase II trial was initiated to evaluate effect and toxicity of gefitinib (250 mg QD) and docetaxel 35 mg/m2 (six of seven weeks) (NCT 00319618). The inclusion of 66 patients was planned. The study was closed due to treatment-related toxicity. Of the 18 included patients, seven (of which three received gefitinib) were withdrawn from the study due to toxicity. Of the nine patients receiving gefitinib and chemotherapy, one achieved a partial response and four stable disease. In the chemotherapy of nine patients, four had a partial response and four stable disease. The breast cancer patients in this study were genotyped using a panel of 14 single-nucleotide polymorphisms (SNPs), previously found associated with docetaxel clearance in a cohort of lung cancer patients. We were unable to identify genes related to toxicity in this study. Nevertheless, toxicity was aggravated by the addition of the tyrosine kinase inhibitor. In conclusion, despite adequately tolerated as monotherapy, combination regimens should be carefully considered for overlapping adverse events in order to avoid increased treatment-related toxicity.
Collapse
Affiliation(s)
- Olav Engebraaten
- Department of Oncology, Oslo University Hospital, 0424 Oslo, Norway
| | | | | | | | | | | | | |
Collapse
|
10
|
Abstract
Recent studies have suggested an increase in the number of retracted scientific publications. It is unclear how broadly the issue of misleading and fraudulent publications pertains to retractions of drug therapy studies. Therefore, we sought to determine the trends and factors associated with retracted publications in drug therapy literature. A PubMed search was conducted to identify retracted drug therapy articles published from 2000-2011. Articles were grouped according to reason for retraction, which was classified as scientific misconduct or error. Scientific misconduct was further divided into data fabrication, data falsification, questions of data veracity, unethical author conduct, and plagiarism. Error was defined as duplicate publication, scientific mistake, journal error, or unstated reasons. Additional data were extracted from the retracted articles, including type of article, funding source, author information, therapeutic area, and retraction issue. A total of 742 retractions were identified from 2000-2011 in the general biomedical literature, and 102 drug studies met our inclusion criteria. Of these, 73 articles (72%) were retracted for a reason classified as scientific misconduct, whereas 29 articles (28%) were retracted for error. Among the 73 articles classified as scientific misconduct, those classified as unethical author conduct (32 articles [44%]) and data fabrication (24 articles [33%]) constituted the majority. The median time from publication of the original article to retraction was 31 months (range 1-130). Fifty percent of retracted articles did not state a funding source, whereas pharmaceutical manufacturer funding accounted for only 13 articles (13%) analyzed. Many retractions were due to repeat offenses by a small number of authors, with nearly 40% of the retracted studies associated with two individuals. We found that a greater proportion of drug therapy articles were retracted for reasons of misconduct and fraud compared with other biomedical studies. It is important for health care practitioners to monitor the literature for retractions so that recommendations for drug therapy and patient management may be modified accordingly.
Collapse
Affiliation(s)
- Jennifer C Samp
- Center for Pharmacoeconomic Research, Department of Pharmacy Practice, College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
11
|
Cytostatic drugs in infants: A review on pharmacokinetic data in infants. Cancer Treat Rev 2012; 38:3-26. [DOI: 10.1016/j.ctrv.2011.03.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/21/2011] [Accepted: 03/24/2011] [Indexed: 01/11/2023]
|
12
|
Yu KD, Huang AJ, Fan L, Li WF, Shao ZM. Genetic Variants in Oxidative Stress–Related Genes Predict Chemoresistance in Primary Breast Cancer: A Prospective Observational Study and Validation. Cancer Res 2011; 72:408-19. [PMID: 22147260 DOI: 10.1158/0008-5472.can-11-2998] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ke-Da Yu
- Department of Breast Surgery, Cancer Center and Cancer Institute, Shanghai Medical College, Fudan University, Shanghai, People's Republic of China
| | | | | | | | | |
Collapse
|
13
|
Li W, Yue W, Yang X, Zhang C, Wang Y. [Relationship between the genetic polymorphisms of phase I and II drug-metabolizing enzymes, as well as the outcome of chemotherapy in advanced non-small cell lung cancer]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2011; 14:858-64. [PMID: 22104220 PMCID: PMC5999999 DOI: 10.3779/j.issn.1009-3419.2011.11.05] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
背景与目的 目前药物代谢酶遗传多态性与化疗疗效关系的研究结果多不一致,本研究旨在探讨细胞色素P4501A1(cytochrome P450 1A1, CYP1A1)、2E1(cytochrome P450 2E1, CYP2E1)、2D6(cytochrome P450 2D6, CYP2D6)和谷胱甘肽硫转移酶M1(glutathione S-transferase M1, GSTM1)基因多态性与晚期非小细胞肺癌化疗疗效以及与肺癌患者预后的关系。 方法 采用PCR和PCR-RFLP技术对肺癌患者4种药物代谢酶基因分型,并对他们进行5年跟踪随访。 结果 携带B型CYP1A1和缺陷性GSTM1肺癌患者比其它基因型患者化疗疗效好(P < 0.001)。携带A型CYP1A1肺癌患者接受非铂类化疗药物治疗比B型和C型患者疗效好(P=0.041); 携带缺陷性GSTM1肺癌患者接受铂类化疗药物治疗疗效比功能型患者疗效好(P=0.011)。4种酶对晚期非小细胞肺癌患者总生存期(overall survival, OS)没有明显影响(P > 0.05)。 结论 A型CYP1A1肺癌患者接受非铂类化疗药物治疗比B型和C型患者疗效好; 缺陷性GSTM1肺癌患者接受铂类化疗药物治疗比功能型患者疗效好。4种酶基因多态对晚期非小细胞肺癌患者OS影响没有明显统计学差异。
Collapse
Affiliation(s)
- Weiying Li
- Department of Cell Molecular Biology, Beijing Thoracic Tumour and Tuberculosis Research Institute, Beijing 101149, China.
| | | | | | | | | |
Collapse
|
14
|
Polymorphisms in GSTM1, CYP1A1, CYP2E1, and CYP2D6 are associated with susceptibility and chemotherapy response in non-small-cell lung cancer patients. Lung 2011; 190:91-8. [PMID: 22109568 DOI: 10.1007/s00408-011-9338-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 09/29/2011] [Indexed: 10/15/2022]
Abstract
BACKGROUND Studies of polymorphisms in CYP1A1, CYP2E1, CYP2D6, and GSTM1 and their relationship to lung cancer susceptibility and chemotherapy response have been reported, but the results are not consistent. In this study we selected four polymorphisms in these genes, several of which have previously been researched, and investigated their association with lung cancer susceptibility and chemotherapy response. METHODS We genotyped the four polymorphisms in a cohort composed of 217 non-small-cell lung cancer (NSCLC) patients and 198 controls. Of these, 145 advanced NSCLC patients underwent chemotherapy and were monitored for 5 years. RESULTS Significant differences in the GSTM1 polymorphism were observed between the case and control groups (P = 0.02). We observed a synergistic effect of smoking and GSTM1. Smokers with deficient-type GSTM1 had a 4.96-fold increased risk of developing lung cancer. Significant differences in GSTM1 and CYP1A1 polymorphisms were observed between the response and nonresponse groups (P = 0.004 and P = 0.026). Moreover, patients with deficient-type GSTM1 were superior responders to platinum drugs than those carrying wild-type GSTM1 (P = 0.014). In addition, patients carrying TT CYP1A1 responded better to nonplatinum drugs than those carrying TC and CC CYP1A1 (P = 0.01). Polymorphisms in the four enzymes had no effect on the overall survival of NSCLC patients. CONCLUSIONS Our findings support the hypothesis that a polymorphism in GSTM1 is associated with lung cancer susceptibility. Furthermore, polymorphisms in GSTM1 and CYP1A1 were associated with chemotherapy response. In particular, smokers carrying deficient-type GSTM1 were at a higher risk of developing lung cancer. Patients carrying deficient-type GSTM1 responded better to platinum drugs, while those with TT CYP1A1 were better responders to nonplatinum drugs.
Collapse
|
15
|
Vashist YK, Uzungolu G, Kutup A, Gebauer F, Koenig A, Deutsch L, Zehler O, Busch P, Kalinin V, Izbicki JR, Yekebas EF. Heme oxygenase-1 germ line GTn promoter polymorphism is an independent prognosticator of tumor recurrence and survival in pancreatic cancer. J Surg Oncol 2011; 104:305-11. [PMID: 21495030 DOI: 10.1002/jso.21926] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2010] [Accepted: 03/11/2011] [Indexed: 02/06/2023]
Abstract
BACKGROUND Heme oxygenase-1 (HO-1) correlates with aggressive tumor behavior and chemotherapy resistance in pancreatic cancer (PC). We evaluated the prognostic value of the basal transcription controlling germ line GTn repeat polymorphism (GTn) in the promoter region of the HO-1 gene in PC. PATIENTS AND METHODS We determined the GTn in 100 controls and 150 PC patients. DNA was extracted from blood leukocytes and GTn determined by PCR, electrophoresis, and sequencing. Clinicopathological parameters, disease-free, and overall survival (DFS, OS) were correlated with GTn. RESULTS Three genotypes were defined based on short (S) <25 and long (L) ≥25 GTn repeat alleles. In PC patients, a steadily increasing risk was evident between LL, SL, and SS genotype patients for larger tumor size, presence of lymph node metastasis, poor tumor differentiation and higher recurrence rate (P < 0.001 each). The SS genotype displayed the most aggressive tumor biology. The LL genotype had the best and the SS genotype the worst DFS and OS (P < 0.001 each). The GTn genotype was the strongest prognostic factor for recurrence and survival (P < 0.001 each). CONCLUSION The GTn repeat polymorphism is a strong prognostic marker for recurrence and survival in PC patients.
Collapse
Affiliation(s)
- Yogesh K Vashist
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Heme Oxygenase-1 Promoter Polymorphism is a Predictor of Disease Relapse in Pancreatic Neuroendocrine Tumors. J Surg Res 2011; 166:e121-7. [DOI: 10.1016/j.jss.2010.11.898] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 10/28/2010] [Accepted: 11/16/2010] [Indexed: 11/23/2022]
|
17
|
Vashist YK, Kutup A, Musici S, Yekebas EF, Mina S, Uzunoglu G, Zehler O, Koenig A, Cataldegirmen G, Bockhorn M, Effenberger K, Kalinin V, Pantel K, Izbicki JR. The GNAS1 T393C single nucleotide polymorphism predicts the natural postoperative course of complete resected esophageal cancer. Cell Oncol (Dordr) 2011; 34:281-8. [PMID: 21340746 DOI: 10.1007/s13402-011-0016-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2011] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Genetic variations in cancer patients may serve as important prognostic indicators of clinical outcome. The GNAS1 T393C single nucleotide polymorphism (SNP) diversely correlates with the clinical outcome in cancer. The aim of this study was to evaluate the potential prognostic value of T393C-SNP in complete resected only surgically treated esophageal cancer (EC). METHODS Genomic DNA was extracted from peripheral blood leucocytes of 190 patients who underwent only complete surgical resection for EC. T393C-SNP was correlated with clinic-pathological parameters, tumor cell dissemination in bone marrow (DTC) and clinical outcome. RESULTS T-allele carriers had more advanced disease due to presence of lymph node metastasis (P < 0.0001) and DTC (P = 0.01) and higher recurrence rate (P = 0.01) compared to CC genotype. The disease-free (P < 0.001) and overall survival (P < 0.001) was better in CC compared to TT and TC patients. In the multivariate Cox regression disease-stage adjusted analysis the T393C-SNP was identified as a strong independent prognostic factor for recurrence (hazard ratio 1.8, P = 0.01) and survival (hazard ratio 2.5, P < 0.001) in EC patients. CONCLUSION Determination of T393C-SNP preoperatively will allow allocation of EC patients into different risk profiles which may help to stratify patients eligible for neoadjuvant and or adjuvant therapy.
Collapse
Affiliation(s)
- Yogesh Kumar Vashist
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Fernandes BJD, Silva CDM, Andrade JM, Matthes ADCS, Coelho EB, Lanchote VL. Pharmacokinetics of cyclophosphamide enantiomers in patients with breast cancer. Cancer Chemother Pharmacol 2011; 68:897-904. [PMID: 21290248 DOI: 10.1007/s00280-011-1554-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Accepted: 01/10/2011] [Indexed: 01/17/2023]
Abstract
PURPOSE Adjuvant chemotherapy with cyclophosphamide (CYC) is used for the treatment of breast cancer. CYC is used as a racemic mixture, although preclinical data have demonstrated differences in the efficacy and toxicity of its enantiomers, with (S)-(-)-CYC exhibiting a higher therapeutic index. The present study investigated the enantioselectivity and influence of CYP2B6, CYP2C9, CYP2C19, and CYP3A on the kinetic disposition of CYC in patients with breast cancer. METHODS Fifteen patients previously submitted to removal of the tumor and treated with racemic CYC (900 or 1,000 mg/m(2)) and epirubicin were included in the study. The in vivo activity of CYP3A was evaluated using midazolam as a marker drug. Serial blood samples were collected up to 24 h after administration of the first cycle of CYC. RESULTS The kinetic disposition of CYC was enantioselective in patients with breast cancer, with plasma accumulation of the (S)-(-)-CYC enantiomer (AUC 195.0 vs. 174.8 μg h/mL) due to the preferential clearance of the (R)-(+)CYC enantiomer (5.1 vs. 5.7 L/h). Clearance of either CYC enantiomer did not differ between the CYP2B6, CYP2C9, and CYP2C19 genotypes or as a function of the in vivo activity of CYP3A evaluated by midazolam clearance. CONCLUSIONS The pharmacokinetics of CYC is enantioselective in patients with breast cancer concomitantly treated with epirubicin and ondansetron. Genotyping or phenotyping did not contribute to adjustment of the CYC dose regimen in patients included in this study.
Collapse
Affiliation(s)
- Bruno José Dumêt Fernandes
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto da Universidade de São Paulo, Avenida do Café s/n, Campus da USP, Ribeirão Preto, SP, 14040-903, Brazil
| | | | | | | | | | | |
Collapse
|
19
|
Valachis A, Mauri D, Neophytou C, Polyzos NP, Tsali L, Garras A, Papanikolau EG. Translational medicine and reliability of single-nucleotide polymorphism studies: can we believe in SNP reports or not? Int J Med Sci 2011; 8:492-500. [PMID: 21897762 PMCID: PMC3167098 DOI: 10.7150/ijms.8.492] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Accepted: 08/09/2011] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The number of genetic association studies is increasing exponentially. Nonetheless, genetic association reports are prone to potential biases which may influence the reported outcome. AIM We hypothesized that positive outcome for a determined polymorphism might be over-reported across genetic association studies analysing a small number of polymorphisms, when compared to studies analysing the same polymorphism together with a high number of other polymorphisms. METHODS We systematically reviewed published reports on the association of glutathione s-transferase (GST) single-nucleotide polymorphisms (SNPs) and cancer outcome. RESULT We identified 79 eligible trials. Most of the studies examined the GSTM1, theGSTP1 Ile105Val mutation, and GSTT1polymorphisms (n = 54, 57 and 46, respectively). Studies analysing one to three polymorphisms (n = 39) were significantly more likely to present positive outcomes, compared to studies examining more than 3 polymorphisms (n=40) p = 0.004; this was particularly evident for studies analysing the GSTM1polymorphism (p =0.001). We found no significant associations between journal impact factor, number of citations, and probability of publishing positive studies or studies with 1-3 polymorphisms examined. CONCLUSIONS We propose a new subtype of publication bias in genetic association studies. Positive results for genetic association studies analysing a small number of polymorphisms (n = 1-3) should be evaluated extremely cautiously, because a very large number of such studies are inconclusive and statistically under-powered. Indeed, publication of misleading reports may affect harmfully medical decision-making and use of resources, both in clinical and pharmacological development setting.
Collapse
|
20
|
Kilburn L, Okcu MF, Wang T, Cao Y, Renfro-Spelman A, Aldape KD, Gilbert MR, Bondy M. Glutathione S-transferase polymorphisms are associated with survival in anaplastic glioma patients. Cancer 2010; 116:2242-9. [PMID: 20187096 PMCID: PMC2861043 DOI: 10.1002/cncr.25006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Glutathione S-transferases (GSTs) are polymorphic enzymes that are responsible for glutathione conjugation of alkylators and scavenging of free radicals created by radiation. GST polymorphisms may result in altered or absent enzyme activity and have been associated with survival in patients with cancer. The authors of this report hypothesized that patients with anaplastic glioma (AG) who have GST genotypes that encode for lower activity enzymes will have longer survival than similar patients who have higher activity genotypes. The current study was performed to investigate the role of GST enzyme polymorphisms in predicting the survival of patients with AG. METHODS The medical records of 207 patients with AG from a single cancer center were reviewed retrospectively. Polymorphisms for the GST micro1 (GSTM1), GST theta1 (GSTT1), and GST pi1 (GSTP1) enzymes were identified. Overall survival was compared using the Kaplan-Meier method and Cox proportional hazards analyses adjusting for age, sex, histology, and therapy. RESULTS Among the patients with oligodendroglial tumors (n = 94), patients who had the GSTT1 null genotype had a 2.9 times increased risk of death (95% confidence interval [CI], 1.3-6.3) compared with patients who had the GSTT1 non-null genotype. Adjustment for 1p/19q status did not change the finding. In the patients who had anaplastic astrocytoma (n = 113), the patients with all GSTP1 genotypes except GSTP1 *B/*B had a 3.8 times increased risk of death (95% CI, 0.5-29.6) compared with patients who had the GSTP1 *B/*B genotype. CONCLUSIONS In patients with anaplastic oligodendroglial tumors, the GSTT1 null genotype may be associated with poor survival, possibly because of modifications in therapy secondary to increased toxicity. This hypothesis is under investigation. In patients with anaplastic astrocytoma, the GSTP1 *B/*B genotype may confer a survival advantage.
Collapse
Affiliation(s)
- Lindsay Kilburn
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
- Texas Children’s Cancer Center, Baylor College of Medicine, 6621 Fannin Street, CC1410.00, Houston, TX 77030
| | - M. Fatih Okcu
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
- Texas Children’s Cancer Center, Baylor College of Medicine, 6621 Fannin Street, CC1410.00, Houston, TX 77030
| | - Tao Wang
- Division of Biostatistics, Dan L. Duncan Cancer Center, Baylor College of Medicine, One Baylor Plaza, BCMM600, Houston, TX 77030
| | - Yumei Cao
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
| | - Amy Renfro-Spelman
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
| | - Kenneth D. Aldape
- Department of Pathology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Box 85, Houston, TX 77030
| | - Mark R. Gilbert
- Department of Neuro-oncology, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd., Box 431, Houston, TX 77030
| | - Melissa Bondy
- Department of Pediatrics, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030
- Department of Epidemiology, Division of Cancer Prevention, The University of Texas M. D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, TX 77030
- Childhood Cancer Prevention and Epidemiology Center, 6621 Fannin Street, CC1510.00, Houston, TX 77030
- Texas Children’s Cancer Center, Baylor College of Medicine, 6621 Fannin Street, CC1410.00, Houston, TX 77030
| |
Collapse
|
21
|
Anderson BO, Yip CH, Smith RA, Shyyan R, Sener SF, Eniu A, Carlson RW, Azavedo E, Harford J. Guideline implementation for breast healthcare in low-income and middle-income countries: overview of the Breast Health Global Initiative Global Summit 2007. Cancer 2009; 113:2221-43. [PMID: 18816619 DOI: 10.1002/cncr.23844] [Citation(s) in RCA: 326] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Breast cancer outcomes in low- and middle-income countries (LMCs) correlate with the degree to which 1) cancers are detected at early stages, 2) newly detected cancers can be diagnosed correctly, and 3) appropriately selected multimodality treatment can be provided properly in a timely fashion. The Breast Health Global Initiative (BHGI) invited international experts to review and revise previously developed BHGI resource-stratified guideline tables for early detection, diagnosis, treatment, and healthcare systems. Focus groups addressed specific issues in breast pathology, radiation therapy, and management of locally advanced disease. Process metrics were developed based on the priorities established in the guideline stratification. The groups indicated that cancer prevention through health behavior modification could influence breast cancer incidence in LMCs. Diagnosing breast cancer at earlier stages will reduce breast cancer mortality. Programs to promote breast self-awareness and clinical breast examination and resource-adapted mammographic screening are important early detection steps. Breast imaging, initially with ultrasound and, at higher resource levels with diagnostic mammography, improves preoperative diagnostic assessment and permits image-guided needle sampling. Multimodality therapy includes surgery, radiation, and systemic therapies. Government intervention is needed to address drug-delivery problems relating to high cost and poor access. Guideline dissemination and implementation research plays a crucial role in improving care. Adaptation of technology is needed in LMCs, especially for breast imaging, pathology, radiation therapy, and systemic treatment. Curricula for education and training in LMCs should be developed, applied, and studied in LMC-based learning laboratories to aid information transfer of evidence-based BHGI guidelines.
Collapse
Affiliation(s)
- Benjamin O Anderson
- Department of Surgery, University of Washington, Seattle, Washington 98195, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Barahmani N, Carpentieri S, Li XN, Wang T, Cao Y, Howe L, Kilburn L, Chintagumpala M, Lau C, Okcu MF. Glutathione S-transferase M1 and T1 polymorphisms may predict adverse effects after therapy in children with medulloblastoma. Neuro Oncol 2008; 11:292-300. [PMID: 18952980 DOI: 10.1215/15228517-2008-089] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Glutathione S-transferases (GSTs) are polymorphic enzymes that catalyze the glutathione conjugation of alkylating agents, platinum compounds, and free radicals formed by radiation used to treat medulloblastoma. We hypothesized that GST polymorphisms may be responsible, in part, for individual differences in toxicity and responses in pediatric medulloblastoma. We investigated the relationship between GSTM1 and GSTT1 polymorphisms and survival and toxicity in 42 children with medulloblastoma diagnosed and treated at the Texas Children's Cancer Center. We conducted Kaplan-Meier analyses to determine if the GST polymorphisms were related to progression-free survival (PFS) and performed logistic regression to explore associations between GST polymorphisms and occurrence of grade 3 or greater (> or =Gr 3) myelosuppression, ototoxicity, nephrotoxicity, neurotoxicity, and intellectual impairment. Patients with at least one null genotype had a 4.3 (95% confidence interval, 1.1-16.8), 3.7 (1-13.6), and 6.4 (1.2-34) times increased risk for any > or =Gr 3 toxicity, any > or =Gr 3 toxicity excluding peripheral neuropathy, and any > or =Gr 3 toxicity requiring omission or cessation of chemotherapy, respectively. Compared with all others, patients with at least one null genotype had, on average, 27.2 (p x= 0.0002), 29 (p = 0.0004), and 21.7 (p = 0.002) lower full-scale, performance, and verbal intelligence quotient (IQ) scores, respectively. GSTM1 and GSTT1 polymorphisms may predict adverse events, including cognitive impairment after therapy, in patients with medulloblastoma. A larger study to validate these findings is under way.
Collapse
Affiliation(s)
- Nadia Barahmani
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
In contrast to western high-income nations, the incidence and mortality from breast cancer are increasing in most low and middle-income countries worldwide. Current approaches to breast cancer control developed for populations of high-income societies should not be directly transferred without evaluation. A relevant research agenda includes population differences in tumor biology and metabolization of systemic therapies, cultural and psychosocial issues, and operations in healthcare systems. Highest priority should be given to assessments of clinical downstaging and basic systemic treatment effectiveness in low and middle-income populations. Partnerships of existing organizations in high-income nations with those in low and middle-income countries are currently the most feasible sources of research support.
Collapse
Affiliation(s)
- Richard R Love
- Department of Internal Medicine, Division of Hematology/Oncology, Ohio State University Comprehensive Cancer Center, Columbus, Ohio 43210, USA.
| |
Collapse
|
24
|
Glutathione-S-transferase P1, T1 and M1 genetic polymorphisms in neoadjuvant-treated locally advanced gastric cancer: GSTM1-present genotype is associated with better prognosis in completely resected patients. Int J Colorectal Dis 2008; 23:773-82. [PMID: 18443805 DOI: 10.1007/s00384-008-0490-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/04/2008] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Neoadjuvant chemotherapy in gastric cancer is now standard in the Western world; however, only 30-40% of the patients respond to induction therapy. Pretherapeutic predictors of response and prognosis would be of utmost interest to individualize treatment. Glutathione-S-transferase enzymes detoxify therapeutic drugs such as platin derivates and may influence outcome of the treated patients. Therefore, glutathione-S-transferase (GST) polymorphisms were assessed as predictive markers in cisplatinum-based neoadjuvant-treated gastric cancer. MATERIALS AND METHODS DNA was isolated from 139 patients with locally advanced gastric cancer (cT3/4 anyN cM0) before chemotherapy. Multiplex polymerase chain reaction was used for GSTT1 and GSTM1 genes, and allelic discrimination assay with the TaqMan system for the GSTP1 gene. RESULTS One hundred ten patients could be analyzed for GSTT1 (T-:23; T+87), 112 for GSTM1 (M-:52; M+:60) and 132 for GSTP1 (Ile/Ile: 55; Ile/Val: 59; Val/Val: 18). There was no significant correlation between any of the GSTT1, GSTM1, or GSTP1 genotypes and patients' characteristics or histopathological data; only the GSTM1+ genotype was associated with the non-intestinal subtype of the Lauren classification (p=0.045). GSTT1, GSTM1, and GSTP1 genotypes were not correlated with response to chemotherapy (p=0.57, p=0.38, p=0.33). In R0 resected patients, we found an improved survival for patients with the GSTM1-present genotype compared to patients with the GSTM1-null genotype (p=0.017). Moreover, the GSTM1-present genotype showed a significantly better tumor-related (p=0.017) and disease-free survival (p=0.029). CONCLUSION None of the common GST polymorphisms predicts response in our study, but the GSTM1+ genotype was associated with a better prognosis in completely resected patients. Further investigations on chemotherapy-associated gene polymorphisms are warranted.
Collapse
|
25
|
Rochat B, Zoete V, Grosdidier A, von Grünigen S, Marull M, Michielin O. In vitro biotransformation of imatinib by the tumor expressed CYP1A1 and CYP1B1. Biopharm Drug Dispos 2008; 29:103-18. [DOI: 10.1002/bdd.598] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
26
|
Influence of germline polymorphisms of GSTT1, GSTM1, and GSTP1 in familial versus sporadic breast cancer susceptibility and survival. Fam Cancer 2007; 7:213-20. [DOI: 10.1007/s10689-007-9177-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Accepted: 12/02/2007] [Indexed: 10/22/2022]
|
27
|
DeMichele A, Gimotty P, Botbyl J, Aplenc R, Colligon T, Foulkes AS, Rebbeck TR. In Response to “Drug Metabolizing Enzyme Polymorphisms Predict Clinical Outcome in a Node-Positive Breast Cancer Cohort”. J Clin Oncol 2007; 25:5675-7. [DOI: 10.1200/jco.2006.10.1485] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
- Angela DeMichele
- Department of Biostatistics and Epidemiology, University of Pennsylvania; Hematology/Oncology Division, Department of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Phyllis Gimotty
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, PA
| | | | - Richard Aplenc
- Division of Hematology/Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Theresa Colligon
- Abramson Family Research Institute, University of Pennsylvania, Philadelphia, PA
| | - Andrea S. Foulkes
- Department of Biostatistics, University of Massachusetts, Amherst, MA
| | - Timothy R. Rebbeck
- Department of Biostatistics and Epidemiology, University of Pennsylvania; Abramson Family Research Institute, University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
28
|
TGFβ1 (Leu10Pro), p53 (Arg72Pro) can predict for increased risk for breast cancer in south Indian women and TGFβ1 Pro (Leu10Pro) allele predicts response to neo-adjuvant chemo-radiotherapy. Breast Cancer Res Treat 2007; 112:81-7. [DOI: 10.1007/s10549-007-9821-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2007] [Accepted: 11/13/2007] [Indexed: 12/15/2022]
|
29
|
Ingelman-Sundberg M, Sim SC, Gomez A, Rodriguez-Antona C. Influence of cytochrome P450 polymorphisms on drug therapies: pharmacogenetic, pharmacoepigenetic and clinical aspects. Pharmacol Ther 2007; 116:496-526. [PMID: 18001838 DOI: 10.1016/j.pharmthera.2007.09.004] [Citation(s) in RCA: 784] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2007] [Accepted: 09/20/2007] [Indexed: 01/11/2023]
Abstract
The polymorphic nature of the cytochrome P450 (CYP) genes affects individual drug response and adverse reactions to a great extent. This variation includes copy number variants (CNV), missense mutations, insertions and deletions, and mutations affecting gene expression and activity of mainly CYP2A6, CYP2B6, CYP2C9, CYP2C19 and CYP2D6, which have been extensively studied and well characterized. CYP1A2 and CYP3A4 expression varies significantly, and the cause has been suggested to be mainly of genetic origin but the exact molecular basis remains unknown. We present a review of the major polymorphic CYP alleles and conclude that this variability is of greatest importance for treatment with several antidepressants, antipsychotics, antiulcer drugs, anti-HIV drugs, anticoagulants, antidiabetics and the anticancer drug tamoxifen. We also present tables illustrating the relative importance of specific common CYP alleles for the extent of enzyme functionality. The field of pharmacoepigenetics has just opened, and we present recent examples wherein gene methylation influences the expression of CYP. In addition microRNA (miRNA) regulation of P450 has been described. Furthermore, this review updates the field with respect to regulatory initiatives and experience of predictive pharmacogenetic investigations in the clinics. It is concluded that the pharmacogenetic knowledge regarding CYP polymorphism now developed to a stage where it can be implemented in drug development and in clinical routine for specific drug treatments, thereby improving the drug response and reducing costs for drug treatment.
Collapse
Affiliation(s)
- Magnus Ingelman-Sundberg
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, SE-17177, Stockholm, Sweden.
| | | | | | | |
Collapse
|
30
|
Genetic variability in CYP3A4 and CYP3A5 in primary liver, gastric and colorectal cancer patients. BMC Cancer 2007; 7:118. [PMID: 17605821 PMCID: PMC1931602 DOI: 10.1186/1471-2407-7-118] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Accepted: 07/02/2007] [Indexed: 01/01/2023] Open
Abstract
Background Drug-metabolizing enzymes play a role in chemical carcinogenesis through enzymatic activation of procarcinogens to biologically reactive metabolites. The role of gene polymorphisms of several cytochrome P450 enzymes in digestive cancer risk has been extensively investigated. However, the drug-metabolizing enzymes with the broader substrate specificity, CYP3A4 and CYP3A5, have not been analyzed so far. This study aims to examine associations between common CYP3A4 and CYP3A5 polymorphisms and digestive cancer risk. Methods CYP3A4 and CYP3A5 genotypes were determined in 574 individuals including 178 patients with primary liver cancer, 82 patients with gastric cancer, 151 patients with colorectal cancer, and 163 healthy individuals. Results The variant allele frequencies for patients with liver cancer, gastric cancer, colorectal cancer and healthy controls, respectively, were: CYP3A4*1B, 4.8 % (95% C.I. 2.6–7.0), 3.7 % (0.8–6.6) 4.3% (2.0–6.6) and 4.3% (2.1–6.5); CYP3A5*3, 91.8 % (93.0–97.4), 95.7% (92.6–98.8), 91.7% (88.6–94.8) and 90.8% (87.7–93.9). The association between CYP3A4*1B and CYP3A5*3 variant alleles did not significantly differ among patients and controls. No differences in genotypes, allele frequencies, or association between variant alleles were observed with regard to gender, age at diagnosis, tumour site or stage. Conclusion Common polymorphisms on CYP3A4 and CYP3A5 genes do not modify the risk of developing digestive cancers in Western Europe.
Collapse
|
31
|
Nakajima M, Komagata S, Fujiki Y, Kanada Y, Ebi H, Itoh K, Mukai H, Yokoi T, Minami H. Genetic polymorphisms of CYP2B6 affect the pharmacokinetics/pharmacodynamics of cyclophosphamide in Japanese cancer patients. Pharmacogenet Genomics 2007; 17:431-45. [PMID: 17502835 DOI: 10.1097/fpc.0b013e328045c4fb] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE To evaluate the effects of genetic polymorphisms of drug metabolizing enzymes on the pharmacokinetics of cyclophosphamide and its active metabolite, 4-hydroxycyclophosphamide, and on the pharmacodynamics. EXPERIMENTAL DESIGN One hundred and three Japanese patients with malignant lymphoma or breast cancer treated with cyclophosphamide (500-750 mg/m) participated in this study. The plasma concentrations of cyclophosphamide and 4-hydroxycyclophosphamide were determined by high-performance liquid chromatography, and pharmacokinetic parameters were calculated. The genotypes of CYP2B6, CYP2C19, CYP3A4, CYP3A5, ALDH1A1, GST genes were determined by allele-specific polymerase chain reaction or polymerase chain reaction-restriction-fragment length polymorphism. RESULTS A large interindividual difference (54-fold) was observed in the area under the curve ratio of 4-hydroxycyclophosphamide/cyclophosphamide calculated as the metabolic index. We first proved that leukocytopenia and neutropenia were significantly (P<0.01) related to the area under the curve of 4-hydroxycyclophosphamide. We found that the homozygotes of CYP2B6*6 (Q172H and K262R) showed significantly (P<0.05) higher clearance and shorter half-life of cyclophosphamide than heterozygotes and homozygotes of CYP2B6*1. The small sample size, however, limited the impact. On the other hand, it was clearly demonstrated that the patients possessing the single nucleotide polymorphisms of the CYP2B6 gene, g.-2320T>C, g.-750T>C (5'-flanking region), g.15582C>T (intron 3), or g.18492T>C (intron 5), had significantly lower area under the curve ratios of 4-hydroxycyclophosphamide/cyclophosphamide, indicating a decreased cyclophosphamide 4-hydroxylation. Of particular importance was the finding that leukocytopenia was significantly related to the single nucleotide polymorphisms g.-2320T>C, g.-750T>C, and g.18492T>C in CYP2B6 gene, which are highly linked. No relationship was observed between the pharmacokinetics of cyclophosphamide or 4-hydroxycyclophosphamide and genetic polymorphisms of the other enzymes. CONCLUSIONS We clarified that the single nucleotide polymorphisms in the promoter region or introns in the CYP2B6 affect the potency of cyclophosphamide activation to 4-hydroxycyclophosphamide. This information would be valuable for predicting adverse reactions and the clinical efficacy of cyclophosphamide.
Collapse
Affiliation(s)
- Miki Nakajima
- Drug Metabolism and Toxicology, Division of Pharmaceutical Sciences, Graduate School of Medical Science, Kanazawa University, Kakuma-machi, Kanazawa 920-1192, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Boddy AV. Recent developments in the clinical pharmacology of classical cytotoxic chemotherapy. Br J Clin Pharmacol 2007; 62:27-34. [PMID: 16842376 PMCID: PMC1885069 DOI: 10.1111/j.1365-2125.2006.02714.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Advances in analytical methods, imaging techniques and an increased understanding of the influence of pharmacogenetic factors have added to our knowledge of the pharmacology of many chemotherapeutic agents. Extending the use of these approaches to pharmacodynamic end-points, together with the application of population-based modelling techniques, offers the potential to develop truly individualized therapy in the future.
Collapse
Affiliation(s)
- Alan V Boddy
- Northern Institute for Cancer Research, Medical School, University of Newcastle, Newcastle upon Tyne, UK.
| |
Collapse
|
33
|
Deeken JF, Figg WD, Bates SE, Sparreboom A. Toward individualized treatment: prediction of anticancer drug disposition and toxicity with pharmacogenetics. Anticancer Drugs 2007; 18:111-26. [PMID: 17159598 DOI: 10.1097/cad.0b013e3280109411] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
A great deal of effort has been spent in defining the pharmacokinetics and pharmacodynamics of investigational and registered anticancer agents. Often, there is a marked variability in drug handling between individual patients, which contributes to variability in the pharmacodynamic effects of a given dose of a drug. A combination of physiological variables, genetic characteristics (pharmacogenetics) and environmental factors is known to alter the relationship between the absolute dose and the concentration-time profile in plasma. A variety of strategies are now being evaluated in patients with cancer to improve the therapeutic index of anticancer drugs by implementation of pharmacogenetic imprinting through genotyping or phenotyping individual patients. The efforts have mainly focused on variants in genes encoding the drug-metabolizing enzymes thiopurine S-methyltransferase, dihydropyrimidine dehydrogenase, members of the cytochrome P450 family, including the CYP2B, 2C, 2D and 3A subfamilies, members of the UDP glucuronosyltransferase family, as well as the ATP-binding cassette transporters ABCB1 (P-glycoprotein) and ABCG2 (breast cancer resistance protein). Several of these genotyping strategies have been shown to have substantial impact on therapeutic outcome and should eventually lead to improved anticancer chemotherapy.
Collapse
Affiliation(s)
- John F Deeken
- Medical Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD 20895, USA
| | | | | | | |
Collapse
|
34
|
Choi JY, Nowell SA, Blanco JG, Ambrosone CB. The role of genetic variability in drug metabolism pathways in breast cancer prognosis. Pharmacogenomics 2006; 7:613-24. [PMID: 16753008 DOI: 10.2217/14622416.7.4.613] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Among patients receiving adjuvant therapy for breast cancer, there is variability in treatment outcomes, and it is unclear which patients will receive the most benefit from treatment and which will have better disease-free survival. To date, most studies of breast cancer prognosis have focused on tumor characteristics, but it is likely that pharmacogenetics, genetic variability in the metabolism of therapeutic agents, also plays a role in the prediction of survival. In this paper, we briefly discuss the metabolic pathways of drugs commonly used for the treatment of breast cancer (cyclophosphamide, doxorubicin, taxanes, tamoxifen and aromatase inhibitors) and describe the known genetic variants that may impact those pathways. Studies that have evaluated potential effects of these genetic variants on treatment outcomes are also discussed. It is likely that the application of pharmacogenetics, particularly in the setting of randomized clinical trials, will contribute to findings that may result in individualized therapeutic dosing.
Collapse
Affiliation(s)
- Ji-Yeob Choi
- Roswell Park Cancer Institute, Department of Epidemiology, Elm & Carlton Sts, Buffalo, NY, 14263, USA
| | | | | | | |
Collapse
|
35
|
Ruzzo A, Graziano F, Kawakami K, Watanabe G, Santini D, Catalano V, Bisonni R, Canestrari E, Ficarelli R, Menichetti ET, Mari D, Testa E, Silva R, Vincenzi B, Giordani P, Cascinu S, Giustini L, Tonini G, Magnani M. Pharmacogenetic profiling and clinical outcome of patients with advanced gastric cancer treated with palliative chemotherapy. J Clin Oncol 2006; 24:1883-91. [PMID: 16622263 DOI: 10.1200/jco.2005.04.8322] [Citation(s) in RCA: 112] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
PURPOSE To investigate whether polymorphisms with putative influence on fluorouracil/cisplatin activity are associated with clinical outcomes of patients with advanced gastric cancer (AGC). PATIENTS AND METHODS Peripheral blood samples from 175 prospectively enrolled AGC patients treated with fluorouracil/cisplatin palliative chemotherapy were used for genotyping 13 polymorphisms in nine genes (TS, MTHFR, XPD, ERCC1, XRCC1, XRCC3, GSTPI, GSTTI, GSTMI). Genotypes were correlated to response and survival. RESULTS The overall response rate was 41%, the median progression-free survival (PFS) was 24 weeks (range, 4 to 50 weeks), and the median overall survival (OS) was 39 weeks (range, 8 to 72+ weeks). Chemoresistance and poor survival were significantly associated with TS 5'-UTR 3G-genotype (2R/3G, 3C/3G, 3G/3G) and GSTP1 105 A/A homozygous genotype. Sixty-one patients (35%) did not show any of these risk genotypes (group 0), 57 patients (32.5%) showed one of the two risk genotypes (group 1), and 57 patients (32.5%) showed both risk genotypes (group 2). Median PFS and OS in group 0 patients were 32 weeks (range, 8 to 50 weeks) and 49 weeks (range, 18 to 72+ weeks), respectively. Group 1 and group 2 patients showed significantly worse PFS (median, 26 weeks [range, 6 to 44 weeks] and 14 weeks [range, 4 to 38 weeks], respectively) and worse OS (median, 39 weeks [range, 10 to 58 weeks] and 28 weeks [range, 8 to 56 weeks]), respectively, than group 0 patients. This adverse effect was retained in multivariate analysis. CONCLUSION Specific polymorphisms may influence clinical outcomes of AGC patients. Selecting palliative chemotherapy on the basis of pretreatment genotyping may represent an innovative strategy that warrants prospective studies.
Collapse
|
36
|
Abstract
The cytochromes P450 (CYPs) are key enzymes in cancer formation and cancer treatment. They mediate the metabolic activation of numerous precarcinogens and participate in the inactivation and activation of anticancer drugs. Since all CYPs that metabolize xenobiotics are polymorphic, much emphasis has been put on the investigation of a relationship between the distribution of specific variant CYP alleles and risk for different types of cancer, but a consistent view does not yet exist. This is to a great extent explained by the fact that the CYPs involved in activation of precarcinogens are in general not functionally polymorphic. This is in contrast to CYPs that are active in drug biotransformation where large interindividual differences in the capacity to metabolize therapeutic drugs are seen as a consequence of polymorphic alleles with altered function. This includes also some anticancer drugs like tamoxifen and cyclophosphamide metabolized by CYP2D6, CYP2C19 and CYP2B6. Some P450 forms are also selectively expressed in tumours, and this could provide a mechanism for drug resistance, but also future therapies using these enzymes as drug targets can be envisioned. This review gives an up-to-date description of our current knowledge in these areas.
Collapse
Affiliation(s)
- C Rodriguez-Antona
- Endocrine Cancer Group, Human Cancer Genetics Programme, Spanish National Cancer Center (CNIO), Madrid, Spain
| | | |
Collapse
|
37
|
Abstract
Due to their enormous substrate spectrum CYP3A4, -3A5 and -3A7 constitute the most important drug-metabolising enzyme subfamily in humans. CYP3As are expressed predominantly, but not exclusively, in the liver and intestine, where they participate in the metabolism of 45 - 60% of currently used drugs and many other compounds such as steroids and carcinogens. CYP3A expression and activity vary interindividually due to a combination of genetic and nongenetic factors such as hormone and health status, and the impact of environmental stimuli. Over the past several years, genetic determinants have been identified for much of the variable expression of CYP3A5 and -3A7, but not for CYP3A4. Using these markers, an effect of CYP3A5 expression status has been demonstrated beyond doubt for therapies with the immunosuppressive drug tacrolimus. Further associations are likely to emerge for drugs metabolised predominantly by CYP3A5 or -3A7, especially for individuals or tissues with concomitant low expression of CYP3A4. However, as exemplified by the controversial association between CYP3A4*1B and prostate cancer, the detection of clinical effects of CYP3A gene variants will be difficult. The most important underlying problems are the continuing absence of activity markers specific for CYP3A4 and the strong contribution of nongenetic factors to CYP3A variability.
Collapse
Affiliation(s)
- Leszek Wojnowski
- Johannes Gutenberg University, Department of Pharmacology, Obere Zahlbacher Strasse 67,55131 Mainz, Germany.
| | | |
Collapse
|