Frequency of RAS Mutations (KRAS, NRAS, HRAS) in Human Solid Cancer

Total Page:16

File Type:pdf, Size:1020Kb

Frequency of RAS Mutations (KRAS, NRAS, HRAS) in Human Solid Cancer DOI: 10.14744/ejmo.2017.22931 EJMO 2017;1(1):1–7 Review Frequency of RAS Mutations (KRAS, NRAS, HRAS) in Human Solid Cancer Hilmi Kodaz,1 Osman Kostek,2 Muhammet Bekir Hacioglu,2 Bulent Erdogan,2 Cagnur Elpen Kodaz,3 Ilhan Hacibekiroglu,4 Esma Turkmen,2 Sernaz Uzunoglu,2 Irfan Cicin2 1Department of Medical Oncology, Acibadem Eskisehir Hospital, Eskisehir, Turkey 2Department of Medical Oncology, Trakya University Faculty of Medicine, Edirne, Turkey 3Department of Health, Acibadem Eskisehir Hospital, Eskisehir, Turkey 4Department of Medical Oncology, Sakarya University Faculty of Medicine, Sakarya, Turkey Abstract RAS oncogene affects numerous cellular functions including growth, proliferation, apoptosis, migration, division and differentiation of the cells. It has 3 known isoforms as Harvey- RAS (HRAS), Kirsten - RAS (KRAS) and Neuroblastoma- RAS (NRAS). RAS has an intrinsic GTPase activity. It encodes proteins binding the guanine nucleotides. KRAS and HRAS were discovered in studies carried out on viruses leading to cancer. Retroviral oncogenes related to murine sarcoma virus genes (Kristen Rat Sarcoma Virus and Murine Sarcoma Virus) were discovered in 1982. These two oncogenes are similar to human KRAS. Approximately 30% of all human cancers have ras genes. Mutations in KRAS account for about 85% for all RAS mutations in human tumors, NRAS is about 11–15%, and HRAS is about 1%. Keywords: KRAS, NRAS, HRAS, cancer Cite This Article: Kodaz H, Kostek O, Hacioglu M, Erdogan B, Elpen Kodaz C, Hacibekiroglu I, Turkmen E, Uzunoglu S, Cicin I. Frequency of Ras Mutations (Kras, Nras, Hras) in Human Solid Cancer. EJMO. 2017; 1(1): 1-7 he RAS oncogene affects numerous cellular functions, in- Frequency of RAS mutation Tcluding growth, proliferation, apoptosis, migration, divi- in intracranial tumors sion, and differentiation of the cells. It has 3 known isoforms: Harvey-RAS (HRAS), Kirsten-RAS (KRAS), and neuroblasto- Gliomas constitute 80% of malignant brain tumors. RAS ma-RAS (NRAS). RAS has an intrinsic GTPase activity; it en- mutations are very rare in malignant gliomas; no RAS mu- codes proteins binding the guanine nucleotides. KRAS and tation was found in a study in which 30 glioblastoma mul- HRAS were discovered in studies conducted on cancer-caus- tiforme (GBM) patients were evaluated. The frequency of ing viruses. Retroviral oncogenes related to murine sarcoma NRAS mutation was 2.1% in another study, although KRAS virus genes (Kristen rat sarcoma virus and Harvey rat sarco- and HRAS mutations were not detected. It was reported ma virus) were discovered in 1982. These 2 oncogenes are that RAS mutation frequency was 12% in a study of isoc- similar to human KRAS. Approximately 30% of all human itrate-dehydrogenase 1-mutant malignant glioma. RAS cancers have RAS gene involvement. Mutations in KRAS ac- genes were studied in 21 cases of glioblastoma multiforme, count for about 85% of all RAS mutations in human tumors, 4 cases of fibrillary astrocytoma, 4 cases of anaplastic astro- NRAS for about 11% to 15%, and HRAS for about 1%.[1–6] cytoma, and 15 normal specimens. RAS mutation was de- Address for correspondence: Hilmi Kodaz, MD. Acibadem Eskisehir Hastanesi, Tibbi Onkoloji Klinigi, Eskisehir, Turkey Phone: +90 507 931 42 86 E-mail: [email protected] Submitted Date: May 21, 2017 Accepted Date: July 03, 2017 Available Online Date: July 26, 2017 ©Copyright 2017 by Eurasian Journal of Medicine and Oncology - Available online at www.ejmo.org 2 Kodaz et al., Frequency of RAS Mutations / doi: 14744/ejmo.2017.22931 tected in 1 normal and 1 pathological sample. It was con- Table 1. Frequency of RAS mutations according to cancer type cluded that brain tumor development was found to involve Cancer KRAS NRAS HRAS KRAS and HRAS mutations, while NRAS mutation may play % % % a lesser role. The frequency of RAS mutation in GBM is 2% in Glioblastoma multiforme 0–2 2.1 0 the atlas of the human genome. KRAS mutation was found Pilocytic astrocytoma 4.7 0 0 in 4.7% of pilocytic astrocytomas, while there was no NRAS Oligodendroglioma LD LD LD and HRAS mutation. RAS mutations in oligodendrogliomas Oral cavity tumor 0–18 <1 35 [7–15] have not yet been sufficiently researched. Larynx cancer 4.8–11.5 0 0 Nasopharynx cancer 0 4 <1 Frequency of RAS mutation Colorectal cancer 27–56 1–7 <1 in head and neck cancers Stomach cancer 2.8–9.4 1,9 <1 Pancreatic cancer 90 <1 <1 Many head and neck cancers are squamous cell carcino- Hepatocellular cancer 0–5 15 29 mas. More than half a million people are affected every Cholangiocarcinoma 16–38 LD <1 year. The prevalence has continued to gradually increase in Small cell lung cancer 1–16 <1 <1 [16] recent years. Overall survival in patients with KRAS mu- Squamous cell lung cancer 3 <1 <1 tation head and neck tumor is worse. KRAS mutations also Lung adenocarcinoma 12–36 <1 <1 demonstrate social differences in head and neck tumors, Large cell lung cancer 14 <1 <1 like many cancers. Though the mutation frequency is 5% in Large cell lung cancer <1 <1 <1 oral cavity tumors in Western societies, it can be seen in up Serous ovarian carcinoma 6 8 <1 to 18% of cases in Eastern societies. The frequency of KRAS Ovarian clear cell carcinoma 14 0 mutation in malignant larynx lesions was 4.8% in one study IMSC 68 performed, while there were no HRAS and NRAS mutations. Low-grade serous tumour 29,5 In Eastern societies, the frequency of HRAS mutation in oral High-grade serous carcinoma 12 cavity tumors can be as much as 35%. KRAS mutation was Mucinous borderline tumor 33–78 not detected at all in mouth and hypopharynx cancers in Serous borderline tumor 60 another study conducted. There is KRAS mutation in 11.5% Mucinous carcinoma 46–75 and 3.3% of patients with laryngeal cancer and oropharynx LGOEC 33 cancer, respectively. NRAS mutation in nasopharynx cancer Endometrial cancer 11–35 LGEEC 18 is reported as 4%, while HRAS mutation is <1% and KRAS NEEC 0–5 mutation is very rare.[17–21] Cervical cancer 5–14 <1 <1 Frequency of RAS mutation Cervical squamous cell carcinoma <1 Cervical adenocarcinoma 10 in gastrointestinal cancer Prostate adenocarcinoma 7–9 Colorectal cancers are one of the leading causes of can- Vulvar cancer 1 9 cer-related death, though incidence and mortality have Germ cell testicular tumor 6 6 LD decreased in recent years. This is probably due to early di- Seminoma 6–18 <1 LD agnosis and screening methods. RAS mutation testing is Non-seminoma 0–9 <1 LD frequently used in everyday practice in cases of metastatic Bladder cancer 0–4 0–80 0–84 colorectal cancer. RAS mutations are significant in the selec- Renal cell carcinoma 0–16 <1 <1 Clear cell renal cell carcinoma 0–100 <1 <1 tion of targeted treatment. There is no marker for the use of Soft tissue sarcoma 0–44 <1 0–16 bevacizumab; however, KRAS wild tumors are a marker for Cutaneous malignant melanoma <1 15–30 <1 the use of cetuximab, and KRAS and NRAS wild colorectal Breast cancer 7–12 5 <1 tumors are a marker for the use of panitumumab. The fre- Luminal A 2 quency of KRAS mutations in colorectal cancers varies from Luminal B 20 27% to 56% from community to community. The frequency HER2 overexpression 17 of NRAS mutations in colorectal cancers is between 1% and Triple negative 0–8 7%. NRAS mutation frequency is 5.3%, 4.1%, and 2.2% in KRAS: Kirsten - RAS; NRAS: Neuroblastoma- RAS; HRAS: Harvey- RAS: IMSC: Romanian, Italian, and Chinese patients, respectively. There Invasive micropapillary serous carcinoma; LGOEC: Low-grade ovarian is almost no HRAS mutation seen in colorectal cancers. endometrioid carcinomas; LGEEC: Low-grade endometrial endometrioid carcinoma; NEEC: Non- endometrioid endometrial cancer; LD: Limited data KRAS mutations do not seem to be prognostic in colorectal EJMO 3 cancers, but are predictive. NRAS mutations also show geo- Frequency of RAS mutation graphical and ethnic differences in colorectal cancers, like in genitourinary cancer KRAS mutations. The frequency of KRAS mutations in the right and left colon is different. Exposure to embryogen- RAS mutation frequency in cancer overall is 27%. Ovarian esis and/or carcinogens may play a role in this difference. clear cell carcinoma constitutes 15% of all ovarian can- The left colon is subject to carbonaceous substances for a cers. KRAS mutation frequency is 14% in ovarian clear cell greater period of time. It is interesting that there are fewer carcinoma, and there is no NRAS mutation. The frequency KRAS mutations in the left colon than in the right colon. of NRAS mutation in epithelial ovarian cancer is 8%. KRAS The first column shadow subject to carcinogenic substanc- mutation prevalence is 68%, 29.5%, 12%, 33%–78%, 60%, es is the right colon. The right colon may be more sensitive 46%–75%, 6%, and 33% in invasive micropapillary serous to carcinogenic substances. carcinoma, low-grade serous tumor, high-grade serous carcinoma, mucinous borderline tumor, serous borderline Stomach cancers are observed more often in Third World tumor, mucinous carcinoma, serous ovarian carcinoma, countries and Eastern societies. KRAS mutation frequency and low-grade ovarian endometrioid carcinoma, respec- in gastric cancer has ranged from 2.8% to 9.4% in studies. tively. In ovarian cancer, the frequency of KRAS mutation NRAS mutation in gastric cancer can be found in 1.9%. varies greatly according to the histological subtype. The HRAS mutation is very rare in gastric cancer. KRAS muta- data regarding KRAS mutation are limited. The frequency tion is present in over 90% of pancreatic cancers. There are of KRAS mutation in endometrial cancer ranges from 11% almost no NRAS and HRAS mutations in pancreatic cancers.
Recommended publications
  • The Rac Gtpase in Cancer: from Old Concepts to New Paradigms Marcelo G
    Published OnlineFirst August 14, 2017; DOI: 10.1158/0008-5472.CAN-17-1456 Cancer Review Research The Rac GTPase in Cancer: From Old Concepts to New Paradigms Marcelo G. Kazanietz1 and Maria J. Caloca2 Abstract Rho family GTPases are critical regulators of cellular func- mislocalization of Rac signaling components. The unexpected tions that play important roles in cancer progression. Aberrant pro-oncogenic functions of Rac GTPase-activating proteins also activity of Rho small G-proteins, particularly Rac1 and their challenged the dogma that these negative Rac regulators solely regulators, is a hallmark of cancer and contributes to the act as tumor suppressors. The potential contribution of Rac tumorigenic and metastatic phenotypes of cancer cells. This hyperactivation to resistance to anticancer agents, including review examines the multiple mechanisms leading to Rac1 targeted therapies, as well as to the suppression of antitumor hyperactivation, particularly focusing on emerging paradigms immune response, highlights the critical need to develop ther- that involve gain-of-function mutations in Rac and guanine apeutic strategies to target the Rac pathway in a clinical setting. nucleotide exchange factors, defects in Rac1 degradation, and Cancer Res; 77(20); 5445–51. Ó2017 AACR. Introduction directed toward targeting Rho-regulated pathways for battling cancer. Exactly 25 years ago, two seminal papers by Alan Hall and Nearly all Rho GTPases act as molecular switches that cycle colleagues illuminated us with one of the most influential dis- between GDP-bound (inactive) and GTP-bound (active) forms. coveries in cancer signaling: the association of Ras-related small Activation is promoted by guanine nucleotide exchange factors GTPases of the Rho family with actin cytoskeleton reorganization (GEF) responsible for GDP dissociation, a process that normally (1, 2).
    [Show full text]
  • A GTP-State Specific Cyclic Peptide Inhibitor of the Gtpase Gαs
    bioRxiv preprint doi: https://doi.org/10.1101/2020.04.25.054080; this version posted April 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. A GTP-state specific cyclic peptide inhibitor of the GTPase Gαs Shizhong A. Dai1,2†, Qi Hu1,2†, Rong Gao3†, Andre Lazar1,4†, Ziyang Zhang1,2, Mark von Zastrow1,4, Hiroaki Suga3*, Kevan M. Shokat1,2* 5 1Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, 94158, USA 2Howard Hughes Medical Institute 3Department of Chemistry, Graduate School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan 10 4Department of Psychiatry, University of California, San Francisco, San Francisco, CA, 94158, USA *Correspondence to: [email protected], [email protected] †These authors contributed equally. 15 20 1 bioRxiv preprint doi: https://doi.org/10.1101/2020.04.25.054080; this version posted April 27, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Abstract: The G protein-coupled receptor (GPCR) cascade leading to production of the second messenger cAMP is replete with pharmacologically targetable receptors and enzymes with the exception of the stimulatory G protein α subunit, Gαs.
    [Show full text]
  • Clinical Utility of Recently Identified Diagnostic, Prognostic, And
    Modern Pathology (2017) 30, 1338–1366 1338 © 2017 USCAP, Inc All rights reserved 0893-3952/17 $32.00 Clinical utility of recently identified diagnostic, prognostic, and predictive molecular biomarkers in mature B-cell neoplasms Arantza Onaindia1, L Jeffrey Medeiros2 and Keyur P Patel2 1Instituto de Investigacion Marques de Valdecilla (IDIVAL)/Hospital Universitario Marques de Valdecilla, Santander, Spain and 2Department of Hematopathology, MD Anderson Cancer Center, Houston, TX, USA Genomic profiling studies have provided new insights into the pathogenesis of mature B-cell neoplasms and have identified markers with prognostic impact. Recurrent mutations in tumor-suppressor genes (TP53, BIRC3, ATM), and common signaling pathways, such as the B-cell receptor (CD79A, CD79B, CARD11, TCF3, ID3), Toll- like receptor (MYD88), NOTCH (NOTCH1/2), nuclear factor-κB, and mitogen activated kinase signaling, have been identified in B-cell neoplasms. Chronic lymphocytic leukemia/small lymphocytic lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, mantle cell lymphoma, Burkitt lymphoma, Waldenström macroglobulinemia, hairy cell leukemia, and marginal zone lymphomas of splenic, nodal, and extranodal types represent examples of B-cell neoplasms in which novel molecular biomarkers have been discovered in recent years. In addition, ongoing retrospective correlative and prospective outcome studies have resulted in an enhanced understanding of the clinical utility of novel biomarkers. This progress is reflected in the 2016 update of the World Health Organization classification of lymphoid neoplasms, which lists as many as 41 mature B-cell neoplasms (including provisional categories). Consequently, molecular genetic studies are increasingly being applied for the clinical workup of many of these neoplasms. In this review, we focus on the diagnostic, prognostic, and/or therapeutic utility of molecular biomarkers in mature B-cell neoplasms.
    [Show full text]
  • Tumor Markers
    Tumor Markers Alan H.B. Wu, Ph.D. Professor, Laboratory Medicine, UCSF Section Chief, Clinical Chemistry, Toxicology, Pharmacogenomics Laboratory, SFGH Learning objectives • Know the ideal characteristics of a tumor marker • Understand the role of tumor markers for diagnosis and management of patients with cancer. • Know the emerging technologies for tumor markers • Understand the role of tumor markers for therapeutic selection How do we diagnose cancer today? Physical Examination Blood tests CT scans Biopsy Human Prostate Cancer Normal Blood Smear Chronic Myeloid Leukemia Death rates for cancer vs. heart disease New cancer cases per year Cancer Site or Type New Cases Prostate 218,000 Lung 222,500 Breast 207,500 Colorectal 149,000 Urinary system 131,500 Skin 68,770 Pancreas 43,100 Ovarian 22,000 Myeloma 20,200 Thyroid 44,700 Germ Cell 9,000 Types of Tumor Markers • Hormones (hCG; calcitonin; gastrin; prolactin;) • Enzymes (acid phosphatase; alkaline phosphatase; PSA) • Cancer antigen proteins & glycoproteins (CA125; CA 15.3; CA19.9) • Metabolites (norepinephrine, epinephrine) • Normal proteins (thyroglobulin) • Oncofetal antigens (CEA, AFP) • Receptors (ER, PR, EGFR) • Genetic changes (mutations/translocations, etc.) Characteristics of an ideal tumor marker • Specificity for a single type of cancer • High sensitivity and specificity for cancerous growth • Correlation of marker level with tumor size • Homogeneous (i.e., minimal post-translational modifications) • Short half-life in circulation Roles for tumor markers • Determine risk (PSA)
    [Show full text]
  • Screening for Tumor Suppressors: Loss of Ephrin PNAS PLUS Receptor A2 Cooperates with Oncogenic Kras in Promoting Lung Adenocarcinoma
    Screening for tumor suppressors: Loss of ephrin PNAS PLUS receptor A2 cooperates with oncogenic KRas in promoting lung adenocarcinoma Narayana Yeddulaa, Yifeng Xiaa, Eugene Kea, Joep Beumera,b, and Inder M. Vermaa,1 aLaboratory of Genetics, The Salk Institute for Biological Studies, La Jolla, CA 92037; and bHubrecht Institute, Utrecht, The Netherlands Contributed by Inder M. Verma, October 12, 2015 (sent for review July 28, 2015; reviewed by Anton Berns, Tyler Jacks, and Frank McCormick) Lung adenocarcinoma, a major form of non-small cell lung cancer, injections in embryonic skin cells identified several potential tu- is the leading cause of cancer deaths. The Cancer Genome Atlas morigenic factors (14–16). None of the reported studies have analysis of lung adenocarcinoma has identified a large number of performed direct shRNA-mediated high-throughput approaches previously unknown copy number alterations and mutations, re- in adult mice recapitulating the mode of tumorigenesis in humans. quiring experimental validation before use in therapeutics. Here, we Activating mutations at positions 12, 13, and 61 amino acids in describe an shRNA-mediated high-throughput approach to test a set Kirsten rat sarcoma viral oncogene homolog (KRas) contributes of genes for their ability to function as tumor suppressors in the to tumorigenesis in 32% of lung adenocarcinoma patients (2) by background of mutant KRas and WT Tp53. We identified several activating downstream signaling cascades. Mice with the KRasG12D candidate genes from tumors originated from lentiviral delivery of allele develop benign adenomatous lesions with long latency to shRNAs along with Cre recombinase into lungs of Loxp-stop-Loxp- develop adenocarcinoma (17, 18).
    [Show full text]
  • The Effects of Oncogenic G12d Mutation on K-Ras Structure, Conformation and Dynamics
    bioRxiv preprint doi: https://doi.org/10.1101/178483; this version posted August 19, 2017. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. THE EFFECTS OF ONCOGENIC G12D MUTATION ON K-RAS STRUCTURE, CONFORMATION AND DYNAMICS Sezen Vatansever1, 2, 3, Zeynep H. Gümüş2, 3*, Burak Erman1* 1Department of Chemical and Biological Engineering, College of Engineering, Koç University, Rumelifeneri Yolu, 34450, Sarıyer, Istanbul, Turkey 2Department of Genetics and Genomics, 3Icahn Institute for Genomics and Multiscale Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 *Correspondence: [email protected] (B.E.) and [email protected] (Z.H.G.) ABSTRACT K-Ras is the most frequently mutated protein in human tumors. Activating K-Ras mutations drive cancer initiation, progression and drug resistance, directly leading to nearly a million deaths per year. To understand the mechanisms by which mutations alter K-Ras function, we need to understand their effects on protein dynamics. However, despite decades of research, how oncogenic mutations in K-Ras alter its conformation and dynamics remain to be understood. Here, we present how the most recurrent K-Ras oncogenic mutation, G12D, leads to structural, conformational and dynamical changes that lead to constitutively active K- Ras. We have developed a new integrated MD simulation data analysis approach to quantify such changes in a protein and applied it to K-Ras. Our results show that G12D mutation induces strong negative correlations between the fluctuations of SII and those of the P-loop, Switch I (SI) and α3 regions in K-RasG12D.
    [Show full text]
  • Agonist-Induced Formation of Unproductive Receptor-G12 Complexes Najeah Okashaha, Shane C
    Agonist-induced formation of unproductive receptor-G12 complexes Najeah Okashaha, Shane C. Wrightb, Kouki Kawakamic, Signe Mathiasend,e,f, Joris Zhoub, Sumin Lua, Jonathan A. Javitchd,e,f, Asuka Inouec, Michel Bouvierb, and Nevin A. Lamberta,1 aDepartment of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912; bInstitute for Research in Immunology and Cancer, Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada; cGraduate School of Pharmaceutical Sciences, Tohoku University, 980-8578 Sendai, Japan; dDepartment of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032; eDepartment of Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032; and fDivision of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032 Edited by Brian K. Kobilka, Stanford University School of Medicine, Stanford, CA, and approved July 24, 2020 (received for review February 28, 2020) G proteins are activated when they associate with G protein- association does not always promote G protein activation and coupled receptors (GPCRs), often in response to agonist-mediated may in some circumstances inhibit downstream signaling. receptor activation. It is generally thought that agonist-induced receptor-G protein association necessarily promotes G protein acti- Results vation and, conversely, that activated GPCRs do not interact with V2R Interacts with G12 Heterotrimers. Conventional GPCR-G pro- G proteins that they do not activate. Here we show that GPCRs can tein coupling is understood as an allosteric interaction where an form agonist-dependent complexes with G proteins that they do agonist-bound active receptor mediates GDP release by stabi- not activate.
    [Show full text]
  • 1 Gα13 Loss Promotes Tumor Progression in the KPC Transgenic
    bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435488; this version posted March 15, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Gα13 loss promotes tumor progression in the KPC transgenic mouse model of advanced pancreatic cancer Mario A. Shields1,2,*, Christina Spaulding1,3, Mahmoud G. Khalafalla1, Thao N.D. Pham1, and Hidayatullah G. Munshi1,2,3,* 1 Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA 2 The Robert H. Lurie Comprehensive Cancer Center, Chicago, IL, USA 3 Jesse Brown VA Medical Center, Chicago, IL, USA Running title: Effects of epithelial Gα13 loss in vivo Keywords: Gα13, KC mouse model, KPC mouse model, human PDAC tumors, E-cadherin, mTOR The authors have declared that no conflict of interest exists. *Address correspondence to Mario A. Shields, Ph.D. or Hidayatullah G. Munshi, M.D. Department of Medicine Northwestern University Feinberg School of Medicine 303 E. Superior Ave., Lurie 3-220 (M.A. Shields) or Lurie 3-117 (H.G. Munshi) Chicago, IL 60611, USA. Phone: (312) 503-0312; [email protected] (M.A. Shields) Phone: (312) 503-2301; [email protected] (H.G. Munshi) 1 bioRxiv preprint doi: https://doi.org/10.1101/2021.03.15.435488; this version posted March 15, 2021. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. SUMMARY Gα13 transduces signals from G protein-coupled receptors.
    [Show full text]
  • The Frequency of Ras Mutations in Cancer
    Author Manuscript Published OnlineFirst on March 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3682 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. The frequency of Ras mutations in cancer Ian A. Prior1*, Fiona E. Hood1 and James L. Hartley2 CAN-19-3682R Running Title: Ras cancer statistics Keywords: HRAS, KRAS, NRAS, cancer, mutation, genetics Word Count: 3220 + references and Figure legends 1. Division of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, L69 3BX, UK. 2. NCI RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, 21701, USA. * Corresponding Author: Ian A. Prior Email: [email protected] Tel: +44-151-794-5332 Fax: +44-151-794-4434 Conflict of interest statement All authors declare that they have no conflicts of interest relevant to this manuscript. Author contributions All authors have contributed to the writing and design of this manuscript. IAP is responsible for the conception, analysis, design and writing. 1 Downloaded from cancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 24, 2020; DOI: 10.1158/0008-5472.CAN-19-3682 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. ABSTRACT Ras is frequently mutated in cancer; however, there is a lack of consensus in the literature regarding the cancer mutation frequency of Ras, with quoted values varying from 10-30%. This variability is at least in part due to the selective aggregation of data from different databases and the dominant influence of particular cancer types and particular Ras isoforms within these datasets.
    [Show full text]
  • Identification of Novel GNAS Mutations in Intramuscular Myxoma Using Next- Generation Sequencing with Single-Molecule Tagged Molecular Inversion Probes Elise M
    Bekers et al. Diagnostic Pathology (2019) 14:15 https://doi.org/10.1186/s13000-019-0787-3 RESEARCH Open Access Identification of novel GNAS mutations in intramuscular myxoma using next- generation sequencing with single-molecule tagged molecular inversion probes Elise M. Bekers1,2* , Astrid Eijkelenboom1, Paul Rombout1, Peter van Zwam3, Suzanne Mol4, Emiel Ruijter5, Blanca Scheijen1 and Uta Flucke1 Abstract Background: Intramuscular myxoma (IM) is a hypocellular benign soft tissue neoplasm characterized by abundant myxoid stroma and occasional hypercellular areas. These tumors can, especially on biopsy material, be difficult to distinguish from low-grade fibromyxoid sarcoma or low-grade myxofibrosarcoma. GNAS mutations are frequently involved in IM, in contrast to these other malignant tumors. Therefore, sensitive molecular techniques for detection of GNAS aberrations in IM, which frequently yield low amounts of DNA due to poor cellularity, will be beneficial for differential diagnosis. Methods: In our study, a total of 34 IM samples from 33 patients were analyzed for the presence of GNAS mutations, of which 29 samples were analyzed using a gene-specific TaqMan genotyping assay for the detection of GNAS hotspot mutations c.601C > T and c602G > A in IM, and 32 samples using a novel next generation sequencing (NGS)-based approach employing single-molecule tagged molecular inversion probes (smMIP) to identify mutations in exon 8 and 9 of GNAS. Results between the two assays were compared for their ability to detect GNAS mutations with high confidence. Results: In total, 23 of 34 samples were successfully analyzed with both techniques showing GNAS mutations in 12 out of 23 (52%) samples.
    [Show full text]
  • Colorectal Cancer Invasion and Atrophy of the Enteric Nervous System: Potential Feedback and Impact on Cancer Progression
    International Journal of Molecular Sciences Review Colorectal Cancer Invasion and Atrophy of the Enteric Nervous System: Potential Feedback and Impact on Cancer Progression Janusz Godlewski 1,* and Zbigniew Kmiec 2 1 Department of Human Histology and Embryology, Collegium Medicum, School of Medicine, University of Warmia and Mazury, 10-082 Olsztyn, Poland 2 Department of Histology, Medical University of Gdansk, 80-210 Gdansk, Poland; [email protected] * Correspondence: [email protected]; Tel.: +48-89-5245302 Received: 31 March 2020; Accepted: 8 May 2020; Published: 11 May 2020 Abstract: Colorectal cancer (CRC) invasion within the large intestine wall results in the replacement of normal tissue architecture by tumour mass. Cancer cells digest the extracellular matrix (ECM) by the release of proteolytic enzymes. The disintegration of matrix ground substance activates several deposited growth factors which stimulate cell proliferation. Stromal (mainly fibroblasts), immune and cancer cells dominate in this area and become involved in a network of multimodal interactions which significantly induce proliferation of colon cancer cells, inhibit their apoptosis and promote their spreading within the local tumour microenvironment. Cancer invasion destroys nerve fibres and neurons of the local enteric nervous system (ENS) and induces subsequent atrophy of the submucosal and myenteric plexuses in areas adjacent to the cancer boundary. Interestingly, the reduction of plexuses’ size is accompanied by the increased number of galanin-immunoreactive neurons and increased galanin content in parts of the colon located close to the tumour. Galanin, a neuroprotective peptide, may inhibit the extrinsic pathway of apoptosis and in this way promote cancer cell survival. The possible role of acetylcholine and some ENS neuropeptides was also discussed.
    [Show full text]
  • Next-Generation Sequencing Expanded NGS Gene List
    Next-Generation Sequencing Expanded NGS Gene List Mutations (DNA) ABI1 BRD4 CRLF2 FOXO4 HOXC11 KLF4 MUC1 PAK3 RHOH TAL2 ABL1 BTG1 DDB2 FSTL3 HOXC13 KLK2 MUTYH PATZ1 RNF213 TBL1XR1 ACKR3 BTK DDIT3 GATA1 HOXD11 LASP1 MYCL (MYCL1) PAX8 RPL10 TCEA1 AKT1 C15orf65 DNM2 GATA2 HOXD13 LMO1 NBN PDE4DIP SEPT5 TCL1A ® AMER1 (FAM123B) CBLC DNMT3A GNA11 HRAS LMO2 NDRG1 PHF6 SEPT6 TERT Tumor Profiling Services from Caris Molecular Intelligence AR CD79B EIF4A2 GPC3 IKBKE MAFB NKX2-1 PHOX2B SFPQ TFE3 ARAF CDH1 ELF4 HEY1 INHBA MAX NONO PIK3CG SLC45A3 TFPT ATP2B3 CDK12 ELN HIST1H3B IRS2 MECOM NOTCH1 PLAG1 SMARCA4 THRAP3 ATRX CDKN2B ERCC1 HIST1H4I JUN MED12 NRAS PMS1 SOCS1 TLX3 BCL11B CDKN2C ETV4 HLF KAT6A (MYST3) MKL1 NUMA1 POU5F1 SOX2 TMPRSS2 BCL2 CEBPA FAM46C HMGN2P46 KAT6B MLLT11 NUTM2B PPP2R1A SPOP UBR5 Next-Generation BCL2L2 CHCHD7 FANCF HNF1A KCNJ5 MN1 OLIG2 PRF1 SRC VHL Sequencing BCOR CNOT3 FEV HOXA11 KDM5C MPL OMD PRKDC SSX1 WAS Multi-platform, solid tumor BCORL1 COL1A1 FOXL2 HOXA13 KDM6A MSN P2RY8 RAD21 STAG2 ZBTB16 Next-Generation Sequencing for BRD3 COX6C FOXO3 HOXA9 KDSR MTCP1 PAFAH1B2 RECQL4 TAL1 ZRSR2 biomarker analysis for therapeutic decision support additional biomarker analysis Mutations and Copy Number Variations (DNA) ABL2 BRCA21 COPB1 ESR1 FUS KIT MYB PER1 RUNX1 TFG ACSL3 BRIP1 CREB1 ETV1 GAS7 KLHL6 MYC PICALM RUNX1T1 TFRC Chemotherapy 4 – ACSL6 BUB1B CREB3L1 ETV5 GATA3 KMT2A (MLL) MYCN PIK3CA SBDS TGFBR2 AFF1 C11orf30 (EMSY) CREB3L2 ETV6 GID4 (C17orf39) KMT2C (MLL3) MYD88 PIK3R1 SDC4 TLX1 Immunotherapy 4 – AFF3 C2orf44 CREBBP EWSR1
    [Show full text]