Skip to main content
Erschienen in: International Journal of Clinical Oncology 7/2019

12.04.2019 | Invited Review Article

Molecular carcinogenesis of gastric cancer: Lauren classification, mucin phenotype expression, and cancer stem cells

verfasst von: Naohide Oue, Kazuhiro Sentani, Naoya Sakamoto, Naohiro Uraoka, Wataru Yasui

Erschienen in: International Journal of Clinical Oncology | Ausgabe 7/2019

Einloggen, um Zugang zu erhalten

Abstract

Gastric cancer (GC), one of the most common human cancers, is a heterogeneous disease with different phenotypes, prognoses, and responses to treatment. Understanding the pathogenesis of GC at the molecular level is important for prognosis prediction and determining treatments. Microsatellite instability (MSI), silencing of MLH1, MGMT, and CDKN2A genes by DNA hypermethylation, KRAS mutation, APC mutation, and ERBB2 amplification are frequently found in intestinal type GC. Inactivation of CDH1 and RARB by DNA hypermethylation, and amplification of FGFR and MET, are frequently detected in diffuse type GC. In addition, BST2 and PCDHB9 genes are overexpressed in intestinal type GC. Both genes are associated with GC progression. GC can be divided into gastric/intestinal mucin phenotypes according to mucin expression. MSI, alterations of TP73, CDH1 mutation, and DNA methylation of MLH are detected frequently in the gastric mucin phenotype. TP53 mutation, deletion of APC, and DNA methylation of MGMT are detected frequently in the intestinal mucin phenotype. FKTN is overexpressed in the intestinal mucin phenotype, and IQGAP3 is overexpressed in the gastric mucin phenotype. These genes are involved in GC progression. To characterize cancer stem cells, a useful method is spheroid colony formation. KIFC1 and KIF11 genes show more than twofold higher expression in spheroid-forming cells than that in parental cells. Both KIF genes are overexpressed in GC, and knockdown of these genes inhibits spheroid formation. Alterations of these molecules may be useful to understand gastric carcinogenesis. Specific inhibitors of these molecules may also be promising anticancer drugs.
Literatur
1.
Zurück zum Zitat Nakamura K, Sugano H, Takagi K (1968) Carcinoma of the stomach in incipient phase: its histogenesis and histological appearances. Gann 59:251–258PubMed Nakamura K, Sugano H, Takagi K (1968) Carcinoma of the stomach in incipient phase: its histogenesis and histological appearances. Gann 59:251–258PubMed
2.
Zurück zum Zitat Lauren P (1965) The two histological main types of gastric carcinoma. Diffuse and so-called intestinal type carcinoma: an attempt at histological classification. Acta Pathol Microbiol Scand 64:31–49CrossRefPubMed Lauren P (1965) The two histological main types of gastric carcinoma. Diffuse and so-called intestinal type carcinoma: an attempt at histological classification. Acta Pathol Microbiol Scand 64:31–49CrossRefPubMed
3.
Zurück zum Zitat Vauhkonen M, Vauhkonen H, Sipponen P (2006) Pathology and molecular biology of gastric cancer. Best Pract Res Clin Gastroenterol 20:651–674CrossRefPubMed Vauhkonen M, Vauhkonen H, Sipponen P (2006) Pathology and molecular biology of gastric cancer. Best Pract Res Clin Gastroenterol 20:651–674CrossRefPubMed
4.
Zurück zum Zitat Yasui W, Oue N, Ito R et al (2004) Search for new biomarkers of gastric cancer through serial analysis of gene expression and its clinical implications. Cancer Sci 95:385–392CrossRefPubMed Yasui W, Oue N, Ito R et al (2004) Search for new biomarkers of gastric cancer through serial analysis of gene expression and its clinical implications. Cancer Sci 95:385–392CrossRefPubMed
5.
Zurück zum Zitat Oue N, Sentani K, Sakamoto N et al (2015) Clinicopathologic and molecular characteristics of gastric cancer showing gastric and intestinal mucin phenotype. Cancer Sci 106:951–958CrossRefPubMedPubMedCentral Oue N, Sentani K, Sakamoto N et al (2015) Clinicopathologic and molecular characteristics of gastric cancer showing gastric and intestinal mucin phenotype. Cancer Sci 106:951–958CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat Kupzig S, Korolchuk V, Rollason R et al (2003) Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology. Traffic 4:694–709CrossRefPubMed Kupzig S, Korolchuk V, Rollason R et al (2003) Bst-2/HM1.24 is a raft-associated apical membrane protein with an unusual topology. Traffic 4:694–709CrossRefPubMed
7.
Zurück zum Zitat Goto T, Kennel SJ, Abe M et al (1994) A novel membrane antigen selectively expressed on terminally differentiated human B cells. Blood 84:1922–1930PubMed Goto T, Kennel SJ, Abe M et al (1994) A novel membrane antigen selectively expressed on terminally differentiated human B cells. Blood 84:1922–1930PubMed
8.
9.
Zurück zum Zitat Wang W, Nishioka Y, Ozaki S et al (2009) HM1.24 (CD317) is a novel target against lung cancer for immunotherapy using anti-HM1.24 antibody. Cancer Immunol Immunother 58:967–976CrossRefPubMed Wang W, Nishioka Y, Ozaki S et al (2009) HM1.24 (CD317) is a novel target against lung cancer for immunotherapy using anti-HM1.24 antibody. Cancer Immunol Immunother 58:967–976CrossRefPubMed
10.
Zurück zum Zitat Mukai S, Oue N, Oshima T et al (2017) Overexpression of transmembrane protein BST2 is associated with poor survival of patients with esophageal, gastric, or colorectal cancer. Ann Surg Oncol 24:594–602CrossRefPubMed Mukai S, Oue N, Oshima T et al (2017) Overexpression of transmembrane protein BST2 is associated with poor survival of patients with esophageal, gastric, or colorectal cancer. Ann Surg Oncol 24:594–602CrossRefPubMed
11.
Zurück zum Zitat Liu W, Cao Y, Guan Y et al (2018) BST2 promotes cell proliferation, migration and induces NF-kappaB activation in gastric cancer. Biotechnol Lett 40:1015–1027CrossRefPubMed Liu W, Cao Y, Guan Y et al (2018) BST2 promotes cell proliferation, migration and induces NF-kappaB activation in gastric cancer. Biotechnol Lett 40:1015–1027CrossRefPubMed
13.
Zurück zum Zitat Mukai S, Oue N, Oshima T et al (2017) Overexpression of PCDHB9 promotes peritoneal metastasis and correlates with poor prognosis in patients with gastric cancer. J Pathol 243:100–110CrossRefPubMed Mukai S, Oue N, Oshima T et al (2017) Overexpression of PCDHB9 promotes peritoneal metastasis and correlates with poor prognosis in patients with gastric cancer. J Pathol 243:100–110CrossRefPubMed
14.
Zurück zum Zitat Sekino Y, Oue N, Mukai S et al (2019) Protocadherin B9 promotes resistance to bicalutamide and is associated with the survival of prostate cancer patients. Prostate 79:234–242CrossRefPubMed Sekino Y, Oue N, Mukai S et al (2019) Protocadherin B9 promotes resistance to bicalutamide and is associated with the survival of prostate cancer patients. Prostate 79:234–242CrossRefPubMed
15.
Zurück zum Zitat Liu JY, Jiang L, He T et al (2019) NETO2 promotes invasion and metastasis of gastric cancer cells via activation of PI3K/Akt/NF-kappaB/Snail axis and predicts outcome of the patients. Cell Death Dis 10:162CrossRefPubMedPubMedCentral Liu JY, Jiang L, He T et al (2019) NETO2 promotes invasion and metastasis of gastric cancer cells via activation of PI3K/Akt/NF-kappaB/Snail axis and predicts outcome of the patients. Cell Death Dis 10:162CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Yasui W, Oue N, Kuniyasu H et al (2001) Molecular diagnosis of gastric cancer: present and future. Gastric Cancer 4:113–121CrossRefPubMed Yasui W, Oue N, Kuniyasu H et al (2001) Molecular diagnosis of gastric cancer: present and future. Gastric Cancer 4:113–121CrossRefPubMed
17.
Zurück zum Zitat Huang KK, Ramnarayanan K, Zhu F et al (2018) Genomic and epigenomic profiling of high-risk intestinal metaplasia reveals molecular determinants of progression to gastric cancer. Cancer Cell 33(137–150):e5 Huang KK, Ramnarayanan K, Zhu F et al (2018) Genomic and epigenomic profiling of high-risk intestinal metaplasia reveals molecular determinants of progression to gastric cancer. Cancer Cell 33(137–150):e5
18.
Zurück zum Zitat Jiang Y, Qi X, Liu X et al (2017) Fbxw7 haploinsufficiency loses its protection against DNA damage and accelerates MNU-induced gastric carcinogenesis. Oncotarget 8:33444–33456PubMedPubMedCentral Jiang Y, Qi X, Liu X et al (2017) Fbxw7 haploinsufficiency loses its protection against DNA damage and accelerates MNU-induced gastric carcinogenesis. Oncotarget 8:33444–33456PubMedPubMedCentral
19.
Zurück zum Zitat Oue N, Sentani K, Yokozaki H et al (2001) Promoter methylation status of the DNA repair genes hMLH1 and MGMT in gastric carcinoma and metaplastic mucosa. Pathobiology 69:143–149CrossRefPubMed Oue N, Sentani K, Yokozaki H et al (2001) Promoter methylation status of the DNA repair genes hMLH1 and MGMT in gastric carcinoma and metaplastic mucosa. Pathobiology 69:143–149CrossRefPubMed
20.
Zurück zum Zitat Zou XP, Zhang B, Zhang XQ et al (2009) Promoter hypermethylation of multiple genes in early gastric adenocarcinoma and precancerous lesions. Hum Pathol 40:1534–1542CrossRefPubMed Zou XP, Zhang B, Zhang XQ et al (2009) Promoter hypermethylation of multiple genes in early gastric adenocarcinoma and precancerous lesions. Hum Pathol 40:1534–1542CrossRefPubMed
21.
Zurück zum Zitat Oue N, Mitani Y, Aung PP et al (2005) Expression and localization of Reg IV in human neoplastic and non-neoplastic tissues: Reg IV expression is associated with intestinal and neuroendocrine differentiation in gastric adenocarcinoma. J Pathol 207:185–198CrossRefPubMed Oue N, Mitani Y, Aung PP et al (2005) Expression and localization of Reg IV in human neoplastic and non-neoplastic tissues: Reg IV expression is associated with intestinal and neuroendocrine differentiation in gastric adenocarcinoma. J Pathol 207:185–198CrossRefPubMed
22.
Zurück zum Zitat Tatematsu M, Tsukamoto T, Inada K (2003) Stem cells and gastric cancer: role of gastric and intestinal mixed intestinal metaplasia. Cancer Sci 94:135–141CrossRefPubMed Tatematsu M, Tsukamoto T, Inada K (2003) Stem cells and gastric cancer: role of gastric and intestinal mixed intestinal metaplasia. Cancer Sci 94:135–141CrossRefPubMed
23.
Zurück zum Zitat Endoh Y, Tamura G, Ajioka Y et al (2000) Frequent hypermethylation of the hMLH1 gene promoter in differentiated-type tumors of the stomach with the gastric foveolar phenotype. Am J Pathol 157:717–722CrossRefPubMedPubMedCentral Endoh Y, Tamura G, Ajioka Y et al (2000) Frequent hypermethylation of the hMLH1 gene promoter in differentiated-type tumors of the stomach with the gastric foveolar phenotype. Am J Pathol 157:717–722CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Yokozaki H, Shitara Y, Fujimoto J et al (1999) Alterations of p73 preferentially occur in gastric adenocarcinomas with foveolar epithelial phenotype. Int J Cancer 83:192–196CrossRefPubMed Yokozaki H, Shitara Y, Fujimoto J et al (1999) Alterations of p73 preferentially occur in gastric adenocarcinomas with foveolar epithelial phenotype. Int J Cancer 83:192–196CrossRefPubMed
25.
Zurück zum Zitat Endoh Y, Tamura G, Watanabe H et al (1999) The common 18-base pair deletion at codons 418-423 of the E-cadherin gene in differentiated-type adenocarcinomas and intramucosal precancerous lesions of the stomach with the features of gastric foveolar epithelium. J Pathol 189:201–206CrossRefPubMed Endoh Y, Tamura G, Watanabe H et al (1999) The common 18-base pair deletion at codons 418-423 of the E-cadherin gene in differentiated-type adenocarcinomas and intramucosal precancerous lesions of the stomach with the features of gastric foveolar epithelium. J Pathol 189:201–206CrossRefPubMed
26.
Zurück zum Zitat Endoh Y, Sakata K, Tamura G et al (2000) Cellular phenotypes of differentiated-type adenocarcinomas and precancerous lesions of the stomach are dependent on the genetic pathways. J Pathol 191:257–263CrossRefPubMed Endoh Y, Sakata K, Tamura G et al (2000) Cellular phenotypes of differentiated-type adenocarcinomas and precancerous lesions of the stomach are dependent on the genetic pathways. J Pathol 191:257–263CrossRefPubMed
27.
Zurück zum Zitat Wu LB, Kushima R, Borchard F et al (1998) Intramucosal carcinomas of the stomach: phenotypic expression and loss of heterozygosity at microsatellites linked to the APC gene. Pathol Res Pract 194:405–411CrossRefPubMed Wu LB, Kushima R, Borchard F et al (1998) Intramucosal carcinomas of the stomach: phenotypic expression and loss of heterozygosity at microsatellites linked to the APC gene. Pathol Res Pract 194:405–411CrossRefPubMed
28.
Zurück zum Zitat Motoshita J, Oue N, Nakayama H et al (2005) DNA methylation profiles of differentiated-type gastric carcinomas with distinct mucin phenotypes. Cancer Sci 96:474–479CrossRefPubMed Motoshita J, Oue N, Nakayama H et al (2005) DNA methylation profiles of differentiated-type gastric carcinomas with distinct mucin phenotypes. Cancer Sci 96:474–479CrossRefPubMed
29.
Zurück zum Zitat Benahmed F, Gross I, Gaunt SJ, et al (2008) Multiple regulatory regions control the complex expression pattern of the mouse Cdx2 homeobox gene. Gastroenterology 135:1238–1247, 47e1–3 Benahmed F, Gross I, Gaunt SJ, et al (2008) Multiple regulatory regions control the complex expression pattern of the mouse Cdx2 homeobox gene. Gastroenterology 135:1238–1247, 47e1–3
30.
Zurück zum Zitat Hinoi T, Lucas PC, Kuick R et al (2002) CDX2 regulates liver intestine-cadherin expression in normal and malignant colon epithelium and intestinal metaplasia. Gastroenterology 123:1565–1577CrossRefPubMed Hinoi T, Lucas PC, Kuick R et al (2002) CDX2 regulates liver intestine-cadherin expression in normal and malignant colon epithelium and intestinal metaplasia. Gastroenterology 123:1565–1577CrossRefPubMed
31.
32.
Zurück zum Zitat Anami K, Oue N, Noguchi T et al (2010) Search for transmembrane protein in gastric cancer by the Escherichia coli ampicillin secretion trap: expression of DSC2 in gastric cancer with intestinal phenotype. J Pathol 221:275–284CrossRefPubMed Anami K, Oue N, Noguchi T et al (2010) Search for transmembrane protein in gastric cancer by the Escherichia coli ampicillin secretion trap: expression of DSC2 in gastric cancer with intestinal phenotype. J Pathol 221:275–284CrossRefPubMed
33.
Zurück zum Zitat Takakura Y, Hinoi T, Oue N et al (2010) CDX2 regulates multidrug resistance 1 gene expression in malignant intestinal epithelium. Cancer Res 70:6767–6778CrossRefPubMedPubMedCentral Takakura Y, Hinoi T, Oue N et al (2010) CDX2 regulates multidrug resistance 1 gene expression in malignant intestinal epithelium. Cancer Res 70:6767–6778CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Tajima Y, Shimoda T, Nakanishi Y et al (2003) Association of gastric and intestinal phenotypic marker expression of gastric carcinomas with tumor thymidylate synthase expression and response to postoperative chemotherapy with 5-fluorouracil. J Cancer Res Clin Oncol 129:683–690CrossRefPubMed Tajima Y, Shimoda T, Nakanishi Y et al (2003) Association of gastric and intestinal phenotypic marker expression of gastric carcinomas with tumor thymidylate synthase expression and response to postoperative chemotherapy with 5-fluorouracil. J Cancer Res Clin Oncol 129:683–690CrossRefPubMed
35.
Zurück zum Zitat Mitani Y, Oue N, Matsumura S et al (2007) Reg IV is a serum biomarker for gastric cancer patients and predicts response to 5-fluorouracil-based chemotherapy. Oncogene 26:4383–4393CrossRefPubMed Mitani Y, Oue N, Matsumura S et al (2007) Reg IV is a serum biomarker for gastric cancer patients and predicts response to 5-fluorouracil-based chemotherapy. Oncogene 26:4383–4393CrossRefPubMed
36.
Zurück zum Zitat Kobayashi K, Nakahori Y, Miyake M et al (1998) An ancient retrotransposal insertion causes Fukuyama-type congenital muscular dystrophy. Nature 394:388–392CrossRefPubMed Kobayashi K, Nakahori Y, Miyake M et al (1998) An ancient retrotransposal insertion causes Fukuyama-type congenital muscular dystrophy. Nature 394:388–392CrossRefPubMed
37.
Zurück zum Zitat Oo HZ, Sentani K, Mukai S et al (2016) Fukutin, identified by the Escherichia coli ampicillin secretion trap (CAST) method, participates in tumor progression in gastric cancer. Gastric Cancer 19:443–452CrossRefPubMed Oo HZ, Sentani K, Mukai S et al (2016) Fukutin, identified by the Escherichia coli ampicillin secretion trap (CAST) method, participates in tumor progression in gastric cancer. Gastric Cancer 19:443–452CrossRefPubMed
38.
39.
Zurück zum Zitat Johnson M, Sharma M, Henderson BR (2009) IQGAP1 regulation and roles in cancer. Cell Signal 21:1471–1478CrossRefPubMed Johnson M, Sharma M, Henderson BR (2009) IQGAP1 regulation and roles in cancer. Cell Signal 21:1471–1478CrossRefPubMed
41.
Zurück zum Zitat Mataraza JM, Briggs MW, Li Z et al (2003) IQGAP1 promotes cell motility and invasion. J Biol Chem 278:41237–41245CrossRefPubMed Mataraza JM, Briggs MW, Li Z et al (2003) IQGAP1 promotes cell motility and invasion. J Biol Chem 278:41237–41245CrossRefPubMed
42.
Zurück zum Zitat Schmidt VA, Chiariello CS, Capilla E et al (2008) Development of hepatocellular carcinoma in Iqgap2-deficient mice is IQGAP1 dependent. Mol Cell Biol 28:1489–1502CrossRefPubMedPubMedCentral Schmidt VA, Chiariello CS, Capilla E et al (2008) Development of hepatocellular carcinoma in Iqgap2-deficient mice is IQGAP1 dependent. Mol Cell Biol 28:1489–1502CrossRefPubMedPubMedCentral
43.
Zurück zum Zitat Nojima H, Adachi M, Matsui T et al (2008) IQGAP3 regulates cell proliferation through the Ras/ERK signalling cascade. Nat Cell Biol 10:971–978CrossRefPubMed Nojima H, Adachi M, Matsui T et al (2008) IQGAP3 regulates cell proliferation through the Ras/ERK signalling cascade. Nat Cell Biol 10:971–978CrossRefPubMed
44.
Zurück zum Zitat Oue N, Yamamoto Y, Oshima T et al (2018) Overexpression of the transmembrane protein IQGAP3 is associated with poor survival of patients with gastric cancer. Pathobiology 85:192–200CrossRefPubMed Oue N, Yamamoto Y, Oshima T et al (2018) Overexpression of the transmembrane protein IQGAP3 is associated with poor survival of patients with gastric cancer. Pathobiology 85:192–200CrossRefPubMed
45.
Zurück zum Zitat Bessede E, Dubus P, Megraud F et al (2015) Helicobacter pylori infection and stem cells at the origin of gastric cancer. Oncogene 34:2547–2555CrossRefPubMed Bessede E, Dubus P, Megraud F et al (2015) Helicobacter pylori infection and stem cells at the origin of gastric cancer. Oncogene 34:2547–2555CrossRefPubMed
46.
Zurück zum Zitat Wakamatsu Y, Sakamoto N, Oo HZ et al (2012) Expression of cancer stem cell markers ALDH1, CD44 and CD133 in primary tumor and lymph node metastasis of gastric cancer. Pathol Int 62:112–119CrossRefPubMed Wakamatsu Y, Sakamoto N, Oo HZ et al (2012) Expression of cancer stem cell markers ALDH1, CD44 and CD133 in primary tumor and lymph node metastasis of gastric cancer. Pathol Int 62:112–119CrossRefPubMed
47.
48.
Zurück zum Zitat Takaishi S, Okumura T, Tu S et al (2009) Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells 27:1006–1020CrossRefPubMedPubMedCentral Takaishi S, Okumura T, Tu S et al (2009) Identification of gastric cancer stem cells using the cell surface marker CD44. Stem Cells 27:1006–1020CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Oue N, Mukai S, Imai T et al (2016) Induction of KIFC1 expression in gastric cancer spheroids. Oncol Rep 36:349–355CrossRefPubMed Oue N, Mukai S, Imai T et al (2016) Induction of KIFC1 expression in gastric cancer spheroids. Oncol Rep 36:349–355CrossRefPubMed
51.
Zurück zum Zitat Imai T, Oue N, Nishioka M et al (2017) Overexpression of KIF11 in gastric cancer with intestinal mucin phenotype. Pathobiology 84:16–24CrossRefPubMed Imai T, Oue N, Nishioka M et al (2017) Overexpression of KIF11 in gastric cancer with intestinal mucin phenotype. Pathobiology 84:16–24CrossRefPubMed
52.
Zurück zum Zitat Zhang S, Huang F, Wang Y et al (2016) KIF2A Overexpression and its association with clinicopathologic characteristics and poor prognoses in patients with gastric cancer. Dis Markers 2016:7484516PubMedPubMedCentral Zhang S, Huang F, Wang Y et al (2016) KIF2A Overexpression and its association with clinicopathologic characteristics and poor prognoses in patients with gastric cancer. Dis Markers 2016:7484516PubMedPubMedCentral
53.
Zurück zum Zitat Zhang H, Ma RR, Wang XJ et al (2017) KIF26B, a novel oncogene, promotes proliferation and metastasis by activating the VEGF pathway in gastric cancer. Oncogene 36:5609–5619CrossRefPubMed Zhang H, Ma RR, Wang XJ et al (2017) KIF26B, a novel oncogene, promotes proliferation and metastasis by activating the VEGF pathway in gastric cancer. Oncogene 36:5609–5619CrossRefPubMed
54.
Zurück zum Zitat Weil D, Garcon L, Harper M et al (2002) Targeting the kinesin Eg5 to monitor siRNA transfection in mammalian cells. Biotechniques 33:1244–1248CrossRefPubMed Weil D, Garcon L, Harper M et al (2002) Targeting the kinesin Eg5 to monitor siRNA transfection in mammalian cells. Biotechniques 33:1244–1248CrossRefPubMed
55.
56.
Zurück zum Zitat Ando A, Kikuti YY, Kawata H et al (1994) Cloning of a new kinesin-related gene located at the centromeric end of the human MHC region. Immunogenetics 39:194–200CrossRefPubMed Ando A, Kikuti YY, Kawata H et al (1994) Cloning of a new kinesin-related gene located at the centromeric end of the human MHC region. Immunogenetics 39:194–200CrossRefPubMed
57.
Zurück zum Zitat DeLuca JG, Newton CN, Himes RH et al (2001) Purification and characterization of native conventional kinesin, HSET, and CENP-E from mitotic hela cells. J Biol Chem 276:28014–28021CrossRefPubMed DeLuca JG, Newton CN, Himes RH et al (2001) Purification and characterization of native conventional kinesin, HSET, and CENP-E from mitotic hela cells. J Biol Chem 276:28014–28021CrossRefPubMed
58.
Zurück zum Zitat Sekino Y, Oue N, Koike Y et al (2019) KIFC1 inhibitor CW069 induces apoptosis and reverses resistance to docetaxel in prostate cancer. J Clin Med 8:225CrossRefPubMedCentral Sekino Y, Oue N, Koike Y et al (2019) KIFC1 inhibitor CW069 induces apoptosis and reverses resistance to docetaxel in prostate cancer. J Clin Med 8:225CrossRefPubMedCentral
59.
Zurück zum Zitat Krzysiak TC, Grabe M, Gilbert SP (2008) Getting in sync with dimeric Eg5. Initiation and regulation of the processive run. J Biol Chem 283:2078–2087CrossRefPubMed Krzysiak TC, Grabe M, Gilbert SP (2008) Getting in sync with dimeric Eg5. Initiation and regulation of the processive run. J Biol Chem 283:2078–2087CrossRefPubMed
60.
Zurück zum Zitat LoRusso PM, Goncalves PH, Casetta L et al (2015) First-in-human phase 1 study of filanesib (ARRY-520), a kinesin spindle protein inhibitor, in patients with advanced solid tumors. Invest New Drugs 33:440–449CrossRefPubMed LoRusso PM, Goncalves PH, Casetta L et al (2015) First-in-human phase 1 study of filanesib (ARRY-520), a kinesin spindle protein inhibitor, in patients with advanced solid tumors. Invest New Drugs 33:440–449CrossRefPubMed
61.
Zurück zum Zitat Network CGAR (2014) Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513:202–209CrossRef Network CGAR (2014) Comprehensive molecular characterization of gastric adenocarcinoma. Nature 513:202–209CrossRef
62.
Zurück zum Zitat Cristescu R, Lee J, Nebozhyn M et al (2015) Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med 21:449–456CrossRefPubMed Cristescu R, Lee J, Nebozhyn M et al (2015) Molecular analysis of gastric cancer identifies subtypes associated with distinct clinical outcomes. Nat Med 21:449–456CrossRefPubMed
63.
Zurück zum Zitat Saito R, Abe H, Kunita A et al (2017) Overexpression and gene amplification of PD-L1 in cancer cells and PD-L1(+) immune cells in Epstein-Barr virus-associated gastric cancer: the prognostic implications. Mod Pathol 30:427–439CrossRefPubMed Saito R, Abe H, Kunita A et al (2017) Overexpression and gene amplification of PD-L1 in cancer cells and PD-L1(+) immune cells in Epstein-Barr virus-associated gastric cancer: the prognostic implications. Mod Pathol 30:427–439CrossRefPubMed
64.
Zurück zum Zitat Tan P, Yeoh KG (2015) Genetics and molecular pathogenesis of gastric adenocarcinoma. Gastroenterology 149:1153–1162CrossRefPubMed Tan P, Yeoh KG (2015) Genetics and molecular pathogenesis of gastric adenocarcinoma. Gastroenterology 149:1153–1162CrossRefPubMed
65.
Zurück zum Zitat Forbes SA, Beare D, Gunasekaran P et al (2015) COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res 43:D805–D811CrossRefPubMed Forbes SA, Beare D, Gunasekaran P et al (2015) COSMIC: exploring the world’s knowledge of somatic mutations in human cancer. Nucleic Acids Res 43:D805–D811CrossRefPubMed
66.
Zurück zum Zitat Wang K, Yuen ST, Xu J et al (2014) Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet 46:573–582CrossRefPubMed Wang K, Yuen ST, Xu J et al (2014) Whole-genome sequencing and comprehensive molecular profiling identify new driver mutations in gastric cancer. Nat Genet 46:573–582CrossRefPubMed
67.
Zurück zum Zitat Bang YJ, Van Cutsem E, Feyereislova A et al (2010) Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376:687–697CrossRefPubMed Bang YJ, Van Cutsem E, Feyereislova A et al (2010) Trastuzumab in combination with chemotherapy versus chemotherapy alone for treatment of HER2-positive advanced gastric or gastro-oesophageal junction cancer (ToGA): a phase 3, open-label, randomised controlled trial. Lancet 376:687–697CrossRefPubMed
68.
Zurück zum Zitat Wilke H, Muro K, Van Cutsem E et al (2014) Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 15:1224–1235CrossRefPubMed Wilke H, Muro K, Van Cutsem E et al (2014) Ramucirumab plus paclitaxel versus placebo plus paclitaxel in patients with previously treated advanced gastric or gastro-oesophageal junction adenocarcinoma (RAINBOW): a double-blind, randomised phase 3 trial. Lancet Oncol 15:1224–1235CrossRefPubMed
69.
Zurück zum Zitat Wang KL, Wu TT, Choi IS et al (2007) Expression of epidermal growth factor receptor in esophageal and esophagogastric junction adenocarcinomas: association with poor outcome. Cancer 109:658–667CrossRefPubMed Wang KL, Wu TT, Choi IS et al (2007) Expression of epidermal growth factor receptor in esophageal and esophagogastric junction adenocarcinomas: association with poor outcome. Cancer 109:658–667CrossRefPubMed
70.
Zurück zum Zitat Lordick F, Kang YK, Chung HC et al (2013) Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol 14:490–499CrossRefPubMed Lordick F, Kang YK, Chung HC et al (2013) Capecitabine and cisplatin with or without cetuximab for patients with previously untreated advanced gastric cancer (EXPAND): a randomised, open-label phase 3 trial. Lancet Oncol 14:490–499CrossRefPubMed
71.
Zurück zum Zitat Nakajima M, Sawada H, Yamada Y et al (1999) The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer 85:1894–1902CrossRefPubMed Nakajima M, Sawada H, Yamada Y et al (1999) The prognostic significance of amplification and overexpression of c-met and c-erb B-2 in human gastric carcinomas. Cancer 85:1894–1902CrossRefPubMed
72.
Metadaten
Titel
Molecular carcinogenesis of gastric cancer: Lauren classification, mucin phenotype expression, and cancer stem cells
verfasst von
Naohide Oue
Kazuhiro Sentani
Naoya Sakamoto
Naohiro Uraoka
Wataru Yasui
Publikationsdatum
12.04.2019
Verlag
Springer Singapore
Erschienen in
International Journal of Clinical Oncology / Ausgabe 7/2019
Print ISSN: 1341-9625
Elektronische ISSN: 1437-7772
DOI
https://doi.org/10.1007/s10147-019-01443-9

Weitere Artikel der Ausgabe 7/2019

International Journal of Clinical Oncology 7/2019 Zur Ausgabe

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.