Skip to main content
Top
Published in: Annals of Surgical Oncology 7/2018

01-07-2018 | Gastrointestinal Oncology

Rates of TP53 Mutation are Significantly Elevated in African American Patients with Gastric Cancer

Authors: Elke J. A. H. van Beek, MD, Jonathan M. Hernandez, MD, Debra A. Goldman, MS, Jeremy L. Davis, MD, Kaitlin McLaughlin, MD, R. Taylor Ripley, MD, Teresa S. Kim, MD, Laura H. Tang, MD, PhD, Jaclyn F. Hechtman, MD, Jian Zheng, MD, Marinela Capanu, PhD, Nikolaus Schultz, PhD, David M. Hyman, MD, Marc Ladanyi, MD, Michael F. Berger, PhD, David B. Solit, MD, Yelena Y. Janjigian, MD, Vivian E. Strong, MD, FACS

Published in: Annals of Surgical Oncology | Issue 7/2018

Login to get access

Abstract

Background

Gastric adenocarcinoma is a heterogenous disease that results from complex interactions between environmental and genetic factors, which may contribute to the disparate outcomes observed between different patient populations. This study aimed to determine whether genomic differences exist in a diverse population of patients by evaluating tumor mutational profiles stratified by race.

Methods

All patients with gastric adenocarcinoma between 2012 and 2016 who underwent targeted next-generation sequencing of cancer genes by the Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets platform were identified. Patient race was categorized as Asian, African American, Hispanic, or Caucasian. Fisher’s exact test was used to examine differences in mutation rates between racial designations for the most common mutations identified. The p values in this study were adjusted using the false discovery rate method.

Results

The study investigated 595 mutations in 119 patients. The DNA alterations identified included missense mutations (66%), frame-shift deletions (13%), and nonsense mutations (9%). Silent mutations were excluded. The most frequently mutated genes were ARID1A, CDH1, ERBB3, KRAS, PIK3CA, and TP53. Of these, TP53 was the most frequently mutated gene, affecting 50% of patients. The proportion of patients with TP53 mutations differed significantly between races (p = 0.012). The findings showed TP53 mutations for 89% (16/18) of the African American patients, 56% (10/18) of the Asian patients, 43% (9/21) of the Hispanic patients, and 40% (25/62) of the Caucasian patients.

Conclusions

Significantly higher rates of TP53 mutations were identified among the African American patients with gastric adenocarcinoma. This is the first study to evaluate tumor genomic differences in a diverse population of patients with gastric adenocarcinoma.
Literature
1.
go back to reference World Health Organization. GLOBOCAN 2012: Stomach cancer estimated cancer incidence, mortality, and prevalence worldwide in 2012. http://globocan.iarc.fr. Accessed 13 July 2017. World Health Organization. GLOBOCAN 2012: Stomach cancer estimated cancer incidence, mortality, and prevalence worldwide in 2012. http://​globocan.​iarc.​fr. Accessed 13 July 2017.
2.
go back to reference Ohtsu A, Yoshida S, Saijo N. Disparities in gastric cancer chemotherapy between the East and West. J Clin Oncol. 2006;24:2188–96.CrossRefPubMed Ohtsu A, Yoshida S, Saijo N. Disparities in gastric cancer chemotherapy between the East and West. J Clin Oncol. 2006;24:2188–96.CrossRefPubMed
3.
go back to reference Allemani C, Weir HK, Carreira H, et al. Global surveillance of cancer survival 1995–2009: analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries (CONCORD-2). Lancet. 2015;385:977–1010.CrossRefPubMed Allemani C, Weir HK, Carreira H, et al. Global surveillance of cancer survival 1995–2009: analysis of individual data for 25,676,887 patients from 279 population-based registries in 67 countries (CONCORD-2). Lancet. 2015;385:977–1010.CrossRefPubMed
4.
go back to reference Strong VE, Song KY, Park CH, et al. Comparison of gastric cancer survival following R0 resection in the United States and Korea using an internationally validated nomogram. Ann Surg. 2010;251:640–6.CrossRefPubMed Strong VE, Song KY, Park CH, et al. Comparison of gastric cancer survival following R0 resection in the United States and Korea using an internationally validated nomogram. Ann Surg. 2010;251:640–6.CrossRefPubMed
5.
go back to reference Noguchi Y, Yoshikawa T, Tsuburaya A, Motohashi H, Karpeh MS, Brennan MF. Is gastric carcinoma different between Japan and the United States? Cancer. 2000;89:2237–46.CrossRefPubMed Noguchi Y, Yoshikawa T, Tsuburaya A, Motohashi H, Karpeh MS, Brennan MF. Is gastric carcinoma different between Japan and the United States? Cancer. 2000;89:2237–46.CrossRefPubMed
6.
go back to reference You WC, Li JY, Zhang L, Jin ML, Chang YS, Ma JL, Pan KF. Etiology and prevention of gastric cancer: a population study in a high-risk area of China. Chin J Dig Dis. 2005;6:149–54.CrossRefPubMed You WC, Li JY, Zhang L, Jin ML, Chang YS, Ma JL, Pan KF. Etiology and prevention of gastric cancer: a population study in a high-risk area of China. Chin J Dig Dis. 2005;6:149–54.CrossRefPubMed
7.
go back to reference Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 2014;513:202–9.CrossRef
8.
go back to reference Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRefPubMed Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRefPubMed
10.
go back to reference Howard JH, Hiles JM, Leung AM, Stern SL, Bilchik AJ. Race influences stage-specific survival in gastric cancer. Am Surg. 2015;81:259–67.PubMed Howard JH, Hiles JM, Leung AM, Stern SL, Bilchik AJ. Race influences stage-specific survival in gastric cancer. Am Surg. 2015;81:259–67.PubMed
11.
go back to reference Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn. 2015;17:251–64.CrossRefPubMedPubMedCentral Cheng DT, Mitchell TN, Zehir A, et al. Memorial Sloan Kettering-Integrated Mutation Profiling of Actionable Cancer Targets (MSK-IMPACT): a hybridization capture-based next-generation sequencing clinical assay for solid tumor molecular oncology. J Mol Diagn. 2015;17:251–64.CrossRefPubMedPubMedCentral
13.
go back to reference DeSantis CE, Siegel RL, Sauer AG, Miller KD, Fedewa SA, Alcaraz KI, Jemal A. Cancer Statistics for African Americans, 2016: progress and opportunities in reducing racial disparities. CA Cancer J Clin. 2016;66:290–308.CrossRefPubMed DeSantis CE, Siegel RL, Sauer AG, Miller KD, Fedewa SA, Alcaraz KI, Jemal A. Cancer Statistics for African Americans, 2016: progress and opportunities in reducing racial disparities. CA Cancer J Clin. 2016;66:290–308.CrossRefPubMed
14.
go back to reference Du XL, Lin CC, Johnson NJ, Altekruse S. Effects of individual-level socioeconomic factors on racial disparities in cancer treatment and survival: findings from the National Longitudinal Mortality Study, 1979–2003. Cancer. 2011;117:3242–51.CrossRefPubMedPubMedCentral Du XL, Lin CC, Johnson NJ, Altekruse S. Effects of individual-level socioeconomic factors on racial disparities in cancer treatment and survival: findings from the National Longitudinal Mortality Study, 1979–2003. Cancer. 2011;117:3242–51.CrossRefPubMedPubMedCentral
15.
go back to reference Albain KS, Unger JM, Crowley JJ, Coltman CA Jr, Hershman DL. Racial disparities in cancer survival among randomized clinical trials patients of the Southwest Oncology Group. J Natl Cancer Inst. 2009;101:984–92.CrossRefPubMedPubMedCentral Albain KS, Unger JM, Crowley JJ, Coltman CA Jr, Hershman DL. Racial disparities in cancer survival among randomized clinical trials patients of the Southwest Oncology Group. J Natl Cancer Inst. 2009;101:984–92.CrossRefPubMedPubMedCentral
16.
go back to reference Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRefPubMed Cristescu R, Lee J, Nebozhyn M, et al. Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med. 2015;21:449–56.CrossRefPubMed
17.
go back to reference Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 mutations in human cancers. Science. 1991;253:49–53.CrossRefPubMed Hollstein M, Sidransky D, Vogelstein B, Harris CC. p53 mutations in human cancers. Science. 1991;253:49–53.CrossRefPubMed
18.
go back to reference Hainaut P, Hollstein M. p53 and human cancer: the first ten thousand mutations. Adv Cancer Res. 2000;77:81–137.CrossRefPubMed Hainaut P, Hollstein M. p53 and human cancer: the first ten thousand mutations. Adv Cancer Res. 2000;77:81–137.CrossRefPubMed
19.
go back to reference Petitjean A, Achatz MI, Borresen-Dale AL, Hainaut P, Olivier M. TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes. Oncogene. 2007;26:2157–65.CrossRefPubMed Petitjean A, Achatz MI, Borresen-Dale AL, Hainaut P, Olivier M. TP53 mutations in human cancers: functional selection and impact on cancer prognosis and outcomes. Oncogene. 2007;26:2157–65.CrossRefPubMed
20.
go back to reference Olivier M, Langerod A, Carrieri P, et al. The clinical value of somatic TP53 gene mutations in 1794 patients with breast cancer. Clin Cancer Res. 2006;12:157–1167.CrossRef Olivier M, Langerod A, Carrieri P, et al. The clinical value of somatic TP53 gene mutations in 1794 patients with breast cancer. Clin Cancer Res. 2006;12:157–1167.CrossRef
21.
go back to reference Bergh J, Norberg T, Sjogren S, Lindgren A, Holmberg L. Complete sequencing of the p53 gene provides prognostic information in breast cancer patients, particularly in relation to adjuvant systemic therapy and radiotherapy. Nat Med. 1995;1:1029–34.CrossRefPubMed Bergh J, Norberg T, Sjogren S, Lindgren A, Holmberg L. Complete sequencing of the p53 gene provides prognostic information in breast cancer patients, particularly in relation to adjuvant systemic therapy and radiotherapy. Nat Med. 1995;1:1029–34.CrossRefPubMed
22.
go back to reference Aas T, Borresen AL, Geisler S, et al. Specific P53 mutations are associated with de novo resistance to doxorubicin in breast cancer patients. Nat Med. 1996;2:811–4.CrossRefPubMed Aas T, Borresen AL, Geisler S, et al. Specific P53 mutations are associated with de novo resistance to doxorubicin in breast cancer patients. Nat Med. 1996;2:811–4.CrossRefPubMed
23.
go back to reference Bull SB, Ozcelik H, Pinnaduwage D, et al. The combination of p53 mutation and neu/erbB-2 amplification is associated with poor survival in node-negative breast cancer. J Clin Oncol. 2004;22:86–96.CrossRefPubMed Bull SB, Ozcelik H, Pinnaduwage D, et al. The combination of p53 mutation and neu/erbB-2 amplification is associated with poor survival in node-negative breast cancer. J Clin Oncol. 2004;22:86–96.CrossRefPubMed
24.
go back to reference Kumar P, Aggarwal R. An overview of triple-negative breast cancer. Arch Gynecol Obstetr. 2016;293:247–69.CrossRef Kumar P, Aggarwal R. An overview of triple-negative breast cancer. Arch Gynecol Obstetr. 2016;293:247–69.CrossRef
25.
go back to reference Papa A, Caruso D, Tomao S, Rossi L, Zaccarelli E, Tomao F. Triple-negative breast cancer: investigating potential molecular therapeutic target. Expert Opin Ther Targets. 2015;19:55–75.CrossRefPubMed Papa A, Caruso D, Tomao S, Rossi L, Zaccarelli E, Tomao F. Triple-negative breast cancer: investigating potential molecular therapeutic target. Expert Opin Ther Targets. 2015;19:55–75.CrossRefPubMed
26.
go back to reference Mathe A, Scott RJ, Avery-Kiejda KA. MiRNAs and other epigenetic changes as biomarkers in triple-negative breast cancer. Int J Mol Sci. 2015;16:28347–76.CrossRefPubMedPubMedCentral Mathe A, Scott RJ, Avery-Kiejda KA. MiRNAs and other epigenetic changes as biomarkers in triple-negative breast cancer. Int J Mol Sci. 2015;16:28347–76.CrossRefPubMedPubMedCentral
27.
go back to reference Turner N, Moretti E, Siclari O, et al. Targeting triple-negative breast cancer: is p53 the answer? Cancer Treat Rev. 2013;39:541–50.CrossRefPubMed Turner N, Moretti E, Siclari O, et al. Targeting triple-negative breast cancer: is p53 the answer? Cancer Treat Rev. 2013;39:541–50.CrossRefPubMed
28.
go back to reference Bertheau P, Espié M, Turpin E, et al. TP53 status and response to chemotherapy in breast cancer. Pathobiology. 2008;75:132–9.CrossRefPubMed Bertheau P, Espié M, Turpin E, et al. TP53 status and response to chemotherapy in breast cancer. Pathobiology. 2008;75:132–9.CrossRefPubMed
Metadata
Title
Rates of TP53 Mutation are Significantly Elevated in African American Patients with Gastric Cancer
Authors
Elke J. A. H. van Beek, MD
Jonathan M. Hernandez, MD
Debra A. Goldman, MS
Jeremy L. Davis, MD
Kaitlin McLaughlin, MD
R. Taylor Ripley, MD
Teresa S. Kim, MD
Laura H. Tang, MD, PhD
Jaclyn F. Hechtman, MD
Jian Zheng, MD
Marinela Capanu, PhD
Nikolaus Schultz, PhD
David M. Hyman, MD
Marc Ladanyi, MD
Michael F. Berger, PhD
David B. Solit, MD
Yelena Y. Janjigian, MD
Vivian E. Strong, MD, FACS
Publication date
01-07-2018
Publisher
Springer International Publishing
Published in
Annals of Surgical Oncology / Issue 7/2018
Print ISSN: 1068-9265
Electronic ISSN: 1534-4681
DOI
https://doi.org/10.1245/s10434-018-6502-x

Other articles of this Issue 7/2018

Annals of Surgical Oncology 7/2018 Go to the issue

Health Services Research and Global Oncology

Will Palliative Care Ever Be Cool?

Health Services Research and Global Oncology

Changing the Status Quo, Including How We Measure It