1
|
Patel JN, Jiang C, Owzar K, Mulkey F, Luzum JA, Mamon HJ, Haller DG, Dragovich T, Alberts SR, Bjarnason G, Willet CG, Niedzwiecki D, Enzinger P, Ratain MJ, Fuchs C, McLeod HL. Pharmacogenetic study in gastric cancer patients treated with adjuvant fluorouracil/leucovorin or epirubicin/cisplatin/fluorouracil before and after chemoradiation on CALGB 80101 (Alliance). Pharmacogenet Genomics 2021; 31:215-220. [PMID: 34149004 PMCID: PMC8490297 DOI: 10.1097/fpc.0000000000000442] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
There is a lack of pharmacogenetic predictors of outcome in gastric cancer patients. The aim of this study was to assess previously identified candidate genes associated with 5-fluorouracil (5-FU), cisplatin, or epirubicin toxicity or response in a cohort of resected gastric cancer patients treated on CALGB (Alliance) 80101. Gastric or gastroesophageal cancer patients randomized to adjuvant 5-FU/leucovorin or epirubicin/cisplatin/5-FU before and after 5-FU chemoradiation were genotyped for single nucleotide polymorphisms (SNPs) in GSTP1 (rs1695), ERCC1 (rs11615 and rs3212986), XRCC1 (rs25487), UGT2B7 (rs7439366) and the 28 base-pair tandem repeats in TYMS (rs34743033). Logistic regression and log rank tests were used to assess the association between each SNP and incidence of grade 3/4 neutropenia and leukopenia, overall (OS) and progression-free survival (PFS), respectively. Toxicity endpoint analyses were adjusted for the treatment arm, while OS and PFS were also adjusted for performance status, sex, age, lymph node involvement, and primary tumor site and size. Of 281 subjects with successful genotyping results and available clinical (toxicity and efficacy) data, 166 self-reported non-Hispanic White patients were included in the final analysis. There was a lack of evidence of an association among any SNPs tested with grade 3/4 neutropenia and leukopenia or OS and PFS. Age, lymph node involvement, and primary tumor size were significantly associated with OS and PFS. This study failed to confirm results of previous gastric cancer pharmacogenetic studies.
Collapse
Affiliation(s)
- Jai N. Patel
- Levine Cancer Institute, Atrium Health, Charlotte, NC, USA
| | - Chen Jiang
- Alliance Statistics and Data Center, Duke University, Durham, NC, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | - Flora Mulkey
- Alliance Statistics and Data Center, Duke University, Durham, NC, USA
| | | | | | - Daniel G. Haller
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Georg Bjarnason
- Sunnybrook Odette Cancer Centre, University of Toronto, Toronto, ON
| | - Christopher G. Willet
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC, USA
| | - Donna Niedzwiecki
- Department of Biostatistics and Bioinformatics, Duke University, Durham, NC, USA
| | | | | | - Charles Fuchs
- Smilow Cancer Hospital, Yale University, New Haven, CT, USA
| | - Howard L. McLeod
- USF Taneja College of Pharmacy and the Geriatric Oncology Consortium, Tampa, FL, USA
| |
Collapse
|
2
|
Cordova-Delgado M, Bravo ML, Cumsille E, Hill CN, Muñoz-Medel M, Pinto MP, Retamal IN, Lavanderos MA, Miquel JF, Rodriguez-Fernandez M, Liao Y, Li Z, Corvalán AH, Armisén R, Garrido M, Quiñones LA, Owen GI. A case-control study of a combination of single nucleotide polymorphisms and clinical parameters to predict clinically relevant toxicity associated with fluoropyrimidine and platinum-based chemotherapy in gastric cancer. BMC Cancer 2021; 21:1030. [PMID: 34525956 PMCID: PMC8444616 DOI: 10.1186/s12885-021-08745-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/22/2021] [Indexed: 12/22/2022] Open
Abstract
Background Fluoropyrimidine plus platinum chemotherapy remains the standard first line treatment for gastric cancer (GC). Guidelines exist for the clinical interpretation of four DPYD genotypes related to severe fluoropyrimidine toxicity within European populations. However, the frequency of these single nucleotide polymorphisms (SNPs) in the Latin American population is low (< 0.7%). No guidelines have been development for platinum. Herein, we present association between clinical factors and common SNPs in the development of grade 3–4 toxicity. Methods Retrospectively, 224 clinical records of GC patient were screened, of which 93 patients were incorporated into the study. Eleven SNPs with minor allelic frequency above 5% in GSTP1, ERCC2, ERCC1, TP53, UMPS, SHMT1, MTHFR, ABCC2 and DPYD were assessed. Association between patient clinical characteristics and toxicity was estimated using logistic regression models and classification algorithms. Results Reported grade ≤ 2 and 3–4 toxicities were 64.6% (61/93) and 34.4% (32/93) respectively. Selected DPYD SNPs were associated with higher toxicity (rs1801265; OR = 4.20; 95% CI = 1.70–10.95, p = 0.002), while others displayed a trend towards lower toxicity (rs1801159; OR = 0.45; 95% CI = 0.19–1.08; p = 0.071). Combination of paired SNPs demonstrated significant associations in DPYD (rs1801265), UMPS (rs1801019), ABCC2 (rs717620) and SHMT1 (rs1979277). Using multivariate logistic regression that combined age, sex, peri-operative chemotherapy, 5-FU regimen, the binary combination of the SNPs DPYD (rs1801265) + ABCC2 (rs717620), and DPYD (rs1801159) displayed the best predictive performance. A nomogram was constructed to assess the risk of developing overall toxicity. Conclusion Pending further validation, this model could predict chemotherapy associated toxicity and improve GC patient quality of life. Supplementary Information The online version contains supplementary material available at 10.1186/s12885-021-08745-0.
Collapse
Affiliation(s)
- Miguel Cordova-Delgado
- Faculty of Chemical and Pharmaceutical Sciences, Universidad de Chile, 8380494, Santiago, Chile.,Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.,Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile
| | - María Loreto Bravo
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile
| | - Elisa Cumsille
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile
| | - Charlotte N Hill
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, 8331150, Santiago, Chile
| | - Matías Muñoz-Medel
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile
| | - Mauricio P Pinto
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile
| | - Ignacio N Retamal
- Faculty of Dentistry, Universidad de Los Andes, 7620001, Santiago, Chile
| | - María A Lavanderos
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, 8380494, Santiago, Chile.,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain.,Escuela de Química y Farmacia, Facultad de Ciencias Médicas, Universidad Bernardo O'Higgins, Santiago, Chile
| | - Juan Francisco Miquel
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile
| | - Maria Rodriguez-Fernandez
- Institute for Biological and Medical Engineering, Schools of Engineering, Medicine and Biological Sciences, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Yuwei Liao
- Central Laboratory, Yangjiang People's Hospital, GuangDong Province, Yangjiang, China.,Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China
| | - Zhiguang Li
- Center of Genome and Personalized Medicine, Institute of Cancer Stem Cell, Dalian Medical University, Dalian, China.,National Institute on Aging, National Institute of Health, Baltimore, USA
| | - Alejandro H Corvalán
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), 8330034, Santiago, Chile
| | - Ricardo Armisén
- Instituto de Ciencias e Innovación en Medicina, Facultad de Medicina, Clínica Alemana, Universidad del Desarrollo, 7590943, Santiago, Chile
| | - Marcelo Garrido
- Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile
| | - Luis A Quiñones
- Laboratory of Chemical Carcinogenesis and Pharmacogenetics, Department of Basic and Clinical Oncology, Faculty of Medicine, Universidad de Chile, 8380494, Santiago, Chile. .,Latin American Network for Implementation and Validation of Clinical Pharmacogenomics Guidelines (RELIVAF-CYTED), Madrid, Spain.
| | - Gareth I Owen
- Department of Physiology, Faculty of Biological Sciences, Pontificia Universidad Católica de Chile, 8331150, Santiago, Chile. .,Department of Hematology and Oncology, Faculty of Medicine, Pontificia Universidad Católica de Chile, 8330032, Santiago, Chile. .,Millennium Institute on Immunology and Immunotherapy, 8331150, Santiago, Chile. .,Advanced Center for Chronic Diseases (ACCDiS), 8330034, Santiago, Chile.
| |
Collapse
|
3
|
Mirzaev KB, Fedorinov DS, Akmalova KA, Abdullaev SP, Kachanova AA, Sozaeva ZA, Grishina EA, Shuev GN, Kitaeva EY, Shprakh VV, Suleymanov SS, Bolieva LZ, Sozaeva MS, Zhuchkova SM, Gimaldinova NE, Sidukova EE, Burashnikova IS, Shikaleva AA, Zabudskaya KG, Sychev DA. [Analysis of carrying clinically significant allelic variants of TPMT and DPYD genes associated with the response to drug therapy in cancer practice among 9 ethnic groups of the Russian Federation]. TERAPEVT ARKH 2020; 92:43-51. [PMID: 33346461 DOI: 10.26442/00403660.2020.08.000719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 11/22/2022]
Abstract
AIM To study the peculiarities of carrying clinically significant allelic variants of TPMT and DPYD genes associated with the response to drug therapy in cancer practice among 9 ethnic groups of the Russian Federation. MATERIALS AND METHODS The study included 1446 conditionally healthy volunteers from 9 ethnic groups. Carriage of polymorphic TPMT and DPYD gene markers was detected by the Real-Time PCR (polymerase chain reaction) method. RESULTS In all ethnic groups, the distribution of genotypes and alleles matched the equilibrium of Hardy-Weinberg. TPMT*3A (rs1800460) and TPMT*3C (rs1142345) were observed in heterozygous state in all investigated ethnic groups. In the Kabardinian group (n=204) the frequency of the TPMT*3A minor allele (MAF, %) was 2.94%; Balkars (n=200) 1.25%; Ossetians (n=239) 1.67%; Chuvashes (n=238) 1.89%: Mari (n=206) 1.21%; Tatars (n=141) 1.77%; Russians (n=134) 4.85%. The frequency of the TPMT*3C minor allele (MAF, %) in the Kabardinian group (n=204) MAF was 4.90%; Balkars (n=200) 1. 75%; Buryats (n=114) 0.44%; Ossetians (n=239) 1.88%; Chuvashes (n=238) 1.68%: Mari (n=206) 1.21%; Tatars (n=141) 1.42%; Russians (n=134) 4.48%. The results of the analysis of DPYD*2A polymorphism (rs3918290) demonstrated ethnic peculiarities of distribution. In the heterozygous state it was found only in the groups of Kabardins (n=204, MAF 1.22%), Balkars (n=200, MAF 2.00%), and Ossetians (n=239, MAF 0.63%). CONCLUSION The results obtained in the study will be useful for developing personalized algorithms of antitumor therapy in cancer practice, including those aimed at increasing the safety of chemotherapy.
Collapse
Affiliation(s)
- K B Mirzaev
- Russian Medical Academy of Continuous Professional Education
| | - D S Fedorinov
- Russian Medical Academy of Continuous Professional Education
| | - K A Akmalova
- Russian Medical Academy of Continuous Professional Education
| | - S P Abdullaev
- Russian Medical Academy of Continuous Professional Education
| | - A A Kachanova
- Russian Medical Academy of Continuous Professional Education
| | - Z A Sozaeva
- Russian Medical Academy of Continuous Professional Education
| | - E A Grishina
- Russian Medical Academy of Continuous Professional Education
| | - G N Shuev
- Russian Medical Academy of Continuous Professional Education
| | - E Y Kitaeva
- Irkutsk State Medical Academy of Postgraduate Education - branch of Russian Medical Academy of Continuous Professional Education
| | - V V Shprakh
- Irkutsk State Medical Academy of Postgraduate Education - branch of Russian Medical Academy of Continuous Professional Education
| | | | | | | | | | | | | | - I S Burashnikova
- Kazan State Medical Academy - branch of Russian Medical Academy of Continuous Professional Education
| | - A A Shikaleva
- Kazan State Medical Academy - branch of Russian Medical Academy of Continuous Professional Education
| | | | - D A Sychev
- Russian Medical Academy of Continuous Professional Education
| |
Collapse
|
4
|
Smyth EC, Cafferkey C, Loehr A, Waddell T, Begum R, Peckitt C, Harding TC, Nguyen M, Okines AF, Raponi M, Rao S, Watkins D, Starling N, Middleton GW, Wadsley J, Mansoor W, Crosby T, Wotherspoon A, Chau I, Cunningham D. Genomic loss of heterozygosity and survival in the REAL3 trial. Oncotarget 2018; 9:36654-36665. [PMID: 30613349 PMCID: PMC6291175 DOI: 10.18632/oncotarget.26336] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 10/06/2018] [Indexed: 12/13/2022] Open
Abstract
Background Homologous recombination deficiency (HRD) measured using a genomic signature for loss of heterozygosity (LOH) predicts benefit from rucaparib in ovarian cancer. We hypothesized that some oesophagogastric cancers will have high-LOH which would be prognostic in patients treated with platinum chemotherapy. Methods Diagnostic biopsy DNA from patients treated in the REAL3 trial was sequenced using the Foundation Medicine T5 next-generation sequencing (NGS) assay. An algorithm quantified the percentage of interrogable genome with LOH. Multidimensional optimization was performed to identify a cut-off dichotomizing the population into LOH-high and low groups associated with differential survival outcomes. Results Of 158 available samples, 117 were successfully sequenced; LOH was derived for 74 of these. A cut-off of 21% genomic LOH defined an LOH-high subgroup (n=10, 14% of population) who had median overall survival (OS) of 18.3 months (m) versus 11m for the LOH-low group (HR 0.55 95% CI 0.19-0.97, p= 0.10). Progression free survival (PFS) for LOH-high and LOH-low groups was 10.7m and 7.3m (HR 0.61 (95% CI 0.21 – 1.09, p=0.09). Sensitivity analysis censoring operated patients (n=4), demonstrated OS of 18.3m vs. 10.2m (HR 0.43, 95% CI (0.20-0.92), p=0.02; PFS was 10.5m vs. 7.2m (HR 0.55, (95% CI 0.26-1.17), p=0.09 for LOH-high and LOH-low. Conclusion HRD assessment using an algorithmically derived LOH signature on a standard NGS panel identifies oesophagogastric cancer patients with high LOH who have prolonged survival when treated with platinum chemotherapy. Validation work will determine the signature's predictive value in patients treated with a PARP inhibitor and with platinum chemotherapy.
Collapse
Affiliation(s)
- Elizabeth C Smyth
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom.,Current affiliation: Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Catherine Cafferkey
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Andrea Loehr
- Clovis Oncology, San Francisco, CA, United States of America
| | - Tom Waddell
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom.,Current affiliation: Department of Medical Oncology, Christie Hospital, Manchester, United Kingdom
| | - Ruwaida Begum
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Clare Peckitt
- Department of Clinical Research & Development, Royal Marsden Hospital, London & Sutton, United Kingdom
| | | | - Minh Nguyen
- Clovis Oncology, San Francisco, CA, United States of America
| | - Alicia F Okines
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Mitch Raponi
- Clovis Oncology, San Francisco, CA, United States of America
| | - Sheela Rao
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - David Watkins
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Naureen Starling
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - Gary W Middleton
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| | - Jonathan Wadsley
- Department of Medical Oncology, Weston Park Hospital, Sheffield, United Kingdom
| | - Wasat Mansoor
- Current affiliation: Department of Medical Oncology, Christie Hospital, Manchester, United Kingdom
| | - Tom Crosby
- Department of Clinical Oncology, Velindre Hospital, Cardiff, Wales, United Kingdom
| | - Andrew Wotherspoon
- Department of Histopathology, Royal Marsden Hospital, London & Surrey, United Kingdom
| | - Ian Chau
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| | - David Cunningham
- Department of Gastrointestinal Oncology and Lymphoma, Royal Marsden Hospital, London & Sutton, United Kingdom
| |
Collapse
|
5
|
Patel JN, Villadolid J. Cancer Drug Delivery. PHARMACEUTICAL SCIENCES 2017. [DOI: 10.4018/978-1-5225-1762-7.ch008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Advancements in cancer drug delivery have led to the development of personalized oncology care through molecularly-driven targeted therapies. Understanding molecular and cellular mechanisms which drive tumor progression and resistance is critical in managing new treatment strategies which have shifted from empiric to biomarker-directed therapy selection. Biomarker-directed therapies have improved clinical outcomes in multiple malignancies as monotherapy and in combination with other treatment modalities, however the changing scope of treatment options presents new opportunities and challenges for research. Furthermore, pharmacogenetics may provide a rationale method of personalizing anticancer drug dosing and supportive care management for oncology patients. This chapter reviews biomarker classifications and pharmacogenetics in anticancer therapy and supportive care. Examples of biomarker-directed therapies and clinical assays, in addition to future directions of molecular profiling in oncology therapy management are discussed.
Collapse
|
6
|
Meulendijks D, Rozeman EA, Cats A, Sikorska K, Joerger M, Deenen MJ, Beijnen JH, Schellens JHM. Pharmacogenetic variants associated with outcome in patients with advanced gastric cancer treated with fluoropyrimidine and platinum-based triplet combinations: a pooled analysis of three prospective studies. THE PHARMACOGENOMICS JOURNAL 2016; 17:441-451. [DOI: 10.1038/tpj.2016.81] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Revised: 07/22/2016] [Accepted: 08/25/2016] [Indexed: 01/08/2023]
|
7
|
Patel JN. Cancer pharmacogenomics, challenges in implementation, and patient-focused perspectives. Pharmgenomics Pers Med 2016; 9:65-77. [PMID: 27471406 PMCID: PMC4948716 DOI: 10.2147/pgpm.s62918] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cancer pharmacogenomics is an evolving landscape and has the potential to significantly impact cancer care and precision medicine. Harnessing and understanding the genetic code of both the patient (germline) and the tumor (somatic) provides the opportunity for personalized dose and therapy selection for cancer patients. While germline DNA is useful in understanding the pharmacokinetic and pharmacodynamic disposition of a drug, somatic DNA is particularly useful in identifying drug targets and predicting drug response. Molecular profiling of somatic DNA has resulted in the current breadth of targeted therapies available, expanding the armamentarium to battle cancer. This review provides an update on cancer pharmacogenomics and genomics-based medicine, challenges in applying pharmacogenomics to the clinical setting, and patient perspectives on the use of pharmacogenomics to personalize cancer therapy.
Collapse
Affiliation(s)
- Jai N Patel
- Department of Cancer Pharmacology, Levine Cancer Institute, Carolinas HealthCare System, Charlotte, NC, USA
| |
Collapse
|
8
|
Patel JN, Fong MK. Personalizing fluoropyrimidine administration in colorectal cancer patients. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2016. [DOI: 10.1080/23808993.2016.1176860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
9
|
Abstract
The goal of pharmacogenomic research is to discover and validate genetic variants that are predictive of drug response, for eventual implementation into clinical practice. Cancer pharmacogenomics provides the opportunity to analyze two sets of DNA, that of the tumor (somatic) and that of the host (germline). Germline variants are inherited variations and are often associated with the pharmacokinetic behavior of a drug, including drug disposition and ultimately drug efficacy and/or toxicity, whereas somatic mutations are often useful in predicting the pharmacodynamic response to drugs. Pharmacoethnicity, or ethnic diversity in drug response or toxicity, is an increasingly recognized factor accounting for interindividual variations in anticancer drug response. Pharmacoethnicity is often determined by germline pharmacogenomic factors and the distribution of single nucleotide polymorphisms across various populations, but it may also be influenced by nongenetic factors, such as environmental factors. This review aims to elucidate the importance of pharmacoethnicity in cancer pharmacogenomic research and implementation, focusing solely on germline variants.
Collapse
|
10
|
Rui LX, Shu SY, Jun WJ, Mo CZ, Wu SZ, Min LS, Yuan L, Yong PJ, Cheng SZ, Sheng WS, Yao TZ. The dual induction of apoptosis and autophagy by SZC014, a synthetic oleanolic acid derivative, in gastric cancer cells via NF-κB pathway. Tumour Biol 2015; 37:5133-44. [PMID: 26547583 DOI: 10.1007/s13277-015-4293-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 10/20/2015] [Indexed: 12/16/2022] Open
Abstract
Oleanolic acid (OA) possesses various pharmacological activities, such as antitumor and anti-inflammation; however, its clinical applications are limited by its relatively weak activities and low bioavailability. In this study, we evaluated the cytotoxic activity of seven novel OA derivatives, one of which, SZC014 [2-(pyrrolidine-1-yl) methyl-3-oxo-olean-12-en-28-oic acid], exhibited the strongest antitumor activity; its anticancer effect on gastric cancer cells and action mechanisms were investigated. The viability of OA and seven synthesized derivatives treating gastric cancer cells was detected using tetrazolium (MTT). Among them, SZC014 exhibited the strongest cytotoxic activity against gastric cancer cells (SGC7901, MGC803, and MKN-45). The effect of SZC014 on cell cycle was identified by propidium iodide (PI) staining assay. The cellular apoptosis induced by SZC014 was tested by annexin V/PI. The cellular morphological changes and ultrastructural structures affected by SZC014 were observed and imaged through inverted phase contrast microscope and transmission electron microscopy. Western blotting was performed to explore the expression of proteins associated with apoptosis (caspase 3, caspase 9, Bax, Bcl-2, and Bcl-xL), autophagy (Beclin 1 and ATG 5), and nuclear factor-κB (NF-κB) signal pathway, respectively. The cytotoxic activities of all the seven synthesized OA derivatives were stronger than that of OA against gastric cancer cells. SZC014 exhibited stronger cytotoxic activity than other OA derivatives, inhibited the proliferation of gastric cancer cells, besides, induced G2/M phase cell cycle arrest in SGC7901 cells. Both apoptosis and autophagy were found simultaneously in SZC014-treated SGC7901 cells. Caspase-dependent apoptosis induced by SZC014 was confirmed to be associated with upregulation of Bax and downregulation of Bcl-2 and Bcl-xL, while upregulation of Beclin 1 and ATG 5 was inferred to be involved in SZC014-induced autophagy. Moreover, treating cells with SZC014 resulted in a decrease in phosphorylation of IκBα and NF-κB/p65 and NF-κB/p65 nuclear translocation. The cytotoxic activities of seven OA derivatives were generally stronger than that of OA, among which, SZC014 possessed the most potent anticancer activity in SGC7901 cells and would be a promising chemotherapic agent for the treatment of gastric cancer.
Collapse
Affiliation(s)
- Li Xiao Rui
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Song Yu Shu
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Wu Jing Jun
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Chen Zi Mo
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Sun Zheng Wu
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Liu Shu Min
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Lin Yuan
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Peng Jin Yong
- Pharmacology Department, Dalian Medical University, Dalian, China
| | - Song Zhi Cheng
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China
| | - Wang Shi Sheng
- College of Pharmaceutical Science and Technology, Dalian University of Technology, Dalian, China.
| | - Tang Ze Yao
- Pharmacology Department, Dalian Medical University, Dalian, China.
| |
Collapse
|
11
|
Abstract
‘Pharmacogenetics or Pharmacogenomics’ (PG) is one of the most practiced cancer therapeutic strategies, tailored for individualized patients. Despite its popularity and rapid advancements in the field, many obstacles for cancer therapy PG still need to be overcome. By borrowing scientific systems from other disciplines such as cancer diagnosis, and therapeutic information from the diversity of tumor origins, categories and stages, cancer therapy PG may hopefully be improved. Furthermore, to quickly acquire genetic and pathologic information and seek therapeutic interventions, possible breakthroughs may come from beyond – changing the cancer therapeutic landscapes. The next generations of PG protocols and hospital routines for searching deadly cancer pathogenic pathways versus drug-targeting predictions are of great clinical significance for the future. Yet, progress of cancer therapy PG is entering into a bottleneck stage owing to simple model of relevant techniques and routines. Promoting or even innovating present PG modular is very necessary. This perspective highlights this issue by introducing new initiatives and ideas.
Collapse
|
12
|
Su B, Su J, He H, Wu Y, Xia H, Zeng X, Dai W, Ai X, Ling H, Jiang H, Su Q. Identification of potential targets for diallyl disulfide in human gastric cancer MGC-803 cells using proteomics approaches. Oncol Rep 2015; 33:2484-94. [PMID: 25812569 DOI: 10.3892/or.2015.3859] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2014] [Accepted: 02/26/2015] [Indexed: 11/05/2022] Open
Abstract
Diallyl disulfide (DADS) is characterized as an effective agent for the prevention and therapy of cancer, however, mechanisms regarding its anticancer effects are not fully clarified. In the present study, we compared the protein expression profile of gastric cancer MGC-803 cells subjected to DADS treatment with that of untreated control cells to explore potential molecules regulated by DADS. Using proteomic approaches, we identified 23 proteins showing statistically significant differences in expression, including 9 upregulated and 14 downregulated proteins. RT-PCR and western blot analysis confirmed that retinoid-related orphan nuclear receptor α (RORα) and nM23 were increased by DADS, whereas LIM kinase-1 (LIMK1), urokinase-type plasminogen activator receptor (uPAR) and cyclin-dependent kinase-1 (CDK1) were decreased. DADS treatment and knockdown of uPAR caused suppression of ERK/Fra-1 pathway, downregulation of urokinase-type plasminogen activator (uPA), matrix metalloproteinase-9 (MMP-9) and vimentin, and upregulation of tissue inhibitor of metalloproteinase-3 (TIMP-3) and E-cadherin, concomitant with inhibition of cell migration and invasion. Moreover, knockdown of uPAR potentiated the effects of DADS on MGC-803 cells. These data demonstrate that downregulation of uPAR may partially be responsible for DADS-induced inhibition of ERK/Fra-1 pathway, as well as cell migration and invasion. Thus, the discovery of DADS-induced differential expression proteins is conducive to reveal unknown mechanisms of DADS anti-gastric cancer.
Collapse
Affiliation(s)
- Bo Su
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Jian Su
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui He
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Youhua Wu
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hong Xia
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xi Zeng
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Wenxiang Dai
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xiaohong Ai
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hui Ling
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Hao Jiang
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Qi Su
- Center for Gastric Cancer Research of Hunan Province, First Affiliated Hospital, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
13
|
Abstract
Given the interpatient biological heterogeneity and narrow therapeutic index of anticancer drugs, a practical method for personalizing cancer therapy is essential. Genotype-guided cancer therapy will provide an optimal approach to normalize systemic drug exposures, predict drug toxicities and/or enrich clinical efficacy. To date, over a dozen anticancer drugs approved by the US FDA require labeling regarding pharmacogenetic biomarkers (both germline and somatic). Many, but not all, have prospective, genotype-guided evidence-based data. Optimizing output from retrospective, prospective, cost-effectiveness and adaptive biomarker driven clinical trials will help drive the success of personalized cancer therapy. This review will discuss prospective genotype-guided clinical trials in patients with solid tumors and address barriers in clinical translation.
Collapse
Affiliation(s)
- Jai N Patel
- Department of Clinical Pharmacology, Levine Cancer Institute, Carolinas HealthCare System, 1021 Morehead Medical Drive, Charlotte, NC 28203, USA.
| |
Collapse
|