1
|
Chen M, Gong N, Sun W, Han J, Liu Y, Zhang S, Zheng A, Butt HJ, Liang XJ, Wu S. Red-Light-Responsive Metallopolymer Nanocarriers with Conjugated and Encapsulated Drugs for Phototherapy Against Multidrug-Resistant Tumors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2201672. [PMID: 35665442 DOI: 10.1002/smll.202201672] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/08/2022] [Indexed: 06/15/2023]
Abstract
It is challenging to treat multidrug-resistant tumors because such tumors are resistant to a broad spectrum of structurally and functionally unrelated drugs. Herein, treatment of multidrug-resistant tumors using red-light-responsive metallopolymer nanocarriers that are conjugated with the anticancer drug chlorambucil (CHL) and encapsulated with the anticancer drug doxorubicin (DOX) is reported. An amphiphilic metallopolymer PolyRuCHL that contains a poly(ethylene glycol) (PEG) block and a red-light-responsive ruthenium (Ru)-containing block is synthesized. Chlorambucil is covalently conjugated to the Ru moieties of PolyRuCHL. Encapsulation of DOX into PolyRuCHL in an aqueous solution results in DOX@PolyRuCHL micelles. The DOX@PolyRuCHL micelles are efficiently taken up by the multidrug-resistant breast cancer cell line MCF-7R and which carries DOX into the cells. Free DOX, without the nanocarriers, is not taken up by MCF-7R or pumped out of MCF-7R via P-glycoproteins. Red light irradiation of DOX@PolyRuCHL micelles triggers the release of chlorambucil-conjugated Ru moieties and DOX. Both act synergistically to inhibit the growth of multidrug-resistant cancer cells. Furthermore, the inhibition of the growth of multidrug-resistant tumors in a mouse model using DOX@PolyRuCHL micelles is demonstrated. The design of red-light-responsive metallopolymer nanocarriers with both conjugated and encapsulated drugs opens up an avenue for photoactivated chemotherapy against multidrug-resistant tumors.
Collapse
Affiliation(s)
- Mingjia Chen
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Ningqiang Gong
- CAS Center for Excellence in Nanoscience and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Wen Sun
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, 2 Linggong Road, Dalian, 116024, China
| | - Jianxiong Han
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Yuanli Liu
- College of Materials Science and Engineering, Guilin University of Technology, Guilin, 541004, China
| | - Shouwen Zhang
- Neurophysiology Department, Beijing ChaoYang Emergency Medical Center, Beijing, 100122, China
| | - Aiping Zheng
- Institute of Pharmacology and Toxicology of Academy of Military Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, China
| | - Hans-Jürgen Butt
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
| | - Xing-Jie Liang
- CAS Center for Excellence in Nanoscience and CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing, 100190, China
| | - Si Wu
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128, Mainz, Germany
- CAS Key Laboratory of Soft Matter Chemistry, Anhui Key Laboratory of Optoelectronic Science and Technology, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, China
| |
Collapse
|
2
|
The circadian clock gene Bmal1 facilitates cisplatin-induced renal injury and hepatization. Cell Death Dis 2020; 11:446. [PMID: 32522976 PMCID: PMC7287064 DOI: 10.1038/s41419-020-2655-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
Cisplatin is one of the most potent chemotherapy drugs to treat cancers, but its clinical application remains limited due to severe nephrotoxicity. Several approaches have been developed to minimize such side effects, notably including chronotherapy, a well-known strategy based on the circadian clock. However, the component of the circadian clock machinery that particularly responses to the cisplatin stimulation remains unknown, including its functions in cisplatin-induced renal injury. In our present study, we demonstrated that Bmal1, as a key clock gene, was induced by the cisplatin stimulation in the mouse kidney and cultured human HK-2 renal cells. Gain- and loss-of-function studies indicated that Bmal1 facilitated cisplatin-induced renal injury both in vivo and in vitro, by aggravating the cell apoptotic process. More importantly, RNA-seq analysis revealed that Bmal1 triggered the expression of hallmark genes involved in renal hepatization, a critical event accompanied by the injury. At the molecular level, Bmal1 activated the transcription of hepatization-associated genes through direct recruitment to the E-box motifs of their promoters. Our findings suggest that Bmal1, a pivotal mediator induced renal injury in response to cisplatin treatment, and the therapeutic intervention targeting Bmal1 in the kidney may be a promising strategy to minimize the toxic side-effects of cisplatin in its clinical applications.
Collapse
|
3
|
Rumiato E, Boldrin E, Malacrida S, Battaglia G, Sileni VC, Ruol A, Amadori A, Saggioro D. Identification of host variants associated with overall survival of esophageal cancer patients receiving platinum-based therapy. Pharmacogenomics 2020; 21:393-402. [PMID: 32285752 DOI: 10.2217/pgs-2019-0165] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Clinical features of esophageal cancer (EC) patients have poor prognostic power. Thus, it is paramount to discover biomarkers that can allow a more accurate survival prediction. Methods: To detect genetic variants associated with survival, DNA from 120 patients treated with cisplatin-based neoadjuvant therapy were genotyped using drug metabolism enzymes and transporters array. Results: We identified two variants: the rs2038067 in PPARD (p = 0.0004) and the rs683369 (F160L) in SLC22A1 (p = 0.001). Their prognostic power was greater than that of clinical stage alone (p = 0.017) and comparable to that of response to neoadjuvant therapy (p = 0.71). Interestingly, the prognostic accuracy of response models increased significantly when genetic variables were included (p = 0.003). Conclusion: Our data, though preliminary, strengthen the potential utility of germline variants for a better-tailored management of EC patients.
Collapse
Affiliation(s)
- Enrica Rumiato
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Elisa Boldrin
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Sandro Malacrida
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Giorgio Battaglia
- Endoscopy Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | - Alberto Ruol
- Department of Surgical Sciences, Oncology & Gastroenterology, University of Padova, Padova, Italy
| | - Alberto Amadori
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.,Department of Surgical Sciences, Oncology & Gastroenterology, University of Padova, Padova, Italy
| | - Daniela Saggioro
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
4
|
Abyarghamsari M, Hosseini Shirazi F, Tavakoli-Ardakani M, Rezvani H, Mirzaei HR, Salamzadeh J. Study of the Relationship between ERCC1 Polymorphisms and Response to Platinum-based Chemotherapy in Iranian Patients with Colorectal and Gastric Cancers. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2020; 18:2163-2171. [PMID: 32184881 PMCID: PMC7059040 DOI: 10.22037/ijpr.2019.1100827] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
This study was designed to evaluate the effect of excision repair cross complementing group 1 (ERCC1) rs11615 codon 118C/T gene polymorphisms on treatment outcomes in Iranian patients receiving oxaliplatin-based regimens for colorectal (CRC) and gastric cancers (GC). Patients, who were candidates to receive oxaliplatin-based chemotherapy, entered into the study. In 2-week intervals, the patients received combination regimen of oxaliplatin, fluorouracil, and leucovorin (FOLFOX) for 3 months. ERCC1 rs11615 codon 118C/T polymorphism was tested by restriction fragment length polymorphism polymerase chain reaction (RFLP-PCR) method using patients’ peripheral blood lymphocytes. The tumor response to chemotherapy was evaluated by examining the size of the tumor using CT scan. Association between response rates, according to the RECIST criteria, and patients’ genotypes was evaluated. Any relationship between response rate and possible explanatory factors was also determined. Overall, 40 patients (13 females (32.5%), and 27 males (67.5%)) enrolled in the study. Four patients (10.0%) carried the homozygous mutation (T/T genotype), ten patients (25.0%) were heterozygous (C/T genotype), and twenty-six patients (65%) were homozygous (C/C genotype). Response rate were 30.77%, 20.00%, and 0.00% for the genotypes C/C, C/T, and T/T, respectively. No significant association between response rate and genotypes was observed (p = 0.64). Patients with well- and moderately-differentiated histological grade of the tumor showed a better response rate (100.00% of 2 patients and 66.66% of 12 patients, respectively) compared to those with poorly differentiated (0.00% of 26 patients) histological grade (p < 0.001). Further multicenter studies are recommended to confirm conclusively our findings.
Collapse
Affiliation(s)
- Mahdiye Abyarghamsari
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medial Sciences, Tehran, Iran
| | - Farshad Hosseini Shirazi
- Pharmaceutical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maria Tavakoli-Ardakani
- Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medial Sciences, Tehran, Iran
| | - Hamid Rezvani
- Ayatollah Taleghani Hospital, Shahid Beheshti University of Medial Sciences, Tehran, Iran
| | - Hamid Reza Mirzaei
- Department of Radiation Oncology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jamshid Salamzadeh
- Food Safety Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Boldrin E, Malacrida S, Rumiato E, Battaglia G, Ruol A, Amadori A, Saggioro D. Association Between ERCC1 rs3212986 and ERCC2/XPD rs1799793 and OS in Patients With Advanced Esophageal Cancer. Front Oncol 2019; 9:85. [PMID: 30847299 PMCID: PMC6393335 DOI: 10.3389/fonc.2019.00085] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 01/29/2019] [Indexed: 12/15/2022] Open
Abstract
Esophageal cancer (EC) is a very aggressive tumor, and no reliable prognostic markers exist especially for resectable advanced neoplasia. The principal aim of this study was to investigate the association of germline polymorphisms in nucleotide excision repair (NER) pathway genes with the overall survival (OS) of patients with advanced EC. As a second aim, we also studied the association of NER gene variants with response to cisplatin-based chemotherapy. Among the EC patients referred to our Institution between 2004 and 2012, we selected a cohort of 180 patients diagnosed with a clinical tumor stage ranging from IIB and IVA. Patients were genotyped for four NER variants, two in the ERCC1 (rs11615 and rs3212986) and two in the ERCC2/XPD (rs1799793 and rs13181) genes. Kaplan–Meier analyses and Cox proportional hazards model were used to evaluate the associations of the selected variants with OS; association with response to neoadjuvant therapy was investigated using logistic regression. Results showed that the ERCC1 rs3212986 and the ERCC2/XPD rs1799793 were significantly associated with shorter OS. On the contrary, response association analysis displayed that, while rs11615 and rs3212986 in ERCC1 were associated with response, both ERCC2/XPD variants were not. By creating survival prediction models, we showed that the rs3212986 and the rs1799793 have a better predictability of the tumor stage alone. Furthermore, they were able to improve the power of the clinical model (AUC = 0.660 vs. AUC = 0.548, p = 0.004). In conclusion, our results indicate that the ERCC1 rs3212986 and the ERCC2/XPD rs1799793 could be used as surrogate markers for a better stratification of EC patients with advanced resectable tumor.
Collapse
Affiliation(s)
- Elisa Boldrin
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Sandro Malacrida
- Eurac Research, Institute of Mountain Emergency Medicine, Bolzano, Italy
| | - Enrica Rumiato
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Giorgio Battaglia
- Endoscopy Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Alberto Ruol
- Department of Surgical Sciences, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Alberto Amadori
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.,Department of Surgical Sciences, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Daniela Saggioro
- Immunology and Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
6
|
Creemers A, Ebbing EA, Pelgrim TC, Lagarde SM, van Etten-Jamaludin FS, van Berge Henegouwen MI, Hulshof MCCM, Krishnadath KK, Meijer SL, Bijlsma MF, van Oijen MGH, van Laarhoven HWM. A systematic review and meta-analysis of prognostic biomarkers in resectable esophageal adenocarcinomas. Sci Rep 2018; 8:13281. [PMID: 30185893 PMCID: PMC6125467 DOI: 10.1038/s41598-018-31548-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 08/20/2018] [Indexed: 02/07/2023] Open
Abstract
Targeted therapy is lagging behind in esophageal adenocarcinoma (EAC). To guide the development of new treatment strategies, we provide an overview of the prognostic biomarkers in resectable EAC treated with curative intent. The Medline, Cochrane and EMBASE databases were systematically searched, focusing on overall survival (OS). The quality of the studies was assessed using a scoring system ranging from 0-7 points based on modified REMARK criteria. To evaluate all identified prognostic biomarkers, the hallmarks of cancer were adapted to fit all biomarkers based on their biological function in EAC, resulting in the features angiogenesis, cell adhesion and extra-cellular matrix remodeling, cell cycle, immune, invasion and metastasis, proliferation, and self-renewal. Pooled hazard ratios (HR) and 95% confidence intervals (CI) were derived by random effects meta-analyses performed on each hallmarks of cancer feature. Of the 3298 unique articles identified, 84 were included, with a mean quality of 5.9 points (range 3.5-7). The hallmarks of cancer feature 'immune' was most significantly associated with worse OS (HR 1.88, (95%CI 1.20-2.93)). Of the 82 unique prognostic biomarkers identified, meta-analyses showed prominent biomarkers, including COX-2, PAK-1, p14ARF, PD-L1, MET, LC3B, IGFBP7 and LGR5, associated to each hallmark of cancer.
Collapse
Affiliation(s)
- Aafke Creemers
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | - Eva A Ebbing
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Thomas C Pelgrim
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sjoerd M Lagarde
- Department of Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Faridi S van Etten-Jamaludin
- Department of Medical Library Science, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | | | - Maarten C C M Hulshof
- Department of Radiotherapy, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Kausilia K Krishnadath
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Gastroenterology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Sybren L Meijer
- Department of Pathology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Maarten F Bijlsma
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Martijn G H van Oijen
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - Hanneke W M van Laarhoven
- Laboratory of Experimental Oncology and Radiobiology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Medical Oncology, Amsterdam UMC, Univ of Amsterdam, Cancer Center Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Arakawa Y, Shirai Y, Hayashi K, Tanaka Y, Matsumoto A, Nishikawa K, Yano S. Effects of gene polymorphisms on the risk of severe hyponatremia during DCF chemotherapy for patients with esophageal squamous cell carcinoma. Oncol Lett 2018; 16:5455-5462. [PMID: 30214618 DOI: 10.3892/ol.2018.9236] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 07/25/2018] [Indexed: 01/01/2023] Open
Abstract
Combination chemotherapy using docetaxel, cisplatin and 5-fluorouracil (DCF) is a promising treatment option for patients with advanced esophageal squamous cell carcinoma (ESCC), although its clinical application is limited by severe systemic toxicities. There are no validated markers for predicting the adverse effects caused by this regimen. This pharmacogenetic study enrolled 57 patients with chemotherapy-naive advanced ESCC between July 2012 and March 2016 (UMIN000008462). All patients received at least one course of DCF chemotherapy (docetaxel, 60 mg/m2 on day 1; cisplatin, 70 mg/m2 on day 1; 5-fluorouracil, 600 mg/m2 on days 1-5). The associations between four gene polymorphisms (ERCC1 rs11615, GSTP1 rs1695, TYMS rs151264360 and XPD rs13181) and the development of grade 3/4 adverse events during the first course of chemotherapy were prospectively investigated. The patients had a median age of 66 years (range, 45-77 years) and the majority were male (51 males vs. 6 females). The treatment settings were neoadjuvant (47 patients), adjuvant (1 patient) and salvage (9 patients), with dose intensities of 100% (51 patients) or 80% (6 patients). The severe adverse events were leukopenia (70.2%), neutropenia (86.0%), febrile neutropenia (36.8%), acute kidney injury (29.1%) and hyponatremia (43.9%). Two polymorphisms were independently associated with the development of severe hyponatremia among patients carrying the minor allele (vs. patients with major homozygote genotype): TYMS 3'-UTR rs151264360 (odds ratio, 3.64; 95% confidence interval, 1.11-11.9) and XPD Lys751Gln rs13181 (odds ratio, 10.1; 95% confidence interval, 1.10-93.3). Therefore, the presence of the TYMS and XPD polymorphisms may aid in identifying patients with a high risk of developing severe hyponatremia during DCF chemotherapy.
Collapse
Affiliation(s)
- Yasuhiro Arakawa
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Yoshihiro Shirai
- Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Kazumi Hayashi
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Yujiro Tanaka
- Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Akira Matsumoto
- Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Katsunori Nishikawa
- Department of Surgery, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| | - Shingo Yano
- Division of Clinical Oncology and Hematology, Department of Internal Medicine, The Jikei University School of Medicine, Tokyo 105-8471, Japan
| |
Collapse
|
8
|
Zhang R, Zhou F, Cheng L, Yu A, Zhu M, Wang M, Zhang Z, Xiang J, Wei Q. Genetic variants in nucleotide excision repair pathway predict survival of esophageal squamous cell cancer patients receiving platinum-based chemotherapy. Mol Carcinog 2018; 57:1553-1565. [PMID: 30035334 DOI: 10.1002/mc.22877] [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: 03/20/2018] [Revised: 06/22/2018] [Accepted: 07/19/2018] [Indexed: 12/19/2022]
Abstract
The benefits of platinum-based chemotherapy (PBC) on survival of esophageal squamous cell carcinoma (ESCC) patients are inexplicit due to the varied therapeutic effects. Nucleotide excision repair (NER) pathway plays a vital role in removing platinum-DNA adducts in tumor cells and hence may modulate the therapeutic effect and survival outcome. The present study assessed the associations of 26 potentially functional regulatory single nucleotide polymorphisms (rSNPs) in nine core NER genes with disease-free survival (DFS) and overall survival (OS) in 339 ESCC patients. We found that ERCC2 rs2097215 T and rs3916788 A, ERCC5 rs3759497 A and XPC rs3731054 C alleles were associated with unfavorable DFS. Patients carrying high-risk allele group (HRG, 5-8 risk alleles) had a significantly shorter DFS, compared with those carrying low-risk alleles (LRG, 0-4 risk alleles) [adjusted hazards ratio (HRadj ) = 1.64, 95%CI = 1.23-2.19, Padj < 0.001]. Three of these SNPs (ie, ERCC2 rs2097215 T and rs3916788 A and ERCC5 rs3759497 A) were also significantly associated with a poorer OS (HRG vs LRG: HRadj = 1.75, 95%CI = 1.23-2.47, Padj = 0.002). The expression quantitative trait loci (eQTL) analysis revealed significant genotype-expression correlations for ERCC5 rs3759497 and ERCC2 2097215 and rs3916788, which suggest regulatory roles of these SNPs. It appears that these NER variants may independently or jointly exert an impact on survival outcome of Chinese ESCC patients undergoing adjuvant platinum-based therapy. Large studies are warranted to validate these findings.
Collapse
Affiliation(s)
- Ruoxin Zhang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Fei Zhou
- Department of Oncology, Shanghai Jiao Tong University affiliated Shanghai General Hospital, Shanghai, China
| | - Lei Cheng
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Alexandria Yu
- Department of Public Health, Johns Hopkins University, Baltimore, Maryland
| | - Meiling Zhu
- Department of Oncology, Xinhua Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Mengyun Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhuanxu Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Jiaqing Xiang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Thoracic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qingyi Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China.,Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
9
|
Obiedat H, Alrabadi N, Sultan E, Al Shatti M, Zihlif M. The effect of ERCC1 and ERCC2 gene polymorphysims on response to cisplatin based therapy in osteosarcoma patients. BMC MEDICAL GENETICS 2018; 19:112. [PMID: 29980176 PMCID: PMC6035436 DOI: 10.1186/s12881-018-0627-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/13/2018] [Indexed: 01/28/2023]
Abstract
Background Cisplatin is one of the major drugs that used in the treatment of osteosarcoma. Cisplatin exerts its function by making cisplatin-DNA adducts culminating in cellular death. These adducts found to be repaired by nucleotide excision repair (NER) pathway. This study aimed to evaluate if polymorphisms in two main genes in the NER pathway, excision repair cross-complementing group 1 and 2 (ERCC1 and ERCC2) could affect the histological response to cisplatin based chemotherapy or clinical outcomes, particularly, event free survival (EFS) and overall survival (OS) rates. Method ERCC1 (C118T (rs11615) and C8092A (rs3212986)) and ERCC2 (A751C (rs171140) and G312A (rs1799793)) polymorphisms were analysed in 44 patients with osteosarcoma, who were treated with cisplatin based neoadjuvant chemotherapy. DNA was extracted from patient’s formalin-fixed paraffin-embedded (FFPE) samples, patient’s genotypes were determined by using polymerase chain reaction-restriction fragment length polymorphism PCR-RFLP assay. The distribution of the patients’ genotype and the allele frequencies were reported. The association between each of these genotypes and many clinical and patho-histological parameters (e.g. EFS, OS and patho-histological response to treatment) was examined. The associations between gender, tumor location, presence of metastasis at diagnosis, histological subtypes, and type of neoadjuvant chemotherapy and between the histological response, EFS and OS rates were also examined. Results This study revealed that there was a positive and significant association between ERCC1 C8092 A genotypes and median EFS rate in years; patients who were carriers of C allele (CC & CA) were found to have longer EFS rates than patients with AA genotype (P value = 0.006) and the median EFS rates were respectively as following: 2.04, 0.24 years. As well, both the presence of metastasis and the histological subtype at the time of diagnosis, were able to affect the EFS rate but not the OS. However, there was a positive correlation between OS rate and the patients’ primary response to treatment. Conclusions Our results suggested that ERCC1 8092 C allele may play a role as a candidate prognostic marker in patients with osteosarcoma. Electronic supplementary material The online version of this article (10.1186/s12881-018-0627-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hadeel Obiedat
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Nasr Alrabadi
- Department of Pharmacology, Faculty of medicine, Jordan University of Science and Technology (JUST), Irbid, 22110, Jordan.
| | - Eyad Sultan
- Department of Pediatric Oncology, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Marwa Al Shatti
- Department of Pathology and Laboratory, King Hussein Cancer Center (KHCC), Amman, Jordan
| | - Malek Zihlif
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
10
|
柏 启, 于 珺, 苟 云, 贺 生, 李 永, 黄 长, 汪 诚. ERCC1-C19007T基因多态性与中晚期食管癌铂类药物化疗敏感性的Meta分析. Shijie Huaren Xiaohua Zazhi 2017; 25:1854-1860. [DOI: 10.11569/wcjd.v25.i20.1854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
目的 定量分析中晚期食管癌患者切除修复交叉互补基因1(excision repair cross comple-menting 1, ERCC1)C-19007T基因多态性与铂类药物化疗敏感性的关系.
方法 计算机检索PubMed、EMBASE、Cochrane Library, 中文科技期刊数据库、中国生物医学文献数据库、中国期刊全文数据库和万方数据库, 收集有关中晚期食管癌患者ERCC1-C19007T基因多态性与基于铂类药物方案化疗敏感性的相关研究, 以临床化疗有效率(完全缓解+部分缓解)作为化疗敏感评价指标, 采用Reviewm5.2及Stata12.1软件进行统计学分析, 计算合并比值比(odd ratio, OR)及95%可信区间(confidence interval, CI).
结果 本研究共纳入8篇文献. Meta分析结果显示, 各基因型之间(CT vs CC: OR = 3.31, 95%CI: 1.94-5.64); (CT vs TT: OR = 5.48, 95%CI: 3.21-9.35); (CT vs CC+TT: OR = 4.06, 95%CI: 2.66-6.18); 差异有统计学意义, 表明ERCC1-C19007T基因多态性与中晚期食管癌对铂类化疗药物敏感性的差异有统计学意义.
结论 ERCC1-C19007T基因多态性可能与食管癌铂类药物化疗耐药相关.
Collapse
|
11
|
Lopes-Aguiar L, Costa EFD, Nogueira GAS, Lima TRP, Visacri MB, Pincinato EC, Calonga L, Mariano FV, de Almeida Milani Altemani AM, Altemani JMC, Coutinho-Camillo CM, Ribeiro Alves MAVF, Moriel P, Ramos CD, Chone CT, Lima CSP. XPD c.934G>A polymorphism of nucleotide excision repair pathway in outcome of head and neck squamous cell carcinoma patients treated with cisplatin chemoradiation. Oncotarget 2017; 8:16190-16201. [PMID: 26918827 PMCID: PMC5369956 DOI: 10.18632/oncotarget.7668] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 02/16/2016] [Indexed: 11/25/2022] Open
Abstract
This study aimed to investigate the associations of XPC c.2815A>C, XPD c.934G>A and c.2251A>C, XPF c.2505T>C and ERCC1 c.354C>T single nucleotide polymorphisms (SNPs) of nucleotide excision repair pathway in outcome of head and neck squamous cell carcinoma (HNSCC) patients treated with cisplatin (CDDP) chemoradiation. Patients with XPC c.2815AC or CC and XPD c.934GA or AA genotypes had 0.20 and 0.38 less chances of presenting moderate/severe ototoxicity and nausea, respectively. Patients with XPD c.934AA and c.2251AC or CC genotypes had 8.64, 12.29 and 3.55 more chances of achieving complete response (CR), consistent ototoxicity and nephrotoxicity, respectively. AA haplotype of XPD and ACT haplotype of XPD and ERCC1 SNPs were associated with 9.30 and 3.41 more chances of achieving CR and consistent nephrotoxicity, respectively. At 24 months of follow-up, patients with XPD c.934AA genotype presented lower progression-free survival and overall survival in Kaplan-Meier estimates, and differences between groups remained the same in univariate Cox analysis. Patients with XPD c.934AA genotype had 2.13 and 2.04 more risks of presenting tumor progression and death than others in multivariate Cox analysis. Our data present preliminary evidence that XPC c.2815A>C, XPD c.934G>A and c.2251A>C, and ERCC1 c.354C>T SNPs alter outcome of HNSCC patients treated with CDDP chemoradiation.
Collapse
Affiliation(s)
- Leisa Lopes-Aguiar
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | - Tathiane Regine Penna Lima
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Marília Berlofa Visacri
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Eder Carvalho Pincinato
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Luciane Calonga
- Department of Ophthalmology and Otorhinolaryngology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Fernanda Viviane Mariano
- Department of Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | | | | | | | | | - Patrícia Moriel
- Department of Clinical Pathology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Celso Dario Ramos
- Department of Radiology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Carlos Takahiro Chone
- Department of Ophthalmology and Otorhinolaryngology, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| | - Carmen Silvia Passos Lima
- Department of Internal Medicine, Faculty of Medical Sciences, University of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
12
|
Dobrzyńska I, Skrzydlewska E, Figaszewski ZA. Effects of Novel Dinuclear Cisplatinum(II) Complexes on the Electrical Properties of Human Molt-4 Leukemia Cells. Cell Biochem Biophys 2016; 71:1517-23. [PMID: 25399303 PMCID: PMC4449378 DOI: 10.1007/s12013-014-0375-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The aim of this study was to determine the influence of cisplatin and novel dinuclear platinum(II) complexes on the membrane electrical properties and lipid peroxidation levels of the Molt-4 human leukemia cell line. Changes in cell function may affect the basal electrical surface properties of cell membranes. These changes can be detected using electrokinetic measurements. Surface charge densities of Molt-4 cells were measured as a function of pH. A four-component equilibrium model was used to describe the interaction between the ions in solution and on cell membrane surfaces. Agreement was found between the experimental and theoretical charge variation curves of the leukemia cells at pH 2.5–9. Lipid peroxidation was estimated by measuring levels of 8-iso-prostaglandine F2α [isoprostanes]. Acid and base functional group concentrations and average association constants with hydroxyl ions were smaller in cisplatin- or dinuclear platinum(II) complex-treated leukemia cell membranes compared to those in untreated cancer cells, and the average association constants with hydrogen ions were higher. Levels of lipid peroxidation products in cisplatin- or dinuclear platinum(II) complex-treated leukemia cell were higher than those found in untreated cancer cells.
Collapse
Affiliation(s)
- Izabela Dobrzyńska
- Institute of Chemistry, University in Białystok, Al. Piłsudskiego 11/4, 15-443, Białystok, Poland.
| | - Elżbieta Skrzydlewska
- Department of Analytical Chemistry, Medical University of Białystok, Mickiewicza 2, 15-230, Białystok, Poland
| | - Zbigniew A Figaszewski
- Institute of Chemistry, University in Białystok, Al. Piłsudskiego 11/4, 15-443, Białystok, Poland
- Laboratory of Electrochemical Power Sources, Faculty of Chemistry, University of Warsaw, Pasteur St. 1, 02-093, Warsaw, Poland
| |
Collapse
|
13
|
Liu YM, Liu YK, Wang LW, Huang YC, Huang PI, Tsai TH, Chen YJ. The medicinal fungus Antrodia cinnamomea regulates DNA repair and enhances the radiosensitivity of human esophageal cancer cells. Onco Targets Ther 2016; 9:6651-6661. [PMID: 27826196 PMCID: PMC5096758 DOI: 10.2147/ott.s96355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
This study investigated the adjunctive effects of Antrodia cinnamomea mycelial fermentation broth (AC-MFB), a Taiwanese medicinal fungus, in enhancing the radiosensitivity of esophageal cancer cells. Human CE81T/VGH squamous and BE3 adenocarcinoma esophageal cancer cells were used in this study. A colony formation assay showed that pretreatment with AC-MFB decreased the survival of irradiated esophageal cancer cells, with a maximum sensitizer enhancement ratio of 1.91% and 37% survival. A DNA histogram study showed that AC-MFB pretreatment enhanced cell cycle arrest at the G2/M phase, the most radiosensitive phase. An immunofluorescence assay and a Western blotting assay showed that AC-MFB delayed the abrogation of γ-H2AX, upregulated p21 expression, and attenuated the radiation-induced phosphorylation of ataxia telangiectasia-mutated kinase and checkpoint kinase 2. An in vivo validation study showed that AC-MFB treatment tended to have a synergistic effect with radiation on the tumor growth delay of CE81T/VGH cells in BALB/c mice. These data suggest that this edible fungus product could enhance the effect of radiotherapy against esophageal cancer.
Collapse
Affiliation(s)
- Yu-Ming Liu
- Department of Oncology, Division of Radiation Oncology, Taipei Veterans General Hospital; School of Medicine, Institute of Traditional Medicine, National Yang Ming University, Taipei; School of Medicine, National Yang Ming University, Taipei
| | - Yu-Kuo Liu
- Department of Chemical and Material Engineering, Chang Gung University, Taoyuan City
| | - Ling-Wei Wang
- Department of Oncology, Division of Radiation Oncology, Taipei Veterans General Hospital; School of Medicine, National Yang Ming University, Taipei
| | - Yu-Chuen Huang
- Department of Medical Research, China Medical University Hospital; School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung
| | - Pin-I Huang
- Department of Oncology, Division of Radiation Oncology, Taipei Veterans General Hospital; School of Medicine, National Yang Ming University, Taipei
| | - Tung-Hu Tsai
- School of Medicine, Institute of Traditional Medicine, National Yang Ming University, Taipei
| | - Yu-Jen Chen
- School of Medicine, Institute of Traditional Medicine, National Yang Ming University, Taipei; Department of Radiation Oncology, MacKay Memorial Hospital, New Taipei City, Taiwan
| |
Collapse
|
14
|
Mlakar V, Huezo-Diaz Curtis P, Satyanarayana Uppugunduri CR, Krajinovic M, Ansari M. Pharmacogenomics in Pediatric Oncology: Review of Gene-Drug Associations for Clinical Use. Int J Mol Sci 2016; 17:ijms17091502. [PMID: 27618021 PMCID: PMC5037779 DOI: 10.3390/ijms17091502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 08/02/2016] [Accepted: 08/15/2016] [Indexed: 02/07/2023] Open
Abstract
During the 3rd congress of the European Society of Pharmacogenomics and Personalised Therapy (ESPT) in Budapest in 2015, a preliminary meeting was held aimed at establishing a pediatric individualized treatment in oncology and hematology committees. The main purpose was to facilitate the transfer and harmonization of pharmacogenetic testing from research into clinics, to bring together basic and translational research and to educate health professionals throughout Europe. The objective of this review was to provide the attendees of the meeting as well as the larger scientific community an insight into the compiled evidence regarding current pharmacogenomics knowledge in pediatric oncology. This preliminary evaluation will help steer the committee’s work and should give the reader an idea at which stage researchers and clinicians are, in terms of personalizing medicine for children with cancer. From the evidence presented here, future recommendations to achieve this goal will also be suggested.
Collapse
Affiliation(s)
- Vid Mlakar
- Cansearch Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
| | - Patricia Huezo-Diaz Curtis
- Cansearch Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
| | | | - Maja Krajinovic
- Charles-Bruneau Cancer Center, Centre hospitalier universitaire Sainte-Justine, 4515 Rue de Rouen, Montreal, QC H1V 1H1, Canada.
- Department of Pediatrics, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, QC H3T 1J4, Canada.
- Department of Pharmacology, Faculty of Medicine, University of Montreal, 2900 Boulevard Edouard-Montpetit, Montreal, QC H3T 1J4, Canada.
| | - Marc Ansari
- Cansearch Research Laboratory, Geneva University Medical School, Avenue de la Roseraie 64, 1205 Geneva, Switzerland.
- Pediatric Department, Onco-Hematology Unit, Geneva University Hospital, Rue Willy-Donzé 6, 1205 Geneva, Switzerland.
| |
Collapse
|
15
|
Rumiato E, Boldrin E, Amadori A, Saggioro D. Predictive role of host constitutive variants in neoadjuvant therapy of esophageal cancer. Pharmacogenomics 2016; 17:805-20. [PMID: 27181132 DOI: 10.2217/pgs-2016-0009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Chemoradiotherapy followed by surgery is at present the standard therapeutic approach for esophageal cancer (EC) in patients with resectable tumor. However, response to neoadjuvant therapy is characterized by a strong interpatient variability, and the identification of markers predictive of outcome is mandatory. In this review, taking into account the currently available literature, we report the impact that host genetic variables can have on EC neoadjuvant therapy. We mainly focused on the gene variants involved in the pharmacokinetics and pharmacodynamics of the common chemotherapeutic drugs used to treat EC patients, commented on the weakness of the present knowledge, and discussed the future strategies to achieve a more personalized and effective EC treatment.
Collapse
Affiliation(s)
- Enrica Rumiato
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Elisa Boldrin
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Alberto Amadori
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy.,Department of Surgical Sciences, Oncology & Gastroenterology, University of Padova, Padova, Italy
| | - Daniela Saggioro
- Immunology & Molecular Oncology, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| |
Collapse
|
16
|
Gusella M, Giacopuzzi S, Bertolaso L, Zanoni A, Pezzolo E, Modena Y, Menon D, Paganin P, Weindelmayer J, Crepaldi G, De Manzoni G, Pasini F. Genetic prediction of long-term survival after neoadjuvant chemoradiation in locally advanced esophageal cancer. THE PHARMACOGENOMICS JOURNAL 2016; 17:252-257. [PMID: 26927287 DOI: 10.1038/tpj.2016.9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 11/12/2015] [Accepted: 12/23/2015] [Indexed: 12/26/2022]
Abstract
Candidate genes involved in DNA repair, 5-fluorouracil metabolism and drug detoxification were genotyped in 124 patients receiving neoadjuvant chemoradiation treatment for locally advanced esophageal cancer and their predictive role for long-term relapse-free survival (RFS) and cancer-specific survival (CSS) were evaluated. A panel including MTHFR 677TT, MDR1 2677GT, GSTP1 114CC, XPC 499CC and XPC 939AC+CC, defined as high-risk genotypes, discriminated subgroups with significantly different outcomes. When the panel was combined with histology, patients split into two subsets with 5-year RFS and CSS rates of 65% vs 27% (hazard ratio (HR) 3.0, P<0.0001) and 69% vs 31% (HR 2.9, P<0.0001), respectively. Combining the 5-single-nucleotide polymorphism (5-SNP) panel with pathological response defined two major informative risk classes with 5-year PFS and CSS rates of 79.4% vs 17.7% (HR 6.71, P<0.0001) and 79.3% vs 26.3% (HR 6.25, P<0.0001), respectively. This classification achieved a sensitivity of 79%, a specificity of 85.4% and an accuracy of 81.8%.
Collapse
Affiliation(s)
- M Gusella
- Department of Oncology, Laboratory of Pharmacology and Molecular Biology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| | - S Giacopuzzi
- Upper Gastrointestinal Surgery Division, University of Verona, Verona, Italy
| | - L Bertolaso
- Department of Oncology, Laboratory of Pharmacology and Molecular Biology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| | - A Zanoni
- Upper Gastrointestinal Surgery Division, University of Verona, Verona, Italy
| | - E Pezzolo
- Department of Oncology, Laboratory of Pharmacology and Molecular Biology, Azienda ULSS 18-Rovigo, Rovigo, Italy.,Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Y Modena
- Department of Oncology, Unit of Medical Oncology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| | - D Menon
- Department of Oncology, Unit of Medical Oncology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| | - P Paganin
- Department of Oncology, Laboratory of Pharmacology and Molecular Biology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| | - J Weindelmayer
- Upper Gastrointestinal Surgery Division, University of Verona, Verona, Italy
| | - G Crepaldi
- Department of Oncology, Unit of Medical Oncology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| | - G De Manzoni
- Upper Gastrointestinal Surgery Division, University of Verona, Verona, Italy
| | - F Pasini
- Department of Oncology, Unit of Medical Oncology, Azienda ULSS 18-Rovigo, Rovigo, Italy
| |
Collapse
|
17
|
Ferreira JA, Peixoto A, Neves M, Gaiteiro C, Reis CA, Assaraf YG, Santos LL. Mechanisms of cisplatin resistance and targeting of cancer stem cells: Adding glycosylation to the equation. Drug Resist Updat 2016; 24:34-54. [DOI: 10.1016/j.drup.2015.11.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 11/09/2015] [Accepted: 11/18/2015] [Indexed: 02/06/2023]
|
18
|
Phase 1a/1b and pharmacogenetic study of docetaxel, oxaliplatin and capecitabine in patients with advanced cancer of the stomach or the gastroesophageal junction. Cancer Chemother Pharmacol 2015; 76:1285-95. [DOI: 10.1007/s00280-015-2872-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 09/08/2015] [Indexed: 11/27/2022]
|
19
|
Deng JH, Deng J, Shi DH, Ouyang XN, Niu PG. Clinical outcome of cisplatin-based chemotherapy is associated with the polymorphisms of GSTP1 and XRCC1 in advanced non-small cell lung cancer patients. Clin Transl Oncol 2015; 17:720-6. [PMID: 26033426 DOI: 10.1007/s12094-015-1299-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Accepted: 05/09/2015] [Indexed: 11/25/2022]
Abstract
INTRODUCTION This study is to evaluate the association of polymorphisms of glutathione S-transferase P1 (GSTP1), copper-transporting P-type adenosine triphosphatase A (ATP7A) and X-ray repair cross-complementing group 1 (XRCC1) with the efficacy and toxicity of cisplatin-based treatment in advanced non-small cell lung cancer (NSCLC) patients. MATERIALS AND METHODS The outcomes of 97 advanced non-small cell lung cancer patients treated with cisplatin-based chemotherapy were estimated. GSTP1, ATP7A, and XRCC1 genetic polymorphisms were determined via polymerase chain reaction of restriction fragment length polymorphism (PCR-RFLP) and DNA sequencing. Association of the polymorphisms with the efficacy and toxicity of cisplatin was analyzed, respectively. RESULTS Significant associations were observed between GSTP1 A313G and response rate (RR) (p = 0.027), disease control rate (DCR) (p = 0.019), and progression-free survival (PFS) (p = 0.044), respectively. Patients with AG and GG of GSTP1 have notably lower risk of anemia (p = 0.046). XRCC1 A1196G was associated with the incidence of lymphopenia (p = 0.024) and diarrhea (p = 0.020). ATP7A C2299G was not related with RR, DCR, PFS, and the risk of toxicity. CONCLUSIONS Advanced NSCLC patients with AA genotype of GSTP1 would obtain better curative effect followed with more risk of anemia when treated by cisplatin-based chemotherapy. ATP7A C2299G does not impact the efficacy and toxicity of cisplatin-based chemotherapy. XRCC1 1196A allele could predict the incidence of lymphopenia and diarrhea.
Collapse
Affiliation(s)
- J-H Deng
- Department of Pharmacy, Fujian Provincial Maternal and Child Health Hospital, 18 Daoshan Road, Fuzhou, Fujian, China
| | | | | | | | | |
Collapse
|
20
|
Han B, Guo Z, Ma Y, Kang S, Wang Y, Wei Q, Wu X. Association of GSTP1 and XRCC1 gene polymorphisms with clinical outcome of advanced non-small cell lung cancer patients with cisplatin-based chemotherapy. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:4113-4119. [PMID: 26097600 PMCID: PMC4466987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/30/2015] [Indexed: 06/04/2023]
Abstract
We investigated the association between the clinical outcome and GSTP1 and XRCC1 gene polymorphisms in advanced NSCLC patients with cisplatin-based chemotherapy. We prospectively recruited 325 NSCLC patients between January 2010 and January 2014. Genotypes of GSTP1 A313G, XRCC1 Arg194Trp, Arg280His and Arg399Gln were conducted using polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) assay. AG and GG genotypes of GSTP1 A313G were correlated with a higher CR + PR when compared with AA genotype. Furthermore, GA and AA genotypes of XRCC1 Arg399Gln were associated with more CR + PR when compared with GG genotype. In the Cox proportional hazards model, GG genotype of GSTP1 A313G was significantly correlated with a longer median survival time when compared with AA genotype, and it is associated with a heavy decreased risk of death from NSCLC. Moreover, GA and AA genotypes of XRCC1 Arg399Gln had a significantly longer median survival time, and GA and AA genotypes were significantly associated with a moderate reduced risk of death from NSCLC. GSTP1 A313G and XRCC1 Arg399Gln gene polymorphisms might influence the response to cisplatin-based chemotherapy and affect the clinical outcome of advanced NSCLC.
Collapse
MESH Headings
- Adenocarcinoma/drug therapy
- Adenocarcinoma/enzymology
- Adenocarcinoma/genetics
- Adenocarcinoma/mortality
- Adenocarcinoma/pathology
- Adenocarcinoma of Lung
- Aged
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Biomarkers, Tumor/genetics
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/enzymology
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/mortality
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/enzymology
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/mortality
- Carcinoma, Squamous Cell/pathology
- Chi-Square Distribution
- Cisplatin/therapeutic use
- DNA-Binding Proteins/genetics
- Female
- Gene Frequency
- Genetic Predisposition to Disease
- Glutathione S-Transferase pi/genetics
- Humans
- Kaplan-Meier Estimate
- Logistic Models
- Lung Neoplasms/drug therapy
- Lung Neoplasms/enzymology
- Lung Neoplasms/genetics
- Lung Neoplasms/mortality
- Lung Neoplasms/pathology
- Male
- Middle Aged
- Multivariate Analysis
- Odds Ratio
- Phenotype
- Polymorphism, Genetic
- Proportional Hazards Models
- Prospective Studies
- Protective Factors
- Risk Factors
- Time Factors
- Treatment Outcome
- X-ray Repair Cross Complementing Protein 1
Collapse
Affiliation(s)
- Bateer Han
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, China
- Department of Thoracic Surgery, Affiliated People’s Hospital of Inner Mongolia Medical UniversityHohhot 010020, China
| | - Zhanlin Guo
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical UniversityHohhot 010050, China
| | - Ying Ma
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical UniversityHohhot 010050, China
| | - Shirong Kang
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical UniversityHohhot 010050, China
| | - Yufei Wang
- Department of Thoracic Surgery, Affiliated Hospital of Inner Mongolia Medical UniversityHohhot 010050, China
| | - Qiyou Wei
- Department of Thoracic Surgery, Affiliated People’s Hospital of Inner Mongolia Medical UniversityHohhot 010020, China
| | - Xu Wu
- Department of Thoracic Surgery, Nanfang Hospital, Southern Medical UniversityGuangzhou 510515, China
| |
Collapse
|
21
|
Yu X, Xiao H, Zhao B, Zhang X, Wang G. DNA repair gene ERCC1 C118T polymorphism predicts sensitivity of recurrent esophageal cancer to radiochemotherapy in a Chinese population. Thorac Cancer 2015; 6:741-8. [PMID: 26557912 PMCID: PMC4632926 DOI: 10.1111/1759-7714.12251] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/04/2015] [Indexed: 02/06/2023] Open
Abstract
Background DNA repair gene polymorphisms could alter DNA repair capacity and therefore associate with tumor sensitivity to radiochemotherapy. This study assessed excision repair cross-complementing group 1 (ERCC1) C118T and X-ray cross-complementing group 1 (XRCC1) G399A single-nucleotide polymorphisms in esophageal patients for an association with sensitivity to radiation and chemotherapy. Methods Esophageal squamous cell carcinoma patients (n = 118) who relapsed after surgery were enrolled for assessment of ERCC1 C118T and XRCC1 G399A polymorphisms by direct DNA sequencing. Results The response rate of treatments was 48.30%: 14 complete response (CR, 11.86%), 43 partial response (PR, 36.44%), 49 stable disease (SD, 41.53%), and 12 progressive disease (PD, 10.17%). ERCC1 C118T was significantly associated with treatment response (C/T vs. C/C + T/T, odds ratio [OR] = 6.035, 95% confidence interval [CI]: 2.114–17.226, P = 0.001) after adjusting for other clinicopathological factors. Patients carrying the C/T genotype had significantly prolonged overall survival (OS) compared with C/C and T/T (median OS 43.00 vs. 27.00, P = 0.027). Multivariate Cox regression showed that a response was only an independent prognostic factor for OS (CR + PR vs. SD+PD, HR = 0.471 95% CI 0.269–0.826, P = 0.009). Grade III and IV adverse events occurred in 12 of 118 patients (10.17%). Only concurrent radiochemotherapy significantly increased these adverse events (OR = 26.529, 95% CI 2.312–304.389, P = 0.008). Conclusion ERCC1 C118T could be a predictive factor for the response to radiotherapy and chemotherapy, but not a prognostic factor for OS in esophageal cancer patients after surgery.
Collapse
Affiliation(s)
- Xian Yu
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University Chongqing, China
| | - He Xiao
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University Chongqing, China
| | - Baojian Zhao
- Beijing Epigenetics Biotechnology Co., Ltd Beijing, China
| | - Xu Zhang
- Beijing Epigenetics Biotechnology Co., Ltd Beijing, China
| | - Ge Wang
- Cancer Center, Daping Hospital and Research Institute of Surgery, The Third Military Medical University Chongqing, China
| |
Collapse
|
22
|
Ma SC, Zhao Y, Zhang T, Ling XL, Zhao D. Association between the ERCC1 rs11615 polymorphism and clinical outcomes of oxaliplatin-based chemotherapies in gastrointestinal cancer: a meta-analysis. Onco Targets Ther 2015; 8:641-8. [PMID: 25834456 PMCID: PMC4365759 DOI: 10.2147/ott.s80913] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Purpose The relationship between the excision repair cross-complementing 1 (ERCC1) rs11615 polymorphism (C/T) and responses to oxaliplatin-based chemotherapy for gastric cancer (GC) and colorectal cancer (CRC) patients is controversial. Therefore, we performed a meta-analysis to assess this relationship. Method Relevant studies were retrieved by searching the PubMed database. A systematic review and meta-analysis was performed to evaluate the predictive value of the ERCC1 rs11615 polymorphism for the clinical outcomes of GC and CRC patients receiving oxaliplatin-based chemotherapy. Therapeutic response to chemotherapy, progression-free survival (PFS), and overall survival (OS) were analyzed. Results A total of 22 studies were included in this meta-analysis, including 1,242 cases of GC and 1,772 cases of CRC. For the ERCC1 rs11615 polymorphism, the T allele was associated with a reduced response to chemotherapy in Asians and GC patients (P<0.05). On the other hand, the T allele was associated with a significant increase in the risk for shorter PFS and OS in all patients (PFS: hazard ratio [HR] =1.22, P<0.001, 95% confidence interval [CI] =0.93–1.51 and OS: HR =1.12, P<0.001, 95% CI =0.85–1.40). Conclusion The ERCC1 rs11615 polymorphism was closely associated with the clinical outcomes of GC and CRC patients treated with oxaliplatin-based chemotherapy.
Collapse
Affiliation(s)
- Shou-Cheng Ma
- Department of Oncology, The First Hospital of Lanzhou University (The Branch Hospital of Donggang), Lanzhou, Gansu Province, People's Republic of China
| | - Yue Zhao
- Department of Gastroenterology, The First Hospital of Lanzhou University (The Branch Hospital of Donggang), Lanzhou, Gansu Province, People's Republic of China
| | - Tao Zhang
- Department of Oncology, The First Hospital of Lanzhou University (The Branch Hospital of Donggang), Lanzhou, Gansu Province, People's Republic of China
| | - Xiao-Ling Ling
- Department of Oncology, The First Hospital of Lanzhou University (The Branch Hospital of Donggang), Lanzhou, Gansu Province, People's Republic of China
| | - Da Zhao
- Department of Oncology, The First Hospital of Lanzhou University (The Branch Hospital of Donggang), Lanzhou, Gansu Province, People's Republic of China
| |
Collapse
|
23
|
Kalikaki A, Voutsina A, Koutsopoulos A, Papadaki C, Sfakianaki M, Yachnakis E, Xyrafas A, Kotsakis A, Agelaki S, Souglakos J, Mavroudis D, Georgoulias V. ERCC1 SNPs as Potential Predictive Biomarkers in Non-Small Cell Lung Cancer Patients Treated With Platinum-Based Chemotherapy. Cancer Invest 2015; 33:107-13. [PMID: 25647444 DOI: 10.3109/07357907.2014.1001897] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Polymorphisms in ERCC1, XPD, and XRCC1 were examined for (a) association with the clinical outcome of 107 non-small cell lung cancer patients receiving front-line platinum-based chemotherapy, and (b) correlation with the ERCC1 mRNA levels of 176 chemo-naive primary tumors. The ERCC1-C8092 allele and the number of ERCC1 polymorphic variants (C8092A and Asn118Asn) were associated with progression-free survival. In non-squamous histology, tumoral ERCC1 mRNA levels were lower in patients homozygous for ERCC1-C8092 as compared with the patients carrying the A allele (p = .024). These findings merit investigation in larger cohorts of patients treated with uniform regimens.
Collapse
Affiliation(s)
- Aristea Kalikaki
- Laboratory of Tumor Cell Biology, School of Medicine, University of Crete , Heraklion, Crete , Greece , 1
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Roco A, Cayún J, Contreras S, Stojanova J, Quiñones L. Can pharmacogenetics explain efficacy and safety of cisplatin pharmacotherapy? Front Genet 2014; 5:391. [PMID: 25452763 PMCID: PMC4231946 DOI: 10.3389/fgene.2014.00391] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2014] [Accepted: 10/25/2014] [Indexed: 12/12/2022] Open
Abstract
Several recent pharmacogenetic studies have investigated the variability in both outcome and toxicity in cisplatin-based therapies. These studies have focused on the genetic variability of therapeutic targets that could affect cisplatin response and toxicity in diverse type of cancer including lung, gastric, ovarian, testicular, and esophageal cancer. In this review, we seek to update the reader in this area of investigation, focusing primarily on DNA reparation enzymes and cisplatin metabolism through Glutathione S-Transferases (GSTs). Current evidence indicates a potential application of pharmacogenetics in therapeutic schemes in which cisplatin is the cornerstone of these treatments. Therefore, a collaborative effort is required to study these molecular characteristics in order to generate a genetic panel with clinical utility.
Collapse
Affiliation(s)
- Angela Roco
- Servicio de Salud Metropolitano Occidente Santiago, Chile ; Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Molecular and Clinical Pharmacology Program, ICBM - Insituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile Santiago, Chile
| | - Juan Cayún
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Molecular and Clinical Pharmacology Program, ICBM - Insituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile Santiago, Chile
| | - Stephania Contreras
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Molecular and Clinical Pharmacology Program, ICBM - Insituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile Santiago, Chile
| | - Jana Stojanova
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Molecular and Clinical Pharmacology Program, ICBM - Insituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile Santiago, Chile
| | - Luis Quiñones
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics (CQF), Molecular and Clinical Pharmacology Program, ICBM - Insituto de Ciencias Biomédicas, Faculty of Medicine, University of Chile Santiago, Chile
| |
Collapse
|
25
|
Razzouk S. Translational genomics and head and neck cancer: toward precision medicine. Clin Genet 2014; 86:412-21. [PMID: 25143247 DOI: 10.1111/cge.12487] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 08/12/2014] [Accepted: 08/18/2014] [Indexed: 12/19/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) comprise a wide spectrum of neoplasms with different tumor biologies, prognosis and response to therapies. Current tumor classification and traditional diagnostic methods (e.g. clinical assessment, histopathology) are limited in their capacity to determine prognosis and clinical decision-making. Despite recent improvements in treatment, the outcome for patients with HNSCC remains poor. Similar to most tumors, several patient-related factors, (e.g. genetics and environment) and disease-related factors (e.g. tumor location, TMN staging) play a significant role on survival. Thus, the problem in defining the prognosis is that the clinical course and response to treatment differ considerably among patients. Such interindividual variability is related to the heterogeneity of the tumor, genetic and epigenetic variations, thus reflecting the interaction of multiple biological components that result in a unique phenotype. Integrative genomics are developed to identify the molecular pathways leading to cancer at the individual level and find novel prognostic markers for HNSCC, hence tailoring a treatment accordingly. Such genetic-based personalized diagnosis allows tumor stratification and implementation of targeted therapy. Modern medicine includes new drugs that disrupt the implicated molecules and their signaling pathways. Here, we summarize the current state of knowledge that elucidates the translation of genetic data into clinical benefit.
Collapse
Affiliation(s)
- S Razzouk
- Department of Periodontology and Implant Dentistry, New York University College of Dentistry, New York City, NY; Private practice, Beirut, Lebanon
| |
Collapse
|
26
|
Findlay JM, Middleton MR, Tomlinson I. A systematic review and meta-analysis of somatic and germline DNA sequence biomarkers of esophageal cancer survival, therapy response and stage. Ann Oncol 2014; 26:624-644. [PMID: 25214541 PMCID: PMC4374384 DOI: 10.1093/annonc/mdu449] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent advances in next generation sequencing reinforce the potential for DNA sequence markers to guide esophageal cancer management. We report the first systematic review and meta-analysis, identifying 94 markers of outcome and 41 of stage. Overall, evidence was poor. Meta-analyses demonstrated outcome associations for 6 tumor and 9 germline variants: priorities for prospective evaluation. Introduction There is an urgent need for biomarkers to help predict prognosis and guide management of esophageal cancer. This review identifies, evaluates and meta-analyses the evidence for reported somatic and germline DNA sequence biomarkers of outcome and stage. Methods A systematic review was carried out of the PubMed, EMBASE and Cochrane databases (20 August 2014), in conjunction with the ASCO Level of Evidence scale for biomarker research. Meta-analyses were carried out for all reported markers associated with outcome measures by more than one study. Results Four thousand and four articles were identified, 762 retrieved and 182 studies included. There were 65 reported markers of survival or recurrence 12 (18.5%) were excluded due to multiple comparisons. Following meta-analysis, significant associations were seen for six tumor variants (mutant TP53 and PIK3CA, copy number gain of ERBB2/HER2, CCND1 and FGF3, and chromosomal instability/ploidy) and seven germline polymorphisms: ERCC1 rs3212986, ERCC2 rs1799793, TP53 rs1042522, MDM2 rs2279744, TYMS rs34743033, ABCB1 rs1045642 and MTHFR rs1801133. Twelve germline markers of treatment complications were reported; 10 were excluded. Two tumor and 15 germline markers (11 excluded) of chemo (radio)therapy response were reported. Following meta-analysis, associations were demonstrated for mutant TP53, ERCC1 rs11615 and XRCC1 rs25487. There were 41 tumor/germline reported markers of stage; 27 (65.9%) were excluded. Conclusions Numerous DNA markers of outcome and stage have been reported, yet few are backed by high-quality evidence. Despite this, a small number of variants appear reliable. These merit evaluation in prospective trials, within the context of high-throughput sequencing and gene expression.
Collapse
Affiliation(s)
- J M Findlay
- Molecular and Population Genetics, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford; Oxford OesophagoGastric Centre
| | - M R Middleton
- NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - I Tomlinson
- Molecular and Population Genetics, Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford; NIHR Oxford Biomedical Research Centre, Churchill Hospital, Oxford, UK.
| |
Collapse
|
27
|
Patel JN, Fuchs CS, Owzar K, Chen Z, McLeod HL. Gastric cancer pharmacogenetics: progress or old tripe? Pharmacogenomics 2014; 14:1053-64. [PMID: 23837480 DOI: 10.2217/pgs.13.88] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer remains the second most frequent cause of cancer-related mortality. While surgery is traditionally the initial treatment for early-stage disease, the addition of chemotherapy has been shown to significantly increase overall survival and progression-free survival in advanced and metastatic stages of disease. However, despite the incorporation of newer chemotherapies and regimens into gastric cancer clinical trials, the response rate and median overall survival for treated patients has not significantly improved throughout the years; therefore, newer therapeutic approaches to improve upon the medication selection process are warranted. Treatment and dose selection based on patient factors, such as genetic variation, may provide a more rational and potentially more powerful means of personalizing chemotherapy. This review provides an update on the current status of pharmacogenetic studies regarding germline DNA mutations that may alter response to chemotherapeutic agents used to treat gastric cancer, including perspectives on clinical translation and future work.
Collapse
Affiliation(s)
- Jai N Patel
- UNC Institute for Pharmacogenomics & Individualized Therapy, University of North Carolina, Chapel Hill, NC 27599-7361, USA
| | | | | | | | | |
Collapse
|
28
|
Nucleotide excision repair: why is it not used to predict response to platinum-based chemotherapy? Cancer Lett 2014; 346:163-71. [PMID: 24462818 DOI: 10.1016/j.canlet.2014.01.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 02/07/2023]
Abstract
Platinum based therapy is one of the most effectively used chemotherapeutic treatments for cancer. The mechanism of action of platinum compounds is to damage DNA and drive cells into apoptosis. The most commonly used platinum containing agents are cis-diammine-dichloroplatinum (II)], more commonly known as cisplatin, its analogue carboplatin, and oxaliplatin. Cisplatin is used to treat a wide variety of tumours such as ovarian, testicular, head and neck and non-small cell lung cancers (NSCLCs). In addition, it forms the basis of most combined treatment regimes. Despite this, cisplatin and its analogues are extremely toxic and although some patients benefit substantially from treatment, a large proportion suffer the toxic side effects without any therapeutic benefit. Nucleotide excision repair (NER) is a versatile DNA repair system that recognises DNA damage induced by platinum based therapy. For many years the components of the NER pathway have been studied to determine mRNA and protein expression levels in response or resistance to cisplatin in many forms of cancer; particularly testicular, ovarian and NSCLCs. Despite the consistent finding that over or under expression of subsets of NER proteins and mRNA highly correlate with response to cisplatin, the translation of these findings into the clinical setting has not been forthcoming. This review summarises the results of previous investigations into NER in cisplatin response and clinical trials where the expression of NER proteins were compared to the response to platinum therapies in treatment.
Collapse
|
29
|
Molecular docking and molecular dynamics study on the effect of ERCC1 deleterious polymorphisms in ERCC1-XPF heterodimer. Appl Biochem Biotechnol 2013; 172:1265-81. [PMID: 24158589 DOI: 10.1007/s12010-013-0592-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2013] [Accepted: 10/03/2013] [Indexed: 11/27/2022]
Abstract
Excision repair cross complementation group 1 (ERCC1) is an important protein in the nucleotide excision repair (NER) pathway, which is responsible for removing DNA adducts induced by platinum based compounds. The heterodimer ERCC1-XPF is one of two endonucleases required for NER. Genetic variations or polymorphisms in ERCC1 gene alter DNA repair capacity. Reduced DNA repair (NER) capacity may result in tumors and enhances cisplatin chemotherapy in cancer patients, which functions by causing DNA damage. Therefore, ERCC1 variants have the potential to be used as a strong candidate biomarker in cancer treatments. In this study we identified five variants V116M, R156Q, A199T, S267P, and R322C of ERCC1 gene as highly deleterious. Further structural and functional analysis has been conducted for ERCC1 protein in the presence of three variants V116M, R156Q, and A199T. Occurrence of theses variations adversely affected the regular interaction between ERCC1 and XPF protein. Analysis of 20 ns molecular dynamics simulation trajectories reveals that the predicted deleterious variants altered the ERCC1-XPF complex stability, flexibility, and surface area. Notably, the number of hydrogen bonds in ERCC1-XPF mutant complexes decreased in the molecular dynamic simulation periods. Overall, this study explores the link between the ERCC1 deleterious variants and cisplatin chemotherapy for various cancers with the help of molecular docking and molecular dynamic approaches.
Collapse
|
30
|
Guo LY, Jin XP, Niu W, Li XF, Liu BH, Wang YL. Association of XPD and XRCC1 genetic polymorphisms with hepatocellular carcinoma risk. Asian Pac J Cancer Prev 2013; 13:4423-6. [PMID: 23167354 DOI: 10.7314/apjcp.2012.13.9.4423] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AIM XRCC1 and XPD are two major repair genes involved in nucleotide excision repair (NER), which is reported to be associated with risk of several cancers. We explored the association of XRCC1 and XPD polymorphisms with the risk of HCC. METHODS A total of 410 cases with HCC and 410 health controls were collected. XRCC1 Arg194Trp, XRCC1 Arg399Gln, XPD Lys751Gln and XPD Asp312Asn genotyping was performed by duplex polymerase-chain-reaction with the confronting-two-pair primer (PCR-CTPP) method. RESULTS XRCC1 194Trp/Trp was strongly significantly associated with an increased risk of HCC cancer when compared with the wide-type genotype (OR=2.26, 95% CI=(1.23-5.38). Individuals carrying the XRCC1 399Gln/ Gln showed increased risk of HCC (OR=1.74, 95%CI=1.06-2.74). The XPD 751Gln/Gln and Gln allele genotype were associated with strong elevated susceptibility to HCC (OR=3.51 and 1.42, respectively). CONCLUSION These results suggest that polymorphisms in XRCC1 and XPD may have functional significance in risk of HCC.
Collapse
Affiliation(s)
- Lian-Yi Guo
- Department of Gastroenterology, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, China
| | | | | | | | | | | |
Collapse
|
31
|
Zhao HL, Han S, Li L, Ding JX, Yang JY. Role of ERCC1 in cisplatin resistance in esophageal cancer. Shijie Huaren Xiaohua Zazhi 2013; 21:1493-1497. [DOI: 10.11569/wcjd.v21.i16.1493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is one of several chemotherapeutic drugs commonly used to treat esophageal cancer. Nucleotide excision repair (NER) pathway plays an important role in repairing cisplatin-caused DNA damage. It has been demonstrated recently that the key enzyme of this pathway, excision repair crosscomplimenting 1 (ERCC1), is a factor determining cisplatin resistance and patient's response to cisplatin treatment. Further studies on the relationship between ERCC1 and cisplatin resistance will improve our understanding of cisplatin resistance in patients with esophageal cancer.
Collapse
|
32
|
Song X, Sturgis EM, Jin L, Wang Z, Wei Q, Li G. Variants in nucleotide excision repair core genes and susceptibility to recurrence of squamous cell carcinoma of the oropharynx. Int J Cancer 2013; 133:695-704. [PMID: 23335232 DOI: 10.1002/ijc.28051] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2012] [Accepted: 01/14/2013] [Indexed: 12/20/2022]
Abstract
Genetically determined capacity for NER may modulate both cancer risk and prognosis. Thus, we evaluated associations of seven selected variants in the NER core genes with recurrence risk in 658 squamous cell carcinoma of the oropharynx (SCCOP) patients treated principally by radiation. The seven polymorphisms in the core NER genes (XPC-rs2228000, XPC-rs2228001, XPD-rs1799793, XPD-rs13181, XPG-rs17655, ERCC1-rs3212986 and XPA-rs1800975) were genotyped using PCR-RFLP method and log-rank test and multivariable Cox models were used to evaluate the associations in both dominant and recessive genetic models. In a dominant model, we found that polymorphisms of XPC-rs2228000, XPD-rs1799793 and XPG-rs17655 were significantly associated with disease-free survival (log-rank, p = 0.014; p = 0.00008; p = 0.0007, respectively), and these polymorphisms were significantly associated with recurrence risk of SCCOP (hazard ratio (HR) = 1.6, 95% confidence interval (CI) 1.1-2.3 for XPC-rs2228000; HR = 0.4, 95% 0.3-0.6 for XPD-rs1799793 and HR = 0.5, 95% CI 0.4-0.8 for XPG-rs17655) after multivariable adjustment. Moreover, the borderline significant or significant associations were also found for these three polymorphisms in HPV16/18-positive SCCOP patients (HR = 1.6, 95% CI 1.0-4.1 for XPC-rs2228000; HR = 0.2, 95% CI 0.1-0.5 for XPD-rs1799793 and HR = 0.1, 95% CI 0.0-0.9 for XPG-rs17655). However, similarly significant associations were not found for these polymorphisms in a recessive model. These findings suggest that polymorphisms of XPC-rs2228000, XPD-rs1799793 and XPG-rs17655 in the NER core genes may contribute to recurrence risk of SCCOP, particularly HPV-positive SCCOP, in a dominant but not in a recessive model. However, validation of these results is warranted.
Collapse
Affiliation(s)
- Xicheng Song
- Department of Head and Neck Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-4009, USA
| | | | | | | | | | | |
Collapse
|
33
|
Vaishampayan UN, Fontana J, Heilbrun LK, Smith D, Heath E, Dickow B, Figg WD. Phase II trial of bevacizumab and satraplatin in docetaxel-pretreated metastatic castrate-resistant prostate cancer. Urol Oncol 2013; 32:31.e25-33. [PMID: 23433892 DOI: 10.1016/j.urolonc.2012.11.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Revised: 11/21/2012] [Accepted: 11/21/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Satraplatin is an oral platinum compound that has demonstrated efficacy and tolerability in prostate cancer. Preclinical synergy between bevacizumab and platinum has been noted. METHODS Docetaxel-pretreated metastatic castrate-resistant prostate cancer patients with disease progression were eligible. Satraplatin 80 mg/m(2) orally on days 1 to 5, prednisone 5mg twice daily, and bevacizumab 10mg/kg on day 1, and 15 mg/kg on day 15 were administered in 35-day cycles. RESULTS Thirty one patients were enrolled. Grade 3 or 4 toxicities were pulmonary embolism in 2 patients and thrombocytopenia in 1 patient. 31% of the patients had a ≥ 30% decline in prostate-specific antigen. Median time to progression was 7.0 months (90% confidence interval [CI] 4.7-8.5mo) and median overall survival was 11.2 months (90% CI 9.1-16.4 mo). Polymorphism in the excision repair cross-complementation-1 (ERCC-1) gene was associated with time to progression (hazard ratio = 1.91). A circulating tumor cell count ≥ 5 was moderately prognostic of overall survival (hazard ratio = 1.49) as compared with CTC <5. CONCLUSIONS The combination was tolerable, and revealed promising efficacy in metastatic castrate-resistant prostate cancer. ERCC1 genotype maybe predictive of clinical benefit with platinum-based therapy in metastatic prostate cancer.
Collapse
Affiliation(s)
- Ulka N Vaishampayan
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI.
| | - Joseph Fontana
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Lance K Heilbrun
- Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Daryn Smith
- Biostatistics Core, Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - Elisabeth Heath
- Department of Oncology, Department of Medicine, Barbara Ann Karmanos Cancer Institute, Wayne State University, Detroit, MI
| | - Brenda Dickow
- Clinical Trials Office, Barbara Ann Karmanos Cancer Institute, Detroit, MI
| | - William D Figg
- Medical Oncology Branch, National Cancer Institute, Bethesda, MD
| |
Collapse
|
34
|
Zanoni A, Verlato G, Giacopuzzi S, Weindelmayer J, Casella F, Pasini F, Zhao E, de Manzoni G. Neoadjuvant concurrent chemoradiotherapy for locally advanced esophageal cancer in a single high-volume center. Ann Surg Oncol 2012; 20:1993-9. [PMID: 23274533 DOI: 10.1245/s10434-012-2822-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Indexed: 12/19/2022]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (CRT) is now considered the standard of care by many centers in the treatment of both squamous cell carcinoma (SCC) and adenocarcinoma of the esophagus. This study evaluates the effectiveness of a neoadjuvant CRT protocol, as regards pathological complete response (pCR) rate and long-term survival. METHODS From 2003 to 2011, at Upper G.I. Surgery Division of Verona University, 155 consecutive patients with locally advanced esophageal cancers (90 SCC, 65 adenocarcinoma) were treated with a single protocol of neoadjuvant CRT (docetaxel, cisplatin, and 5-fluorouracil with 50.4 Gy of concurrent radiotherapy). Response to CRT was evaluated through percentage of pathological complete response (pCR or ypT0N0), overall (OS) and disease-related survival (DRS), and pattern of relapse. RESULTS One hundred thirty-one patients (84.5 %) underwent surgery. Radical resection (R0) was achieved in 123 patients (79.3 %), and pCR in 65 (41.9 %). Postoperative mortality was 0.7 % (one case). Five-year OS and DRS were respectively 43 and 49 % in the entire cohort, 52 and 59 % in R0 cases, and 72 and 81 % in pCR cases. Survival did not significantly differ between SCC and adenocarcinoma, except for pCR cases. Forty-nine patients suffered from relapse, which was mainly systemic in adenocarcinoma. Only three out of 26 pCR patients with previous adenocarcinoma developed relapse, always systemic. CONCLUSIONS This study suggests that patients treated with the present protocol achieve good survival and high pCR rate. Further research is necessary to evaluate whether surgery on demand is feasible in selected patients, such as pCR patients with adenocarcinoma.
Collapse
Affiliation(s)
- A Zanoni
- Upper G.I. Surgery Division, University of Verona, Verona, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Ayers D, Nasti A. Utilisation of nanoparticle technology in cancer chemoresistance. JOURNAL OF DRUG DELIVERY 2012; 2012:265691. [PMID: 23213536 PMCID: PMC3505656 DOI: 10.1155/2012/265691] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Revised: 10/11/2012] [Accepted: 10/11/2012] [Indexed: 01/08/2023]
Abstract
The implementation of cytotoxic chemotherapeutic drugs in the fight against cancer has played an invariably essential role for minimizing the extent of tumour progression and/or metastases in the patient and thus allowing for longer event free survival periods following chemotherapy. However, such therapeutics are nonspecific and bring with them dose-dependent cumulative adverse effects which can severely exacerbate patient suffering. In addition, the emergence of innate and/or acquired chemoresistance to the exposed cytotoxic agents undoubtedly serves to thwart effective clinical efficacy of chemotherapy in the cancer patient. The advent of nanotechnology has led to the development of a myriad of nanoparticle-based strategies with the specific goal to overcome such therapeutic hurdles in multiple cancer conditions. This paper aims to provide a brief overview and recollection of all the latest advances in the last few years concerning the application of nanoparticle technology to enhance the safe and effective delivery of chemotherapeutic agents to the tumour site, together with providing possible solutions to circumvent cancer chemoresistance in the clinical setting.
Collapse
Affiliation(s)
- Duncan Ayers
- Department of Pathology, Faculty of Medicine & Surgery, University of Malta, Msida MSD 2060, Malta
| | - Alessandro Nasti
- School of Medicine, Kanazawa University Hospital, University of Kanazawa, Kanazawa 920-1192, Japan
| |
Collapse
|
36
|
Soo RA, Yong WP, Innocenti F. Systemic therapies for pancreatic cancer--the role of pharmacogenetics. Curr Drug Targets 2012; 13:811-28. [PMID: 22458528 DOI: 10.2174/138945012800564068] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 02/23/2012] [Accepted: 03/27/2012] [Indexed: 12/17/2022]
Abstract
Effective systemic treatment of pancreatic cancer remains a major challenge, with progress hampered by drug resistance and treatment related toxicities. Currently available cytotoxic agents as monotherapy or in combination have provided only a modest survival benefit for patients with advanced disease. Disappointing phase III results with gemcitabine-based combinations in patients with advanced pancreatic cancer might be related to poor efficacy of systemic therapies in unselected patients. Future research strategies should prioritize identification of predictive markers through pharmacogenetic investigations. The individualization of patient treatment through pharmacogenetics may help to improve outcome by maximizing efficacy whilst lowering toxicity. This review provides an update on the pharmacogenetics of pancreatic cancer treatment and its influence on treatment benefits and toxicity.
Collapse
Affiliation(s)
- Ross A Soo
- Department of Hematology-Oncology, National University Health System, Singapore
| | | | | |
Collapse
|
37
|
Soo RA, Yong WP, Innocenti F. Systemic therapies for pancreatic cancer--the role of pharmacogenetics. Curr Drug Targets 2012. [PMID: 22458528 DOI: 10.1016/j.pestbp.2011.02.012.investigations] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Effective systemic treatment of pancreatic cancer remains a major challenge, with progress hampered by drug resistance and treatment related toxicities. Currently available cytotoxic agents as monotherapy or in combination have provided only a modest survival benefit for patients with advanced disease. Disappointing phase III results with gemcitabine-based combinations in patients with advanced pancreatic cancer might be related to poor efficacy of systemic therapies in unselected patients. Future research strategies should prioritize identification of predictive markers through pharmacogenetic investigations. The individualization of patient treatment through pharmacogenetics may help to improve outcome by maximizing efficacy whilst lowering toxicity. This review provides an update on the pharmacogenetics of pancreatic cancer treatment and its influence on treatment benefits and toxicity.
Collapse
Affiliation(s)
- Ross A Soo
- Department of Hematology-Oncology, National University Health System, Singapore
| | | | | |
Collapse
|
38
|
Marin JJG, Briz O, Monte MJ, Blazquez AG, Macias RIR. Genetic variants in genes involved in mechanisms of chemoresistance to anticancer drugs. Curr Cancer Drug Targets 2012. [PMID: 22229248 DOI: 10.1002/9780470015902.a0025217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Refractoriness to the pharmacological treatment of cancer is dependent on the expression levels of genes involved in mechanisms of chemoresistance and on the existence of genetic variants that may affect their function. Thus, changes in genes encoding solute carriers may account for considerable inter-individual variability in drug uptake and the lack of sensitivity to the substrates of these transporters. Moreover, changes in proteins involved in drug export can affect their subcellular localization and transport ability and hence may also modify the bioavailability of antitumor agents. Regarding pro-drug activation or drug inactivation, genetic variants are responsible for changes in the activity of drug-metabolizing enzymes, which affect drug clearance and may determine the lack of response to anticancer chemotherapy. The presence of genetic variants may also decrease the sensitivity to pharmacological agents acting through molecular targets or signaling pathways. Recent investigations suggest that changes in genes involved in DNA repair may affect the response to platinum-based drugs. Since most anticancer agents activate cell death pathways, the evasion of apoptosis plays an important role in chemoresistance. Several genetic variants affecting death-receptor pathways, the mitochondrial pathway, downstream caspases and their natural modulators, and the p53 pathway, whose elements are mutated in more than half of tumors, and survival pathways, have been reported. The present review summarizes the available data regarding the role of genetic variants in the different mechanisms of chemoresistance and discusses their potential impact in clinical practice and in the development of tools to predict and overcome chemoresistance.
Collapse
Affiliation(s)
- J J G Marin
- Department of Physiology and Pharmacology, Campus Miguel de Unamuno E.I.D., Salamanca, Spain.
| | | | | | | | | |
Collapse
|
39
|
Pharmacogenetics of oxaliplatin as adjuvant treatment in colon carcinoma: are single nucleotide polymorphisms in GSTP1, ERCC1, and ERCC2 good predictive markers? Mol Diagn Ther 2012; 15:277-83. [PMID: 21958378 DOI: 10.1007/bf03256419] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Adjuvant chemotherapy improves survival in stage III colon cancer patients. However, a subgroup of patients still develops recurrent disease at some point in time, partly because of the ineffectiveness of the chemotherapy. Predictive markers of response are therefore crucial. Our aim was to study the predictive value of functional polymorphisms in genes involved in the metabolism of oxaliplatin and in DNA repair in stage III colon cancer patients. MATERIALS AND METHODS Normal DNA was isolated from 98 patients diagnosed with stage III colon carcinoma. Single nucleotide polymorphisms (SNPs) in three genes (the excision repair cross-complementing genes ERCC1 [19007T>C] and ERCC2 [2251A>C], and the glutathione S-transferase pi 1 gene [GSTP1 313A>G]) were tested by PCR followed by digestion with restriction enzymes or by direct sequencing. These genes and SNPs were selected on the basis of their reported associations with oxaliplatin response in colorectal cancer. RESULTS The genotype frequencies were in Hardy-Weinberg equilibrium. GSTP1 and ERCC2 polymorphisms were significantly associated with sex. The AA genotype of GSTP1 313A>G was more frequent in men than in women (59% vs 30%, p = 0.02), and the CC genotype of ERCC2 2251A>C was significantly more frequent in women than in men (24% vs 6%, p = 0.02). In univariate and multivariate survival analysis, none of the tested polymorphisms seemed to influence disease-free survival. The GSTP1 AA genotype had different effects on survival between men and women; homozygous A men had significantly worse cancer-specific survival and overall survival than women with the same genotype (log rank p = 0.029 and p = 0.015, respectively). CONCLUSION None of the tested polymorphisms is likely to be a reliable marker of response to oxaliplatin therapy. The GSTP1 313A>G homozygous A genotype may have a prognostic value in male patients.
Collapse
|
40
|
Fariña Sarasqueta A, van Lijnschoten G, Lemmens VEPP, Rutten HJT, van den Brule AJC. Pharmacogenetics of oxaliplatin as adjuvant treatment in colon carcinoma: are single nucleotide polymorphisms in GSTP1, ERCC1, and ERCC2 good predictive markers? Mol Diagn Ther 2012. [PMID: 21958378 DOI: 10.2165/11592080-000000000-00000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE Adjuvant chemotherapy improves survival in stage III colon cancer patients. However, a subgroup of patients still develops recurrent disease at some point in time, partly because of the ineffectiveness of the chemotherapy. Predictive markers of response are therefore crucial. Our aim was to study the predictive value of functional polymorphisms in genes involved in the metabolism of oxaliplatin and in DNA repair in stage III colon cancer patients. MATERIALS AND METHODS Normal DNA was isolated from 98 patients diagnosed with stage III colon carcinoma. Single nucleotide polymorphisms (SNPs) in three genes (the excision repair cross-complementing genes ERCC1 [19007T>C] and ERCC2 [2251A>C], and the glutathione S-transferase pi 1 gene [GSTP1 313A>G]) were tested by PCR followed by digestion with restriction enzymes or by direct sequencing. These genes and SNPs were selected on the basis of their reported associations with oxaliplatin response in colorectal cancer. RESULTS The genotype frequencies were in Hardy-Weinberg equilibrium. GSTP1 and ERCC2 polymorphisms were significantly associated with sex. The AA genotype of GSTP1 313A>G was more frequent in men than in women (59% vs 30%, p = 0.02), and the CC genotype of ERCC2 2251A>C was significantly more frequent in women than in men (24% vs 6%, p = 0.02). In univariate and multivariate survival analysis, none of the tested polymorphisms seemed to influence disease-free survival. The GSTP1 AA genotype had different effects on survival between men and women; homozygous A men had significantly worse cancer-specific survival and overall survival than women with the same genotype (log rank p = 0.029 and p = 0.015, respectively). CONCLUSION None of the tested polymorphisms is likely to be a reliable marker of response to oxaliplatin therapy. The GSTP1 313A>G homozygous A genotype may have a prognostic value in male patients.
Collapse
|
41
|
Erčulj N, Kovač V, Hmeljak J, Dolžan V. The influence of platinum pathway polymorphisms on the outcome in patients with malignant mesothelioma. Ann Oncol 2012; 23:961-7. [DOI: 10.1093/annonc/mdr324] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
42
|
|
43
|
Neoadjuvant radiochemotherapy in adenocarcinoma of the esophagus: ERCC1 gene polymorphisms for prediction of response and prognosis. J Gastrointest Surg 2012; 16:26-34; discussion 34. [PMID: 21956434 DOI: 10.1007/s11605-011-1700-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2011] [Accepted: 09/14/2011] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Neoadjuvant radiochemotherapy (RT/CTx) regimens were primarily designed for treatment of squamous cell carcinoma of the esophagus. Own preliminary results demonstrate that also patients with locally advanced adenocarcinoma of the esophagus may achieve a major response in 30% with a 3-year survival rate of 80%. To identify these patients, ERCC1 (rs11615) gene polymorphisms were analyzed. ERCC1 is a key enzyme of the nucleotide excision and repair (NER) complex to prevent DNA inter- and intra-strand crosslinks. PATIENTS AND METHODS Genomic DNA from 217 patients with cT3/4 adenocarcinoma of the esophagus was extracted from paraffin-embedded tissues. Of these patients, 153 underwent neoadjuvant RT/CTx (CDDP, 5-FU, 36 Gy). For analysis of ERCC1 single nucleotide polymorphisms (SNPs), allelic discrimination was performed by quantitative real-time PCR. Two allele-specific TaqMan probes in competition were used for amplification of ERCC1 (rs11615). Allelic genotyping was correlated with histomorphologic tumor regression after neoadjuvant RT/CTx and survival. Major response (MaHR) was defined as <10% vital residual tumor cells (VRTC). RESULTS Analysis of tumor regression revealed a MaHR in 56/153 (36.6%) patients with a 5-year survival rate (5-YSR) of 74% (p < 0.001). ERCC1 gene polymorphisms for all patients showed the following expression pattern: ERCC1 polymorphism (rs11615) CC: n = 27 (12.4%), TT: n = 98 (45.2%), C/T: n = 92 (42.4%). ERCC1 polymorphism CT was identified as a predictor for response to the neoadjuvant RT/CTx (p < 0.001). The 5-YSR for patients with C/T genotype was 51%. Contrary to this, the 5-YSR for the group of patients with a CC/TT polymorphism decreased to 34%. CONCLUSION Analysis of ERCC1 (rs11615) gene polymorphisms reveals a significant correlation with response and survival in patients with adenocarcinoma of the esophagus treated with a neoadjuvant radiochemotherapy. Single nucleotide polymorphisms of ERCC1 (rs11615) could therefore be applied to further individualize therapy in esophageal cancer.
Collapse
|
44
|
ERCC1 Gene +262A/C Polymorphism Associated with Risk of Gastric Cardiac Adenocarcinoma in Nonsmokers. Arch Med Res 2012; 43:67-74. [DOI: 10.1016/j.arcmed.2012.01.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 01/18/2012] [Indexed: 11/22/2022]
|
45
|
Tzvetkov MV, Behrens G, O'Brien VP, Hohloch K, Brockmöller J, Benöhr P. Pharmacogenetic analyses of cisplatin-induced nephrotoxicity indicate a renoprotective effect of ERCC1 polymorphisms. Pharmacogenomics 2011; 12:1417-27. [PMID: 21902499 DOI: 10.2217/pgs.11.93] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
AIM We investigated whether genetic polymorphisms may contribute to the interpatient variability of cisplatin-induced nephrotoxicity. PATIENTS & METHODS Polymorphisms in the candidate genes GSTM1, GSTT1, OCT1, OCT2, LARP2, ERCC1, XRCC1 and EPO were analyzed for associations with nephrotoxicity in 79 cancer patients receiving cisplatin-containing chemotherapy. RESULTS Higher cisplatin dose was associated with strongly decreased estimated glomerular filtration rates (eGFR) (r(2) = 0.205). Two highly genetically linked polymorphisms in the ERCC1 gene, 8092C>A and Asn118Asn, were significantly associated with change in eGFR, accounting for an additional 13% of interindividual variability. Homozygous carriers of the 8092A allele in ERCC1 showed no reduction in eGFR, compared with the 11.5% mean eGFR decrease in C allele carriers (p = 0.004). Homozygous carriers of the C allele of Asn118Asn showed no reduction in eGFR, compared with the 12.8% mean eGFR decrease seen in T allele carriers (p = 0.047). Polymorphisms in the other candidate genes were not associated with cisplatin-induced nephrotoxicity. CONCLUSION Genetic polymorphisms in ERCC1 may be valuable predictors of cisplatin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Mladen V Tzvetkov
- Department of Clinical Pharmacology, University Medical Center, Georg-August-University, Robert-Koch-Strasse 40, Göttingen, Germany.
| | | | | | | | | | | |
Collapse
|
46
|
Zhong S, Nukui T, Buch S, Diergaarde B, Weissfeld LA, Grandis J, Romkes M, Weissfeld JL. Effects of ERCC2 Lys751Gln (A35931C) and CCND1 (G870A) polymorphism on outcome of advanced-stage squamous cell carcinoma of the head and neck are treatment dependent. Cancer Epidemiol Biomarkers Prev 2011; 20:2429-37. [PMID: 21890746 DOI: 10.1158/1055-9965.epi-11-0520] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Germline variation in DNA damage response may explain variable treatment outcomes in squamous cell carcinoma of the head and neck (SCCHN). By grouping patients according to stage and radiation treatment, we compared SCCHN survival with regard to ERCC2 A35931C (Lys751Gln, rs13181) and CCND1 G870A (Pro241Pro, rs9344) genotypes. METHODS In a hospital-based SCCHN case series (all white, 24.7% female, mean age 58.4 years), this treatment-outcome cohort study genotyped 275 stage III-IV cases that were initially treated with radiation (with or without chemotherapy) and 80 stage III-IV and 130 stage I-II cases that were initially treated without radiation or chemotherapy and used Kaplan-Meier and Cox regression analyses to compare genotype groups on the basis of overall, disease-specific, progression-free, and recurrence-free survival rates. RESULTS ERCC2 35931 AA predicted worse survival in stage III-IV cases treated with radiation [multiply-adjusted HR = 1.66, 95% confidence interval (CI), 1.15-2.40; HR over the first 3 follow-up years = 1.92; 95% CI, 1.28-2.88] and better survival in stage III-IV cases not treated with radiation (HR = 0.26; 95% CI, 0.11-0.62). Although not associated with survival in stage III-IV cancers treated with radiation (HR = 1.00; 95% CI, 0.67-1.51), CCND1-870 GG predicted better survival in stage III-IV cancers not treated with radiation (HR = 0.14; 95% CI, 0.04-0.50). Survival in stage I-II did not depend on ERCC2 A35931C or CCND1 G870A genotype. CONCLUSIONS Although promoting tumor progression in untreated patients, germline differences in DNA-repair or cell-cycle control may improve treatment outcome in patients treated with DNA-damaging agents. IMPACT ERCC2 A35931C may help distinguish advanced stage SCCHN with better outcomes from radiation treatment.
Collapse
Affiliation(s)
- Shilong Zhong
- University of Pittsburgh Cancer Institute with Center for Clinical Pharmacology, Pittsburgh, PA, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Windsor RE, Strauss SJ, Kallis C, Wood NE, Whelan JS. Germline genetic polymorphisms may influence chemotherapy response and disease outcome in osteosarcoma: a pilot study. Cancer 2011; 118:1856-67. [PMID: 21887680 DOI: 10.1002/cncr.26472] [Citation(s) in RCA: 108] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Revised: 06/23/2011] [Accepted: 07/06/2011] [Indexed: 11/09/2022]
Abstract
BACKGROUND Osteosarcoma is the most common malignant bone tumor in children and young people. Efficacy of multiagent MAP (methotrexate, doxorubicin [Adriamycin], cisplatin) chemotherapy may be influenced by multiple cellular pathways. This pilot study aimed to investigate the association of 36 candidate genetic polymorphisms in MAP pathway genes with histological response, survival, and grade 3-4 chemotherapy toxicity in osteosarcoma. METHODS Blood samples were obtained from 60 patients who had completed MAP chemotherapy. All patients were manually genotyped for 5 polymorphisms. The remaining 31 polymorphisms were genotyped in 50 patients using the Illumina 610-Quad microarray. Associations between candidate polymorphisms and histological response, progression-free survival, and toxicity were estimated using Pearson chi-square and Fisher exact tests, the Kaplan-Meier method, the log-rank test, and the Cox proportional hazards model. RESULTS Poor histological response was increased in variants of ABCC2 c.24C>T (P = .011) and GSTP1 c.313A>G p.Ile(105)Val (P = .009), whereas MTHFD1 c.1958G>A p.Arg(653)Gln was protective (P = .03). Methotrexate toxicity was increased in variants of MTHFR c.1298A>C p.Glu(429)Ala (P = .038), ABCB1 c.3435T>C Ile(145)Ile (P = .027), and ABCC2 c.3563T>A p.Val(1188)Glu (P = .028). Variants of GSTP1 c.313A>G p.Ile(105)Val were at increased risk of myelosuppression (P = .024) and cardiac damage (P = .008). CONCLUSIONS This pilot study represents the most comprehensive study to date examining the role of genetic polymorphisms in osteosarcoma. Although small and retrospective, it shows that several polymorphisms appear to significantly influence toxicity and clinical outcome. These deserve prospective validation in the hope of optimizing treatment for resistant disease and reducing the late effects burden.
Collapse
|
48
|
Biason P, Hattinger CM, Innocenti F, Talamini R, Alberghini M, Scotlandi K, Zanusso C, Serra M, Toffoli G. Nucleotide excision repair gene variants and association with survival in osteosarcoma patients treated with neoadjuvant chemotherapy. THE PHARMACOGENOMICS JOURNAL 2011; 12:476-83. [PMID: 21826087 DOI: 10.1038/tpj.2011.33] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The aim of this study was to investigate the role of common polymorphisms in the nucleotide excision repair pathway genes in the tumorigenesis of osteosarcoma and in the response to DNA damaging therapies, such as cisplatin-based neoadjuvant therapy. Excision repair cross-complementing (ERCC) group 2 (XPD; rs13181 and rs1799793), group 5 (XPG; rs17655) and group 1 (XPA; rs3212986 and rs11615) polymorphisms were analyzed in a group of 130 homogenously treated patients with high-grade osteosarcoma, for association with event-free survival (EFS), using the Kaplan-Meier plots and log-rank test. A positive association was observed between both XPD single-nucleotide polymorphisms and an increased EFS (hazards ratio (HR) = 0.34, 95% confidence interval (CI) 0.12-0.98 and HR = 0.19, 95% CI 0.05-0.77, respectively). We had also performed a case-control study for relative risk to develop osteosarcoma. Patients carrying at least one variant allele of XPD rs1799793 had a reduced risk of developing osteosarcoma, compared with wild-type patients (odds ratio = 0.55, 95% CI 0.36-0.84). This study suggests that XPD rs1799793 could be a marker of osteosarcoma associated with features conferring either a better prognosis or a better outcome after platinum therapy, or both.
Collapse
Affiliation(s)
- P Biason
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico, National Cancer Institute, Aviano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
OBJECTIVE To investigate the association of the genetic variants in excision repair cross-complementation group 2 (ERCC2) R156R and ERCC4 rs3136038 with survival duration for patients with esophageal cancer. BACKGROUND ERCC2 and ERCC4 are important molecules participating nucleotide excision repair system. The clinical relevance of the genetic variants of these genes is largely unknown currently. PATIENTS AND METHODS A total of 400 patients with a diagnosis of esophageal cancer were included. The genetic variants in the promoter regions of ERCC2 on R156R and ERCC4 on rs3136038 were analyzed with the TaqMan assay from leukocyte DNA collected before treatment and correlated to survival of the patients. RESULTS Presence with ERCC2 R156R C/C or ERCC4 rs3136038 C/T genotype of the patients could additively increase risk of death and disease progression. Under multivariate analysis, T, N staging and simultaneous presentation of these unfavorable genotypes were found significant for prognosis (P < 0.05). Accumulation of each unfavorable genotype would associate with adjusted HRs [95% CI] of 1.35 [1.10-1.65] and 1.37 [1.12-1.68] (P ≤ 0.05) for death and disease progression respectively. The prognostic impact of these genotypes were more evident in the subgroup of patients with early disease status including T staging (II or less), free from lymph node metastasis or being able to undergo surgical resection (P < 0.05 for both overall and disease progression-free survival duration, respectively). CONCLUSION Genetic variants in ERCC2 and ERCC4 may provide further survival prediction in addition to TNM staging system of esophageal cancer, which is more evident in the patients with early disease status.
Collapse
|
50
|
Earley JN, Turchi JJ. Interrogation of nucleotide excision repair capacity: impact on platinum-based cancer therapy. Antioxid Redox Signal 2011; 14:2465-77. [PMID: 20812782 PMCID: PMC3096502 DOI: 10.1089/ars.2010.3369] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
DNA repair is essential for routine monitoring and repair of damage imparted to our genetic material by exposure to endogenous and exogenous carcinogens, including reactive oxygen species, UV light, and chemicals such as those found in cigarette smoke. Without DNA repair pathways, the continual assault on our DNA would be highly mutagenic and the risk of cancer increased. Paradoxically, the same pathways that help prevent cancer development are detrimental to the efficacy of DNA-damaging cancer therapeutics such as cisplatin. Recent studies demonstrate the inverse relationship between DNA repair capacity and efficacy of platinum-based chemotherapeutics: increased DNA repair capacity leads to resistance, while decreased capacity leads to increased sensitivities. Cisplatin's cytotoxic effects are mediated by formation of intrastrand DNA crosslinks, which are predominantly repaired via the nucleotide excision repair (NER) pathway. In an effort to personalize the treatment of cancers based on DNA repair capacity, we developed an ELISA-based assay to measure NER activity accurately and reproducibly as a prognostic for platinum-based treatments. Here we present an overview of DNA repair and its link to cancer and therapeutics. We also present data demonstrating the ability to detect the proteins of the pre-incision complex within the NER pathway from cell and tissue extracts.
Collapse
Affiliation(s)
- Jennifer N Earley
- Department of Medicine/Hematology and Oncology, Indiana University School of Medicine, 980 W. Walnut Street, Indianapolis, IN 46202, USA
| | | |
Collapse
|