#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Integrated Genomic Characterization Reveals Novel, Therapeutically Relevant Drug Targets in FGFR and EGFR Pathways in Sporadic Intrahepatic Cholangiocarcinoma


Advanced cholangiocarcinoma continues to harbor a difficult prognosis and therapeutic options have been limited. During the course of a clinical trial of whole genomic sequencing seeking druggable targets, we examined six patients with advanced cholangiocarcinoma. Integrated genome-wide and whole transcriptome sequence analyses were performed on tumors from six patients with advanced, sporadic intrahepatic cholangiocarcinoma (SIC) to identify potential therapeutically actionable events. Among the somatic events captured in our analysis, we uncovered two novel therapeutically relevant genomic contexts that when acted upon, resulted in preliminary evidence of anti-tumor activity. Genome-wide structural analysis of sequence data revealed recurrent translocation events involving the FGFR2 locus in three of six assessed patients. These observations and supporting evidence triggered the use of FGFR inhibitors in these patients. In one example, preliminary anti-tumor activity of pazopanib (in vitro FGFR2 IC50≈350 nM) was noted in a patient with an FGFR2-TACC3 fusion. After progression on pazopanib, the same patient also had stable disease on ponatinib, a pan-FGFR inhibitor (in vitro, FGFR2 IC50≈8 nM). In an independent non-FGFR2 translocation patient, exome and transcriptome analysis revealed an allele specific somatic nonsense mutation (E384X) in ERRFI1, a direct negative regulator of EGFR activation. Rapid and robust disease regression was noted in this ERRFI1 inactivated tumor when treated with erlotinib, an EGFR kinase inhibitor. FGFR2 fusions and ERRFI mutations may represent novel targets in sporadic intrahepatic cholangiocarcinoma and trials should be characterized in larger cohorts of patients with these aberrations.


Vyšlo v časopise: Integrated Genomic Characterization Reveals Novel, Therapeutically Relevant Drug Targets in FGFR and EGFR Pathways in Sporadic Intrahepatic Cholangiocarcinoma. PLoS Genet 10(2): e32767. doi:10.1371/journal.pgen.1004135
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004135

Souhrn

Advanced cholangiocarcinoma continues to harbor a difficult prognosis and therapeutic options have been limited. During the course of a clinical trial of whole genomic sequencing seeking druggable targets, we examined six patients with advanced cholangiocarcinoma. Integrated genome-wide and whole transcriptome sequence analyses were performed on tumors from six patients with advanced, sporadic intrahepatic cholangiocarcinoma (SIC) to identify potential therapeutically actionable events. Among the somatic events captured in our analysis, we uncovered two novel therapeutically relevant genomic contexts that when acted upon, resulted in preliminary evidence of anti-tumor activity. Genome-wide structural analysis of sequence data revealed recurrent translocation events involving the FGFR2 locus in three of six assessed patients. These observations and supporting evidence triggered the use of FGFR inhibitors in these patients. In one example, preliminary anti-tumor activity of pazopanib (in vitro FGFR2 IC50≈350 nM) was noted in a patient with an FGFR2-TACC3 fusion. After progression on pazopanib, the same patient also had stable disease on ponatinib, a pan-FGFR inhibitor (in vitro, FGFR2 IC50≈8 nM). In an independent non-FGFR2 translocation patient, exome and transcriptome analysis revealed an allele specific somatic nonsense mutation (E384X) in ERRFI1, a direct negative regulator of EGFR activation. Rapid and robust disease regression was noted in this ERRFI1 inactivated tumor when treated with erlotinib, an EGFR kinase inhibitor. FGFR2 fusions and ERRFI mutations may represent novel targets in sporadic intrahepatic cholangiocarcinoma and trials should be characterized in larger cohorts of patients with these aberrations.


Zdroje

1. ShinHR, LeeCU, ParkHJ, SeolSY, ChungJM, et al. (1996) Hepatitis B and C virus, Clonorchis sinensis for the risk of liver cancer: a case-control study in Pusan, Korea. International journal of epidemiology 25: 933–940.

2. WatanapaP (1996) Cholangiocarcinoma in patients with opisthorchiasis. The British journal of surgery 83: 1062–1064.

3. WatanapaP, WatanapaWB (2002) Liver fluke-associated cholangiocarcinoma. The British journal of surgery 89: 962–970.

4. BergquistA, EkbomA, OlssonR, KornfeldtD, LoofL, et al. (2002) Hepatic and extrahepatic malignancies in primary sclerosing cholangitis. Journal of hepatology 36: 321–327.

5. BergquistA, GlaumannH, PerssonB, BroomeU (1998) Risk factors and clinical presentation of hepatobiliary carcinoma in patients with primary sclerosing cholangitis: a case-control study. Hepatology 27: 311–316.

6. BurakK, AnguloP, PashaTM, EganK, PetzJ, et al. (2004) Incidence and risk factors for cholangiocarcinoma in primary sclerosing cholangitis. The American journal of gastroenterology 99: 523–526.

7. ClaessenMM, VleggaarFP, TytgatKM, SiersemaPD, van BuurenHR (2009) High lifetime risk of cancer in primary sclerosing cholangitis. Journal of hepatology 50: 158–164.

8. VisserBC, SuhI, WayLW, KangSM (2004) Congenital choledochal cysts in adults. Archives of surgery 139: 855–860 discussion 860–852.

9. HsingAW, ZhangM, RashidA, McGlynnKA, WangBS, et al. (2008) Hepatitis B and C virus infection and the risk of biliary tract cancer: a population-based study in China. International journal of cancer Journal international du cancer 122: 1849–1853.

10. KobayashiM, IkedaK, SaitohS, SuzukiF, TsubotaA, et al. (2000) Incidence of primary cholangiocellular carcinoma of the liver in japanese patients with hepatitis C virus-related cirrhosis. Cancer 88: 2471–2477.

11. LiuXF, ZouSQ, QiuFZ (2003) Pathogenesis of cholangiocarcinoma in the porta hepatis and infection of hepatitis virus. Hepatobiliary & pancreatic diseases international : HBPD INT 2: 285–289.

12. ShaibYH, El-SeragHB, DavilaJA, MorganR, McGlynnKA (2005) Risk factors of intrahepatic cholangiocarcinoma in the United States: a case-control study. Gastroenterology 128: 620–626.

13. WelzelTM, GraubardBI, El-SeragHB, ShaibYH, HsingAW, et al. (2007) Risk factors for intrahepatic and extrahepatic cholangiocarcinoma in the United States: a population-based case-control study. Clinical gastroenterology and hepatology : the official clinical practice journal of the American Gastroenterological Association 5: 1221–1228.

14. YamamotoS, KuboS, HaiS, UenishiT, YamamotoT, et al. (2004) Hepatitis C virus infection as a likely etiology of intrahepatic cholangiocarcinoma. Cancer science 95: 592–595.

15. DonatoF, GelattiU, TaggerA, FavretM, RiberoML, et al. (2001) Intrahepatic cholangiocarcinoma and hepatitis C and B virus infection, alcohol intake, and hepatolithiasis: a case-control study in Italy. Cancer causes & control : CCC 12: 959–964.

16. LeeCC, WuCY, ChenGH (2002) What is the impact of coexistence of hepatolithiasis on cholangiocarcinoma? Journal of gastroenterology and hepatology 17: 1015–1020.

17. BeckerN, LiebermannD, WeschH, Van KaickG (2008) Mortality among Thorotrast-exposed patients and an unexposed comparison group in the German Thorotrast study. European journal of cancer 44: 1259–1268.

18. TravisLB, HauptmannM, GaulLK, StormHH, GoldmanMB, et al. (2003) Site-specific cancer incidence and mortality after cerebral angiography with radioactive thorotrast. Radiation research 160: 691–706.

19. KhanSA, ThomasHC, DavidsonBR, Taylor-RobinsonSD (2005) Cholangiocarcinoma. Lancet 366: 1303–1314.

20. ValleJ, WasanH, PalmerDH, CunninghamD, AnthoneyA, et al. (2010) Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. The New England journal of medicine 362: 1273–1281.

21. KibaT, TsudaH, PairojkulC, InoueS, SugimuraT, et al. (1993) Mutations of the p53 tumor suppressor gene and the ras gene family in intrahepatic cholangiocellular carcinomas in Japan and Thailand. Molecular carcinogenesis 8: 312–318.

22. OhashiK, TsutsumiM, NakajimaY, NoguchiO, OkitaS, et al. (1994) High rates of Ki-ras point mutation in both intra- and extra-hepatic cholangiocarcinomas. Japanese journal of clinical oncology 24: 305–310.

23. PetmitrS, PinlaorS, ThousungnoenA, KaralakA, MigasenaP (1998) K-ras oncogene and p53 gene mutations in cholangiocarcinoma from Thai patients. The Southeast Asian journal of tropical medicine and public health 29: 71–75.

24. RoaJC, RoaI, de AretxabalaX, MeloA, FariaG, et al. (2004) [K-ras gene mutation in gallbladder carcinoma]. Revista medica de Chile 132: 955–960.

25. SturmPD, BaasIO, ClementMJ, NakeebA, JohanG, et al. (1998) Alterations of the p53 tumor-suppressor gene and K-ras oncogene in perihilar cholangiocarcinomas from a high-incidence area. International journal of cancer Journal international du cancer 78: 695–698.

26. TadaM, OmataM, OhtoM (1990) Analysis of ras gene mutations in human hepatic malignant tumors by polymerase chain reaction and direct sequencing. Cancer research 50: 1121–1124.

27. TadaM, OmataM, OhtoM (1990) [Detection of ras gene mutations of primary hepatic malignant tumors by polymerase chain reaction and direct sequencing method]. Nihon Shokakibyo Gakkai zasshi = The Japanese journal of gastro-enterology 87: 811–815.

28. TadaM, OmataM, OhtoM (1992) High incidence of ras gene mutation in intrahepatic cholangiocarcinoma. Cancer 69: 1115–1118.

29. WattanasirichaigoonS, TasanakhajornU, JesadapatarakulS (1998) The incidence of K-ras codon 12 mutations in cholangiocarcinoma detected by polymerase chain reaction technique. Journal of the Medical Association of Thailand = Chotmaihet thangphaet 81: 316–323.

30. BobergKM, SchrumpfE, BergquistA, BroomeU, ParesA, et al. (2000) Cholangiocarcinoma in primary sclerosing cholangitis: K-ras mutations and Tp53 dysfunction are implicated in the neoplastic development. Journal of hepatology 32: 374–380.

31. TannapfelA, WeinansL, GeisslerF, SchutzA, KatalinicA, et al. (2000) Mutations of p53 tumor suppressor gene, apoptosis, and proliferation in intrahepatic cholangiocellular carcinoma of the liver. Digestive diseases and sciences 45: 317–324.

32. TulloA, D'ErchiaAM, HondaK, KellyMD, HabibNA, et al. (2000) New p53 mutations in hilar cholangiocarcinoma. European journal of clinical investigation 30: 798–803.

33. HahnSA, BartschD, SchroersA, GalehdariH, BeckerM, et al. (1998) Mutations of the DPC4/Smad4 gene in biliary tract carcinoma. Cancer research 58: 1124–1126.

34. Iacobuzio-DonahueCA, SongJ, ParmiagianiG, YeoCJ, HrubanRH, et al. (2004) Missense mutations of MADH4: characterization of the mutational hot spot and functional consequences in human tumors. Clinical cancer research : an official journal of the American Association for Cancer Research 10: 1597–1604.

35. AhrendtSA, EisenbergerCF, YipL, RashidA, ChowJT, et al. (1999) Chromosome 9p21 loss and p16 inactivation in primary sclerosing cholangitis-associated cholangiocarcinoma. The Journal of surgical research 84: 88–93.

36. TadokoroH, ShigiharaT, IkedaT, TakaseM, SuyamaM (2007) Two distinct pathways of p16 gene inactivation in gallbladder cancer. World journal of gastroenterology : WJG 13: 6396–6403.

37. TaniaiM, HiguchiH, BurgartLJ, GoresGJ (2002) p16INK4a promoter mutations are frequent in primary sclerosing cholangitis (PSC) and PSC-associated cholangiocarcinoma. Gastroenterology 123: 1090–1098.

38. TannapfelA, BenickeM, KatalinicA, UhlmannD, KockerlingF, et al. (2000) Frequency of p16(INK4A) alterations and K-ras mutations in intrahepatic cholangiocarcinoma of the liver. Gut 47: 721–727.

39. YoshidaS, TodorokiT, IchikawaY, HanaiS, SuzukiH, et al. (1995) Mutations of p16Ink4/CDKN2 and p15Ink4B/MTS2 genes in biliary tract cancers. Cancer research 55: 2756–2760.

40. BorgerDR, TanabeKK, FanKC, LopezHU, FantinVR, et al. (2012) Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. The oncologist 17: 72–79.

41. KippBR, VossJS, KerrSE, Barr FritcherEG, GrahamRP, et al. (2012) Isocitrate dehydrogenase 1 and 2 mutations in cholangiocarcinoma. Human pathology 43: 1552–1558.

42. GuTL, DengX, HuangF, TuckerM, CrosbyK, et al. (2011) Survey of tyrosine kinase signaling reveals ROS kinase fusions in human cholangiocarcinoma. PloS one 6: e15640.

43. GwakGY, YoonJH, ShinCM, AhnYJ, ChungJK, et al. (2005) Detection of response-predicting mutations in the kinase domain of the epidermal growth factor receptor gene in cholangiocarcinomas. Journal of cancer research and clinical oncology 131: 649–652.

44. LeoneF, CavalloniG, PignochinoY, SarottoI, FerrarisR, et al. (2006) Somatic mutations of epidermal growth factor receptor in bile duct and gallbladder carcinoma. Clinical cancer research : an official journal of the American Association for Cancer Research 12: 1680–1685.

45. TannapfelA, SommererF, BenickeM, KatalinicA, UhlmannD, et al. (2003) Mutations of the BRAF gene in cholangiocarcinoma but not in hepatocellular carcinoma. Gut 52: 706–712.

46. DeshpandeV, NduagubaA, ZimmermanSM, KehoeSM, MacconaillLE, et al. (2011) Mutational profiling reveals PIK3CA mutations in gallbladder carcinoma. BMC cancer 11: 60.

47. RienerMO, BawohlM, ClavienPA, JochumW (2008) Rare PIK3CA hotspot mutations in carcinomas of the biliary tract. Genes, chromosomes & cancer 47: 363–367.

48. WendumD, BarbuV, RosmorducO, ArriveL, FlejouJF, et al. (2012) Aspects of liver pathology in adult patients with MDR3/ABCB4 gene mutations. Virchows Archiv : an international journal of pathology 460: 291–298.

49. ScheimannAO, StrautnieksSS, KniselyAS, ByrneJA, ThompsonRJ, et al. (2007) Mutations in bile salt export pump (ABCB11) in two children with progressive familial intrahepatic cholestasis and cholangiocarcinoma. The Journal of pediatrics 150: 556–559.

50. StrautnieksSS, ByrneJA, PawlikowskaL, CebecauerovaD, RaynerA, et al. (2008) Severe bile salt export pump deficiency: 82 different ABCB11 mutations in 109 families. Gastroenterology 134: 1203–1214.

51. EndoK, AshidaK, MiyakeN, TeradaT (2001) E-cadherin gene mutations in human intrahepatic cholangiocarcinoma. The Journal of pathology 193: 310–317.

52. UkitaY, KatoM, TeradaT (2002) Gene amplification and mRNA and protein overexpression of c-erbB-2 (HER-2/neu) in human intrahepatic cholangiocarcinoma as detected by fluorescence in situ hybridization, in situ hybridization, and immunohistochemistry. Journal of hepatology 36: 780–785.

53. AraiY, TotokiY, HosodaF, ShirotaT, HamaN, et al. (2013) FGFR2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology 2013 Oct 9 doi:[]10.1002/hep.26890. [Epub ahead of print]

54. WuYM, SuF, Kalyana-SundaramS, KhazanovN, AteeqB, et al. (2013) Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discov 3: 636–647.

55. NgPC, HenikoffS (2001) Predicting deleterious amino acid substitutions. Genome Res 11: 863–874.

56. AdzhubeiIA, SchmidtS, PeshkinL, RamenskyVE, GerasimovaA, et al. (2010) A method and server for predicting damaging missense mutations. Nat Methods 7: 248–249.

57. RevaB, AntipinY, SanderC (2007) Determinants of protein function revealed by combinatorial entropy optimization. Genome Biol 8: R232.

58. SchwarzJM, RodelspergerC, SchuelkeM, SeelowD (2010) MutationTaster evaluates disease-causing potential of sequence alterations. Nat Methods 7: 575–576.

59. CraigDW, O'ShaughnessyJA, KieferJA, AldrichJ, SinariS, et al. (2013) Genome and transcriptome sequencing in prospective metastatic triple-negative breast cancer uncovers therapeutic vulnerabilities. Mol Cancer Ther 12: 104–116.

60. CirulliV, YebraM (2007) Netrins: beyond the brain. Nat Rev Mol Cell Biol 8: 296–306.

61. QiJH, EbrahemQ, MooreN, MurphyG, Claesson-WelshL, et al. (2003) A novel function for tissue inhibitor of metalloproteinases-3 (TIMP3): inhibition of angiogenesis by blockage of VEGF binding to VEGF receptor-2. Nat Med 9: 407–415.

62. SalvucciO, TosatoG (2012) Essential roles of EphB receptors and EphrinB ligands in endothelial cell function and angiogenesis. Adv Cancer Res 114: 21–57.

63. CavallaroU, ChristoforiG (2004) Cell adhesion and signalling by cadherins and Ig-CAMs in cancer. Nat Rev Cancer 4: 118–132.

64. KrausMR, ClauinS, PfisterY, Di MaioM, UlinskiT, et al. (2012) Two mutations in human BICC1 resulting in Wnt pathway hyperactivity associated with cystic renal dysplasia. Hum Mutat 33: 86–90.

65. WangX, OsadaT, WangY, YuL, SakakuraK, et al. (2010) CSPG4 protein as a new target for the antibody-based immunotherapy of triple-negative breast cancer. J Natl Cancer Inst 102: 1496–1512.

66. KrzeslakA, FormaE, BernaciakM, RomanowiczH, BrysM (2012) Gene expression of O-GlcNAc cycling enzymes in human breast cancers. Clin Exp Med 12: 61–65.

67. AntonescuCR, ZhangL, NielsenGP, RosenbergAE, Dal CinP, et al. (2011) Consistent t(1;10) with rearrangements of TGFBR3 and MGEA5 in both myxoinflammatory fibroblastic sarcoma and hemosiderotic fibrolipomatous tumor. Genes Chromosomes Cancer 50: 757–764.

68. HallorKH, SciotR, StaafJ, HeidenbladM, RydholmA, et al. (2009) Two genetic pathways, t(1;10) and amplification of 3p11-12, in myxoinflammatory fibroblastic sarcoma, haemosiderotic fibrolipomatous tumour, and morphologically similar lesions. J Pathol 217: 716–727.

69. KumarR, CrouthamelMC, RomingerDH, GontarekRR, TumminoPJ, et al. (2009) Myelosuppression and kinase selectivity of multikinase angiogenesis inhibitors. Br J Cancer 101: 1717–1723.

70. HaGH, ParkJS, BreuerEK (2013) TACC3 promotes epithelial-mesenchymal transition (EMT) through the activation of PI3K/Akt and ERK signaling pathways. Cancer Lett 332: 63–73.

71. TurnerN, GroseR (2010) Fibroblast growth factor signalling: from development to cancer. Nature reviews Cancer 10: 116–129.

72. WilliamsSV, HurstCD, KnowlesMA (2012) Oncogenic FGFR3 gene fusions in bladder cancer. Human molecular genetics. Hum Mol Genet 22: 795–803.

73. MaedaT, YagasakiF, IshikawaM, TakahashiN, BesshoM (2005) Transforming property of TEL-FGFR3 mediated through PI3-K in a T-cell lymphoma that subsequently progressed to AML. Blood 105: 2115–2123.

74. RenM, QinH, RenR, TidwellJ, CowellJK (2011) Src activation plays an important key role in lymphomagenesis induced by FGFR1 fusion kinases. Cancer research 71: 7312–7322.

75. Ren M, Qin H, Ren R, Cowell JK (2012) Ponatinib suppresses the development of myeloid and lymphoid malignancies associated with FGFR1 abnormalities. Leukemia: official journal of the Leukemia Society of America, Leukemia Research Fund, UK.

76. RicheldaR, RonchettiD, BaldiniL, CroL, ViggianoL, et al. (1997) A novel chromosomal translocation t(4; 14)(p16.3; q32) in multiple myeloma involves the fibroblast growth-factor receptor 3 gene. Blood 90: 4062–4070.

77. CrossNC, ReiterA (2008) Fibroblast growth factor receptor and platelet-derived growth factor receptor abnormalities in eosinophilic myeloproliferative disorders. Acta haematologica 119: 199–206.

78. SinghD, ChanJM, ZoppoliP, NiolaF, SullivanR, et al. (2012) Transforming fusions of FGFR and TACC genes in human glioblastoma. Science 337: 1231–1235.

79. WangP, DongQ, ZhangC, KuanPF, LiuY, et al. (2012) Mutations in isocitrate dehydrogenase 1 and 2 occur frequently in intrahepatic cholangiocarcinomas and share hypermethylation targets with glioblastomas. Oncogene 32: 3091–100.

80. MaisonneuveC, GuilleretI, VickP, WeberT, AndreP, et al. (2009) Bicaudal C, a novel regulator of Dvl signaling abutting RNA-processing bodies, controls cilia orientation and leftward flow. Development 136: 3019–3030.

81. PetiotA, ContiFJ, GroseR, RevestJM, Hodivala-DilkeKM, et al. (2003) A crucial role for Fgfr2-IIIb signalling in epidermal development and hair follicle patterning. Development 130: 5493–5501.

82. FengS, WangF, MatsubaraA, KanM, McKeehanWL (1997) Fibroblast growth factor receptor 2 limits and receptor 1 accelerates tumorigenicity of prostate epithelial cells. Cancer Res 57: 5369–5378.

83. RicolD, CappellenD, El MarjouA, Gil-Diez-de-MedinaS, GiraultJM, et al. (1999) Tumour suppressive properties of fibroblast growth factor receptor 2-IIIb in human bladder cancer. Oncogene 18: 7234–7243.

84. LorenziMV, CastagninoP, ChenQ, ChedidM, MikiT (1997) Ligand-independent activation of fibroblast growth factor receptor-2 by carboxyl terminal alterations. Oncogene 15: 817–826.

85. ChaseA, GrandFH, CrossNC (2007) Activity of TKI258 against primary cells and cell lines with FGFR1 fusion genes associated with the 8p11 myeloproliferative syndrome. Blood 110: 3729–3734.

86. GuagnanoV, KauffmannA, WohrleS, StammC, ItoM, et al. (2012) FGFR Genetic Alterations Predict for Sensitivity to NVP-BGJ398, a Selective Pan-FGFR Inhibitor. Cancer discovery 2: 1118–1133.

87. GozgitJM, WongMJ, MoranL, WardwellS, MohemmadQK, et al. (2012) Ponatinib (AP24534), a multitargeted pan-FGFR inhibitor with activity in multiple FGFR-amplified or mutated cancer models. Molecular cancer therapeutics 11: 690–699.

88. AmanoR, YamadaN, DoiY, YashiroM, OhiraM, et al. (2010) Significance of keratinocyte growth factor receptor in the proliferation of biliary tract cancer. Anticancer Res 30: 4115–4121.

89. KumarR, KnickVB, RudolphSK, JohnsonJH, CrosbyRM, et al. (2007) Pharmacokinetic-pharmacodynamic correlation from mouse to human with pazopanib, a multikinase angiogenesis inhibitor with potent antitumor and antiangiogenic activity. Mol Cancer Ther 6: 2012–2021.

90. BallaroC, CeccarelliS, TiveronC, TatangeloL, SalvatoreAM, et al. (2005) Targeted expression of RALT in mouse skin inhibits epidermal growth factor receptor signalling and generates a Waved-like phenotype. EMBO reports 6: 755–761.

91. FerbyI, ReschkeM, KudlacekO, KnyazevP, PanteG, et al. (2006) Mig6 is a negative regulator of EGF receptor-mediated skin morphogenesis and tumor formation. Nature medicine 12: 568–573.

92. JeongJW, LeeHS, LeeKY, WhiteLD, BroaddusRR, et al. (2009) Mig-6 modulates uterine steroid hormone responsiveness and exhibits altered expression in endometrial disease. Proceedings of the National Academy of Sciences of the United States of America 106: 8677–8682.

93. LinCI, DuJ, ShenWT, WhangEE, DonnerDB, et al. (2011) Mitogen-inducible gene-6 is a multifunctional adaptor protein with tumor suppressor-like activity in papillary thyroid cancer. The Journal of clinical endocrinology and metabolism 96: E554–565.

94. ReschkeM, FerbyI, StepniakE, SeitzerN, HorstD, et al. (2010) Mitogen-inducible gene-6 is a negative regulator of epidermal growth factor receptor signaling in hepatocytes and human hepatocellular carcinoma. Hepatology 51: 1383–1390.

95. AnastasiS, FiorentinoL, FioriniM, FraioliR, SalaG, et al. (2003) Feedback inhibition by RALT controls signal output by the ErbB network. Oncogene 22: 4221–4234.

96. ZhangX, PickinKA, BoseR, JuraN, ColePA, et al. (2007) Inhibition of the EGF receptor by binding of MIG6 to an activating kinase domain interface. Nature 450: 741–744.

97. FrosiY, AnastasiS, BallaroC, VarsanoG, CastellaniL, et al. (2010) A two-tiered mechanism of EGFR inhibition by RALT/MIG6 via kinase suppression and receptor degradation. The Journal of cell biology 189: 557–571.

98. AnastasiS, SalaG, HuipingC, CapriniE, RussoG, et al. (2005) Loss of RALT/MIG-6 expression in ERBB2-amplified breast carcinomas enhances ErbB-2 oncogenic potency and favors resistance to Herceptin. Oncogene 24: 4540–4548.

99. DuncanCG, KillelaPJ, PayneCA, LampsonB, ChenWC, et al. (2010) Integrated genomic analyses identify ERRFI1 and TACC3 as glioblastoma-targeted genes. Oncotarget 1: 265–277.

100. YingH, ZhengH, ScottK, WiedemeyerR, YanH, et al. (2010) Mig-6 controls EGFR trafficking and suppresses gliomagenesis. Proceedings of the National Academy of Sciences of the United States of America 107: 6912–6917.

101. LiZ, DongQ, WangY, QuL, QiuX, et al. (2012) Downregulation of Mig-6 in nonsmall-cell lung cancer is associated with EGFR signaling. Molecular carcinogenesis 51: 522–534.

102. ZhangYW, StaalB, DykemaKJ, FurgeKA, Vande WoudeGF (2012) Cancer-type regulation of MIG-6 expression by inhibitors of methylation and histone deacetylation. PloS one 7: e38955.

103. ZhangYW, StaalB, SuY, SwiatekP, ZhaoP, et al. (2007) Evidence that MIG-6 is a tumor-suppressor gene. Oncogene 26: 269–276.

104. BaiA, MeetzeK, VoNY, KolliparaS, MazsaEK, et al. (2010) GP369, an FGFR2-IIIb-specific antibody, exhibits potent antitumor activity against human cancers driven by activated FGFR2 signaling. Cancer Res 70: 7630–7639.

105. ChristoforidesA, CarptenJD, WeissGJ, DemeureMJ, Von HoffDD, et al. (2013) Identification of somatic mutations in cancer through Bayesian-based analysis of sequenced genome pairs. BMC Genomics 14: 302.

106. KimD, SalzbergSL (2011) TopHat-Fusion: an algorithm for discovery of novel fusion transcripts. Genome Biol 12: R72.

107. IyerMK, ChinnaiyanAM, MaherCA (2011) ChimeraScan: a tool for identifying chimeric transcription in sequencing data. Bioinformatics 27: 2903–2904.

108. McPhersonA, HormozdiariF, ZayedA, GiulianyR, HaG, et al. (2011) deFuse: an algorithm for gene fusion discovery in tumor RNA-Seq data. PLoS Comput Biol 7: e1001138.

109. AsmannYW, HossainA, NecelaBM, MiddhaS, KalariKR, et al. (2011) A novel bioinformatics pipeline for identification and characterization of fusion transcripts in breast cancer and normal cell lines. Nucleic Acids Res 39: e100.

110. DiepCH, ZuckerKM, HostetterG, WatanabeA, HuC, et al. (2012) Down-regulation of Yes Associated Protein 1 expression reduces cell proliferation and clonogenicity of pancreatic cancer cells. PLoS One 7: e32783.

111. OngCK, SubimerbC, PairojkulC, WongkhamS, CutcutacheI, et al. (2012) Exome sequencing of liver fluke-associated cholangiocarcinoma. Nature genetics 44: 690–693.

112. BiankinAV, WaddellN, KassahnKS, GingrasMC, MuthuswamyLB, et al. (2012) Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 491: 399–405.

113. GuichardC, AmaddeoG, ImbeaudS, LadeiroY, PelletierL, et al. (2012) Integrated analysis of somatic mutations and focal copy-number changes identifies key genes and pathways in hepatocellular carcinoma. Nature genetics 44: 694–698.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 2
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#