Skip to main content
Top
Published in: Breast Cancer Research and Treatment 2/2011

01-09-2011 | Preclinical study

Ras-related protein 1 and the insulin-like growth factor type I receptor are associated with risk of progression in patients diagnosed with carcinoma in situ

Authors: Dana K. Furstenau, Nandita Mitra, Fei Wan, Robert Lewis, Michael D. Feldman, Douglas L. Fraker, Marina A. Guvakova

Published in: Breast Cancer Research and Treatment | Issue 2/2011

Login to get access

Abstract

Currently, there are no applied molecular markers to aid in predicting risk of carcinoma in situ (CIS) progression to invasive cancer, and therefore, all women diagnosed with CIS undergo surgery. Standard assessment of protein expression in fixed tissue by immunohistochemistry (IHC) is not quantitative and hence is not well suited for measuring biomarkers. In this study, we developed an original analytical method for IHC quantification. Using our novel image-based uniplex (IBU) method, quantitative protein profiling was performed on 90 samples of the breast (17 histologically normal tissues, 16 benign lesions, 15 CIS, and 42 invasive carcinomas). Differences between groups were assessed using analysis of variance (ANOVA) and mixed effects models. Measuring protein expression on a continuous scale revealed a significant increase in Ras-related protein 1 (Rap1) and the insulin-like growth factor type I receptor (IGF-IR) in conjunction with the presence of cancer invasion. Women with invasive cancers were four times more likely to have increased levels of Rap1 [odds ratio (OR) = 3.91; P = 0.0002] and IGF-IR (OR = 4.33; P < 0.0001) than women with non-invasive lesions. Furthermore, expression of both proteins was also increased significantly in CIS adjacent to invasive tumors compared with non-cancerous tissue. These novel findings of a significant up-regulation of Rap1 and IGF-IR in CIS progressing to invasive cancers warrant further investigation of Rap1 and IGF-IR together as a dual biomarker to aid in predicting risk of progression and ultimately providing non-surgical treatment options to those at lower risk.
Appendix
Available only for authorised users
Literature
1.
go back to reference Allegra CJ, Aberle DR, Ganschow P, Hahn SM, Lee CN, Millon-Underwood S, Pike MC, Reed SD, Saftlas AF, Scarvalone SA, Schwartz AM, Slomski C, Yothers G, Zon R (2010) National Institutes of Health State-of-the-Science Conference statement: diagnosis and management of ductal carcinoma in situ September 22–24, 2009. J Natl Cancer Inst 102:161–169PubMedCrossRef Allegra CJ, Aberle DR, Ganschow P, Hahn SM, Lee CN, Millon-Underwood S, Pike MC, Reed SD, Saftlas AF, Scarvalone SA, Schwartz AM, Slomski C, Yothers G, Zon R (2010) National Institutes of Health State-of-the-Science Conference statement: diagnosis and management of ductal carcinoma in situ September 22–24, 2009. J Natl Cancer Inst 102:161–169PubMedCrossRef
2.
go back to reference Kerlikowske K, Molinaro AM, Gauthier ML, Berman HK, Waldman F, Bennington J, Sanchez H, Jimenez C, Stewart K, Chew K, Ljung BM, Tlsty TD (2010) Biomarker expression and risk of subsequent tumors after initial ductal carcinoma in situ diagnosis. J Natl Cancer Inst 102:627–637PubMedCrossRef Kerlikowske K, Molinaro AM, Gauthier ML, Berman HK, Waldman F, Bennington J, Sanchez H, Jimenez C, Stewart K, Chew K, Ljung BM, Tlsty TD (2010) Biomarker expression and risk of subsequent tumors after initial ductal carcinoma in situ diagnosis. J Natl Cancer Inst 102:627–637PubMedCrossRef
3.
go back to reference Kuerer HM, Albarracin CT, Yang WT, Cardiff RD, Brewster AM, Symmans WF, Hylton NM, Middleton LP, Krishnamurthy S, Perkins GH, Babiera G, Edgerton ME, Czerniecki BJ, Arun BK, Hortobagyi GN (2009) Ductal carcinoma in situ: state of the science and roadmap to advance the field. J Clin Oncol 27:279–288PubMedCrossRef Kuerer HM, Albarracin CT, Yang WT, Cardiff RD, Brewster AM, Symmans WF, Hylton NM, Middleton LP, Krishnamurthy S, Perkins GH, Babiera G, Edgerton ME, Czerniecki BJ, Arun BK, Hortobagyi GN (2009) Ductal carcinoma in situ: state of the science and roadmap to advance the field. J Clin Oncol 27:279–288PubMedCrossRef
4.
go back to reference Hoorntje LE, Schipper ME, Peeters PH, Bellot F, Storm RK, Borel RI (2003) The finding of invasive cancer after a preoperative diagnosis of ductal carcinoma-in situ: causes of ductal carcinoma-in situ underestimates with stereotactic 14-gauge needle biopsy. Ann Surg Oncol 10:748–753PubMedCrossRef Hoorntje LE, Schipper ME, Peeters PH, Bellot F, Storm RK, Borel RI (2003) The finding of invasive cancer after a preoperative diagnosis of ductal carcinoma-in situ: causes of ductal carcinoma-in situ underestimates with stereotactic 14-gauge needle biopsy. Ann Surg Oncol 10:748–753PubMedCrossRef
5.
go back to reference Huo L, Sneige N, Hunt KK, Albarracin CT, Lopez A, Resetkova E (2006) Predictors of invasion in patients with core-needle biopsy-diagnosed ductal carcinoma in situ and recommendations for a selective approach to sentinel lymph node biopsy in ductal carcinoma in situ. Cancer 107:1760–1768PubMedCrossRef Huo L, Sneige N, Hunt KK, Albarracin CT, Lopez A, Resetkova E (2006) Predictors of invasion in patients with core-needle biopsy-diagnosed ductal carcinoma in situ and recommendations for a selective approach to sentinel lymph node biopsy in ductal carcinoma in situ. Cancer 107:1760–1768PubMedCrossRef
6.
go back to reference Meijnen P, Oldenburg HS, Loo CE, Nieweg OE, Peterse JL, Rutgers EJ (2007) Risk of invasion and axillary lymph node metastasis in ductal carcinoma in situ diagnosed by core-needle biopsy. Br J Surg 94:952–956PubMedCrossRef Meijnen P, Oldenburg HS, Loo CE, Nieweg OE, Peterse JL, Rutgers EJ (2007) Risk of invasion and axillary lymph node metastasis in ductal carcinoma in situ diagnosed by core-needle biopsy. Br J Surg 94:952–956PubMedCrossRef
7.
go back to reference Renshaw AA (2002) Predicting invasion in the excision specimen from breast core needle biopsy specimens with only ductal carcinoma in situ. Arch Pathol Lab Med 126:39–41PubMed Renshaw AA (2002) Predicting invasion in the excision specimen from breast core needle biopsy specimens with only ductal carcinoma in situ. Arch Pathol Lab Med 126:39–41PubMed
8.
go back to reference Roses RE, Paulson EC, Sharma A, Schueller JE, Nisenbaum H, Weinstein S, Fox KR, Zhang PJ, Czerniecki BJ (2009) HER-2/neu overexpression as a predictor for the transition from in situ to invasive breast cancer. Cancer Epidemiol Biomarkers Prev 18:1386–1389PubMedCrossRef Roses RE, Paulson EC, Sharma A, Schueller JE, Nisenbaum H, Weinstein S, Fox KR, Zhang PJ, Czerniecki BJ (2009) HER-2/neu overexpression as a predictor for the transition from in situ to invasive breast cancer. Cancer Epidemiol Biomarkers Prev 18:1386–1389PubMedCrossRef
9.
go back to reference Kohn EC, Francis EA, Liotta LA, Schiffmann E (1990) Heterogeneity of the motility responses in malignant tumor cells: a biological basis for the diversity and homing of metastatic cells. Int J Cancer 46:287–292PubMedCrossRef Kohn EC, Francis EA, Liotta LA, Schiffmann E (1990) Heterogeneity of the motility responses in malignant tumor cells: a biological basis for the diversity and homing of metastatic cells. Int J Cancer 46:287–292PubMedCrossRef
10.
go back to reference Doerr ME, Jones JI (1996) The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells. J Biol Chem 271:2443–2447PubMedCrossRef Doerr ME, Jones JI (1996) The roles of integrins and extracellular matrix proteins in the insulin-like growth factor I-stimulated chemotaxis of human breast cancer cells. J Biol Chem 271:2443–2447PubMedCrossRef
11.
go back to reference Guvakova MA, Surmacz E (1997) Overexpressed IGF-I receptors reduce estrogen growth requirements, enhance survival, and promote E-cadherin-mediated cell–cell adhesion in human breast cancer cells. Exp Cell Res 231:149–162PubMedCrossRef Guvakova MA, Surmacz E (1997) Overexpressed IGF-I receptors reduce estrogen growth requirements, enhance survival, and promote E-cadherin-mediated cell–cell adhesion in human breast cancer cells. Exp Cell Res 231:149–162PubMedCrossRef
12.
go back to reference Shakibaei M, John T, De Souza P, Rahmanzadeh R, Merker HJ (1999) Signal transduction by beta1 integrin receptors in human chondrocytes in vitro: collaboration with the insulin-like growth factor-I receptor. Biochem J 342(Pt 3):615–623PubMedCrossRef Shakibaei M, John T, De Souza P, Rahmanzadeh R, Merker HJ (1999) Signal transduction by beta1 integrin receptors in human chondrocytes in vitro: collaboration with the insulin-like growth factor-I receptor. Biochem J 342(Pt 3):615–623PubMedCrossRef
13.
go back to reference Vuori K, Ruoslahti E (1994) Association of insulin receptor substrate-1 with integrins. Science 266:1576–1578PubMedCrossRef Vuori K, Ruoslahti E (1994) Association of insulin receptor substrate-1 with integrins. Science 266:1576–1578PubMedCrossRef
14.
go back to reference Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353PubMedCrossRef Lopez T, Hanahan D (2002) Elevated levels of IGF-1 receptor convey invasive and metastatic capability in a mouse model of pancreatic islet tumorigenesis. Cancer Cell 1:339–353PubMedCrossRef
15.
go back to reference All-Ericsson C, Girnita L, Seregard S, Bartolazzi A, Jager MJ, Larsson O (2002) Insulin-like growth factor-1 receptor in uveal melanoma: a predictor for metastatic disease and a potential therapeutic target. Invest Ophthalmol Vis Sci 43:1–8PubMed All-Ericsson C, Girnita L, Seregard S, Bartolazzi A, Jager MJ, Larsson O (2002) Insulin-like growth factor-1 receptor in uveal melanoma: a predictor for metastatic disease and a potential therapeutic target. Invest Ophthalmol Vis Sci 43:1–8PubMed
16.
go back to reference Brodt P, Samani A, Navab R (2000) Inhibition of the type I insulin-like growth factor receptor expression and signaling: novel strategies for antimetastatic therapy. Biochem Pharmacol 60:1101–1107PubMedCrossRef Brodt P, Samani A, Navab R (2000) Inhibition of the type I insulin-like growth factor receptor expression and signaling: novel strategies for antimetastatic therapy. Biochem Pharmacol 60:1101–1107PubMedCrossRef
17.
go back to reference Jackson JG, Zhang X, Yoneda T, Yee D (2001) Regulation of breast cancer cell motility by insulin receptor substrate-2 (IRS-2) in metastatic variants of human breast cancer cell lines. Oncogene 20:7318–7325PubMedCrossRef Jackson JG, Zhang X, Yoneda T, Yee D (2001) Regulation of breast cancer cell motility by insulin receptor substrate-2 (IRS-2) in metastatic variants of human breast cancer cell lines. Oncogene 20:7318–7325PubMedCrossRef
18.
go back to reference Nishizuka I, Ishikawa T, Hamaguchi Y, Kamiyama M, Ichikawa Y, Kadota K, Miki R, Tomaru Y, Mizuno Y, Tominaga N, Yano R, Goto H, Nitanda H, Togo S, Okazaki Y, Hayashizaki Y, Shimada H (2002) Analysis of gene expression involved in brain metastasis from breast cancer using cDNA microarray. Breast Cancer 9:26–32PubMedCrossRef Nishizuka I, Ishikawa T, Hamaguchi Y, Kamiyama M, Ichikawa Y, Kadota K, Miki R, Tomaru Y, Mizuno Y, Tominaga N, Yano R, Goto H, Nitanda H, Togo S, Okazaki Y, Hayashizaki Y, Shimada H (2002) Analysis of gene expression involved in brain metastasis from breast cancer using cDNA microarray. Breast Cancer 9:26–32PubMedCrossRef
19.
go back to reference Happerfield LC, Miles DW, Barnes DM, Thomsen LL, Smith P, Hanby A (1997) The localization of the insulin-like growth factor receptor 1 (IGFR-1) in benign and malignant breast tissue. J Pathol 183:412–417PubMedCrossRef Happerfield LC, Miles DW, Barnes DM, Thomsen LL, Smith P, Hanby A (1997) The localization of the insulin-like growth factor receptor 1 (IGFR-1) in benign and malignant breast tissue. J Pathol 183:412–417PubMedCrossRef
20.
go back to reference Resnik JL, Reichart DB, Huey K, Webster NJ, Seely BL (1998) Elevated insulin-like growth factor I receptor autophosphorylation and kinase activity in human breast cancer. Cancer Res 58:1159–1164PubMed Resnik JL, Reichart DB, Huey K, Webster NJ, Seely BL (1998) Elevated insulin-like growth factor I receptor autophosphorylation and kinase activity in human breast cancer. Cancer Res 58:1159–1164PubMed
21.
go back to reference Shimizu C, Hasegawa T, Tani Y, Takahashi F, Takeuchi M, Watanabe T, Ando M, Katsumata N, Fujiwara Y (2004) Expression of insulin-like growth factor 1 receptor in primary breast cancer: immunohistochemical analysis. Hum Pathol 35:1537–1542PubMedCrossRef Shimizu C, Hasegawa T, Tani Y, Takahashi F, Takeuchi M, Watanabe T, Ando M, Katsumata N, Fujiwara Y (2004) Expression of insulin-like growth factor 1 receptor in primary breast cancer: immunohistochemical analysis. Hum Pathol 35:1537–1542PubMedCrossRef
22.
go back to reference Nielsen TO, Andrews HN, Cheang M, Kucab JE, Hsu FD, Ragaz J, Gilks CB, Makretsov N, Bajdik CD, Brookes C, Neckers LM, Evdokimova V, Huntsman DG, Dunn SE (2004) Expression of the insulin-like growth factor I receptor and urokinase plasminogen activator in breast cancer is associated with poor survival: potential for intervention with 17-allylamino geldanamycin. Cancer Res 64:286–291PubMedCrossRef Nielsen TO, Andrews HN, Cheang M, Kucab JE, Hsu FD, Ragaz J, Gilks CB, Makretsov N, Bajdik CD, Brookes C, Neckers LM, Evdokimova V, Huntsman DG, Dunn SE (2004) Expression of the insulin-like growth factor I receptor and urokinase plasminogen activator in breast cancer is associated with poor survival: potential for intervention with 17-allylamino geldanamycin. Cancer Res 64:286–291PubMedCrossRef
23.
go back to reference van Golen KL (2003) Inflammatory breast cancer: relationship between growth factor signaling and motility in aggressive cancers. Breast Cancer Res 5:174–179PubMedCrossRef van Golen KL (2003) Inflammatory breast cancer: relationship between growth factor signaling and motility in aggressive cancers. Breast Cancer Res 5:174–179PubMedCrossRef
24.
go back to reference Chitnis MM, Yuen JS, Protheroe AS, Pollak M, Macaulay VM (2008) The type 1 insulin-like growth factor receptor pathway. Clin Cancer Res 14:6364–6370PubMedCrossRef Chitnis MM, Yuen JS, Protheroe AS, Pollak M, Macaulay VM (2008) The type 1 insulin-like growth factor receptor pathway. Clin Cancer Res 14:6364–6370PubMedCrossRef
25.
go back to reference Romano D, Pertuit M, Rasolonjanahary R, Barnier JV, Magalon K, Enjalbert A, Gerard C (2006) Regulation of the RAP1/RAF-1/extracellularly regulated kinase-1/2 cascade and prolactin release by the phosphoinositide 3-kinase/AKT pathway in pituitary cells. Endocrinology 147:6036–6045PubMedCrossRef Romano D, Pertuit M, Rasolonjanahary R, Barnier JV, Magalon K, Enjalbert A, Gerard C (2006) Regulation of the RAP1/RAF-1/extracellularly regulated kinase-1/2 cascade and prolactin release by the phosphoinositide 3-kinase/AKT pathway in pituitary cells. Endocrinology 147:6036–6045PubMedCrossRef
26.
go back to reference Guvakova MA, Lee WSY (2009) Tuberin and hamartin in moving breast cancer cells expression localization and function. In: Abreu T, Silva G (eds) Cell movement new research trends. Nova Science Publishers, New York, pp 187–207 Guvakova MA, Lee WSY (2009) Tuberin and hamartin in moving breast cancer cells expression localization and function. In: Abreu T, Silva G (eds) Cell movement new research trends. Nova Science Publishers, New York, pp 187–207
27.
go back to reference Paganini S, Guidetti GF, Catricala S, Trionfini P, Panelli S, Balduini C, Torti M (2006) Identification and biochemical characterization of Rap2C, a new member of the Rap family of small GTP-binding proteins. Biochimie 88:285–295PubMedCrossRef Paganini S, Guidetti GF, Catricala S, Trionfini P, Panelli S, Balduini C, Torti M (2006) Identification and biochemical characterization of Rap2C, a new member of the Rap family of small GTP-binding proteins. Biochimie 88:285–295PubMedCrossRef
28.
go back to reference Bos JL, de Rooij J, Reedquist KA (2001) Rap1 signalling: adhering to new models. Nat Rev Mol Cell Biol 2:369–377PubMedCrossRef Bos JL, de Rooij J, Reedquist KA (2001) Rap1 signalling: adhering to new models. Nat Rev Mol Cell Biol 2:369–377PubMedCrossRef
29.
go back to reference Bokoch GM (1993) Biology of the Rap proteins, members of the ras superfamily of GTP-binding proteins. Biochem J 289(Pt 1):17–24PubMed Bokoch GM (1993) Biology of the Rap proteins, members of the ras superfamily of GTP-binding proteins. Biochem J 289(Pt 1):17–24PubMed
31.
go back to reference Caron E (2003) Cellular functions of the Rap1 GTP-binding protein: a pattern emerges. J Cell Sci 116:435–440PubMedCrossRef Caron E (2003) Cellular functions of the Rap1 GTP-binding protein: a pattern emerges. J Cell Sci 116:435–440PubMedCrossRef
32.
go back to reference Hattori M, Minato N (2003) Rap1 GTPase: functions, regulation, and malignancy. J Biochem 134:479–484PubMedCrossRef Hattori M, Minato N (2003) Rap1 GTPase: functions, regulation, and malignancy. J Biochem 134:479–484PubMedCrossRef
33.
34.
go back to reference Gutmann DH, Saporito-Irwin S, De Clue JE, Wienecke R, Guha A (1997) Alterations in the rap1 signaling pathway are common in human gliomas. Oncogene 15:1611–1616PubMedCrossRef Gutmann DH, Saporito-Irwin S, De Clue JE, Wienecke R, Guha A (1997) Alterations in the rap1 signaling pathway are common in human gliomas. Oncogene 15:1611–1616PubMedCrossRef
35.
go back to reference Jiang WG, Sampson J, Martin TA, Lee-Jones L, Watkins G, Douglas-Jones A, Mokbel K, Mansel RE (2005) Tuberin and hamartin are aberrantly expressed and linked to clinical outcome in human breast cancer: the role of promoter methylation of TSC genes. Eur J Cancer 41:1628–1636PubMedCrossRef Jiang WG, Sampson J, Martin TA, Lee-Jones L, Watkins G, Douglas-Jones A, Mokbel K, Mansel RE (2005) Tuberin and hamartin are aberrantly expressed and linked to clinical outcome in human breast cancer: the role of promoter methylation of TSC genes. Eur J Cancer 41:1628–1636PubMedCrossRef
36.
go back to reference Zhang L, Chenwei L, Mahmood R, van Golen K, Greenson J, Li G, D’Silva NJ, Li X, Burant CF, Logsdon CD, Simeone DM (2006) Identification of a putative tumor suppressor gene Rap1GAP in pancreatic cancer. Cancer Res 66:898–906PubMedCrossRef Zhang L, Chenwei L, Mahmood R, van Golen K, Greenson J, Li G, D’Silva NJ, Li X, Burant CF, Logsdon CD, Simeone DM (2006) Identification of a putative tumor suppressor gene Rap1GAP in pancreatic cancer. Cancer Res 66:898–906PubMedCrossRef
37.
go back to reference Nellore A, Paziana K, Ma C, Tsygankova OM, Wang Y, Puttaswamy K, Iqbal AU, Franks SR, Lv Y, Troxel AB, Feldman MD, Meinkoth JL, Brose MS (2009) Loss of Rap1GAP in papillary thyroid cancer. J Clin Endocrinol Metab 94:1026–1032PubMedCrossRef Nellore A, Paziana K, Ma C, Tsygankova OM, Wang Y, Puttaswamy K, Iqbal AU, Franks SR, Lv Y, Troxel AB, Feldman MD, Meinkoth JL, Brose MS (2009) Loss of Rap1GAP in papillary thyroid cancer. J Clin Endocrinol Metab 94:1026–1032PubMedCrossRef
38.
go back to reference Yajnik V, Paulding C, Sordella R, McClatchey AI, Saito M, Wahrer DC, Reynolds P, Bell DW, Lake R, Van, den Heuvel S, Settleman J, Haber DA (2003) DOCK4, a GTPase activator, is disrupted during tumorigenesis. Cell 112:673–684PubMedCrossRef Yajnik V, Paulding C, Sordella R, McClatchey AI, Saito M, Wahrer DC, Reynolds P, Bell DW, Lake R, Van, den Heuvel S, Settleman J, Haber DA (2003) DOCK4, a GTPase activator, is disrupted during tumorigenesis. Cell 112:673–684PubMedCrossRef
39.
go back to reference Hirata T, Nagai H, Koizumi K, Okino K, Harada A, Onda M, Nagahata T, Mikami I, Hirai K, Haraguchi S, Jin E, Kawanami O, Shimizu K, Emi M (2004) Amplification, up-regulation and over-expression of C3G (CRK SH3 domain-binding guanine nucleotide-releasing factor) in non-small cell lung cancers. J Hum Genet 49(6):290–295PubMedCrossRef Hirata T, Nagai H, Koizumi K, Okino K, Harada A, Onda M, Nagahata T, Mikami I, Hirai K, Haraguchi S, Jin E, Kawanami O, Shimizu K, Emi M (2004) Amplification, up-regulation and over-expression of C3G (CRK SH3 domain-binding guanine nucleotide-releasing factor) in non-small cell lung cancers. J Hum Genet 49(6):290–295PubMedCrossRef
40.
go back to reference Kinashi T, Katagiri K (2004) Regulation of lymphocyte adhesion and migration by the small GTPase Rap1 and its effector molecule, RAPL. Immunol Lett 93:1–5PubMedCrossRef Kinashi T, Katagiri K (2004) Regulation of lymphocyte adhesion and migration by the small GTPase Rap1 and its effector molecule, RAPL. Immunol Lett 93:1–5PubMedCrossRef
41.
go back to reference Fujita H, Fukuhara S, Sakurai A, Yamagishi A, Kamioka Y, Nakaoka Y, Masuda M, Mochizuki N (2005) Local activation of Rap1 contributes to directional vascular endothelial cell migration accompanied by extension of microtubules on which RAPL, a Rap1-associating molecule, localizes. J Biol Chem 280:5022–5031PubMedCrossRef Fujita H, Fukuhara S, Sakurai A, Yamagishi A, Kamioka Y, Nakaoka Y, Masuda M, Mochizuki N (2005) Local activation of Rap1 contributes to directional vascular endothelial cell migration accompanied by extension of microtubules on which RAPL, a Rap1-associating molecule, localizes. J Biol Chem 280:5022–5031PubMedCrossRef
42.
go back to reference Takahashi M, Rikitake Y, Nagamatsu Y, Hara T, Ikeda W, Hirata K, Takai Y (2008) Sequential activation of Rap1 and Rac1 small G proteins by PDGF locally at leading edges of NIH3T3 cells. Genes Cells 13:549–569PubMedCrossRef Takahashi M, Rikitake Y, Nagamatsu Y, Hara T, Ikeda W, Hirata K, Takai Y (2008) Sequential activation of Rap1 and Rac1 small G proteins by PDGF locally at leading edges of NIH3T3 cells. Genes Cells 13:549–569PubMedCrossRef
43.
go back to reference Itoh M, Nelson CM, Myers CA, Bissell MJ (2007) Rap1 integrates tissue polarity, lumen formation, and tumorigenic potential in human breast epithelial cells. Cancer Res 67:4759–4766PubMedCrossRef Itoh M, Nelson CM, Myers CA, Bissell MJ (2007) Rap1 integrates tissue polarity, lumen formation, and tumorigenic potential in human breast epithelial cells. Cancer Res 67:4759–4766PubMedCrossRef
44.
go back to reference McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM (2006) Reporting recommendations for tumor marker prognostic studies (remark). Exp Oncol 28:99–105PubMed McShane LM, Altman DG, Sauerbrei W, Taube SE, Gion M, Clark GM (2006) Reporting recommendations for tumor marker prognostic studies (remark). Exp Oncol 28:99–105PubMed
45.
go back to reference Becker KF, Schott C, Hipp S, Metzger V, Porschewski P, Beck R, Nahrig J, Becker I, Hofler H (2007) Quantitative protein analysis from formalin-fixed tissues: implications for translational clinical research and nanoscale molecular diagnosis. J Pathol 211:370–378PubMedCrossRef Becker KF, Schott C, Hipp S, Metzger V, Porschewski P, Beck R, Nahrig J, Becker I, Hofler H (2007) Quantitative protein analysis from formalin-fixed tissues: implications for translational clinical research and nanoscale molecular diagnosis. J Pathol 211:370–378PubMedCrossRef
46.
go back to reference McCarty KS Jr, Szabo E, Flowers JL, Cox EB, Leight GS, Miller L, Konrath J, Soper JT, Budwit DA, Creasman WT (1986) Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res 46:4244s–4248sPubMed McCarty KS Jr, Szabo E, Flowers JL, Cox EB, Leight GS, Miller L, Konrath J, Soper JT, Budwit DA, Creasman WT (1986) Use of a monoclonal anti-estrogen receptor antibody in the immunohistochemical evaluation of human tumors. Cancer Res 46:4244s–4248sPubMed
47.
go back to reference Cregger M, Berger AJ, Rimm DL (2006) Immunohistochemistry and quantitative analysis of protein expression. Arch Pathol Lab Med 130:1026–1030PubMed Cregger M, Berger AJ, Rimm DL (2006) Immunohistochemistry and quantitative analysis of protein expression. Arch Pathol Lab Med 130:1026–1030PubMed
48.
go back to reference Guvakova MA (2007) Insulin-like growth factors control cell migration in health and disease. Int J Biochem Cell Biol 39:890–909PubMedCrossRef Guvakova MA (2007) Insulin-like growth factors control cell migration in health and disease. Int J Biochem Cell Biol 39:890–909PubMedCrossRef
49.
go back to reference Zha J, Lackner MR (2010) Targeting the insulin-like growth factor receptor-1R pathway for cancer therapy. Clin Cancer Res 16:2512–2517PubMedCrossRef Zha J, Lackner MR (2010) Targeting the insulin-like growth factor receptor-1R pathway for cancer therapy. Clin Cancer Res 16:2512–2517PubMedCrossRef
50.
go back to reference Polyak K (2002) Molecular alterations in ductal carcinoma in situ of the breast. Curr Opin Oncol 14:92–96PubMedCrossRef Polyak K (2002) Molecular alterations in ductal carcinoma in situ of the breast. Curr Opin Oncol 14:92–96PubMedCrossRef
51.
go back to reference Wiechmann L, Kuerer HM (2008) The molecular journey from ductal carcinoma in situ to invasive breast cancer. Cancer 112:2130–2142PubMedCrossRef Wiechmann L, Kuerer HM (2008) The molecular journey from ductal carcinoma in situ to invasive breast cancer. Cancer 112:2130–2142PubMedCrossRef
52.
go back to reference Lann D, LeRoith D (2008) The role of endocrine insulin-like growth factor-I and insulin in breast cancer. J Mammary Gland Biol Neoplasia 13:371–379PubMedCrossRef Lann D, LeRoith D (2008) The role of endocrine insulin-like growth factor-I and insulin in breast cancer. J Mammary Gland Biol Neoplasia 13:371–379PubMedCrossRef
53.
go back to reference Schnarr B, Strunz K, Ohsam J, Benner A, Wacker J, Mayer D (2000) Down-regulation of insulin-like growth factor-I receptor and insulin receptor substrate-1 expression in advanced human breast cancer. Int J Cancer 89:506–513PubMedCrossRef Schnarr B, Strunz K, Ohsam J, Benner A, Wacker J, Mayer D (2000) Down-regulation of insulin-like growth factor-I receptor and insulin receptor substrate-1 expression in advanced human breast cancer. Int J Cancer 89:506–513PubMedCrossRef
54.
go back to reference Peyrat JP, Bonneterre J, Beuscart R, Djiane J, Demaille A (1988) Insulin-like growth factor 1 receptors in human breast cancer and their relation to estradiol and progesterone receptors. Cancer Res 48:6429–6433PubMed Peyrat JP, Bonneterre J, Beuscart R, Djiane J, Demaille A (1988) Insulin-like growth factor 1 receptors in human breast cancer and their relation to estradiol and progesterone receptors. Cancer Res 48:6429–6433PubMed
55.
go back to reference Jones RA, Campbell CI, Gunther EJ, Chodosh LA, Petrik JJ, Khokha R, Moorehead RA (2007) Transgenic overexpression of IGF-IR disrupts mammary ductal morphogenesis and induces tumor formation. Oncogene 26:1636–1644PubMedCrossRef Jones RA, Campbell CI, Gunther EJ, Chodosh LA, Petrik JJ, Khokha R, Moorehead RA (2007) Transgenic overexpression of IGF-IR disrupts mammary ductal morphogenesis and induces tumor formation. Oncogene 26:1636–1644PubMedCrossRef
56.
go back to reference Kucab JE, Dunn SE (2003) Role of IGF-1R in mediating breast cancer invasion and metastasis. Breast Dis 17:41–47PubMed Kucab JE, Dunn SE (2003) Role of IGF-1R in mediating breast cancer invasion and metastasis. Breast Dis 17:41–47PubMed
57.
go back to reference Waldman FM, De Vries S, Chew KL, Moore DH, Kerlikowske K, Ljung BM (2000) Chromosomal alterations in ductal carcinomas in situ and their in situ recurrences. J Natl Cancer Inst 92:313–320PubMedCrossRef Waldman FM, De Vries S, Chew KL, Moore DH, Kerlikowske K, Ljung BM (2000) Chromosomal alterations in ductal carcinomas in situ and their in situ recurrences. J Natl Cancer Inst 92:313–320PubMedCrossRef
58.
go back to reference Miller BS, Yee D (2005) Type I insulin-like growth factor receptor as a therapeutic target in cancer. Cancer Res 65:10123–10127PubMedCrossRef Miller BS, Yee D (2005) Type I insulin-like growth factor receptor as a therapeutic target in cancer. Cancer Res 65:10123–10127PubMedCrossRef
59.
go back to reference Yee D (2007) Targeting insulin-like growth factor pathways. Br J Cancer 96(Supp l):7–10 Yee D (2007) Targeting insulin-like growth factor pathways. Br J Cancer 96(Supp l):7–10
60.
go back to reference Rodon J, De Santos V, Ferry RJ Jr, Kurzrock R (2008) Early drug development of inhibitors of the insulin-like growth factor-I receptor pathway: lessons from the first clinical trials. Mol Cancer Ther 7:2575–2588PubMedCrossRef Rodon J, De Santos V, Ferry RJ Jr, Kurzrock R (2008) Early drug development of inhibitors of the insulin-like growth factor-I receptor pathway: lessons from the first clinical trials. Mol Cancer Ther 7:2575–2588PubMedCrossRef
61.
go back to reference Lobell RB, Liu D, Buser CA, Davide JP, De Puy E, Hamilton K, Koblan KS, Lee Y, Mosser S, Motzel SL, Abbruzzese JL, Fuchs CS, Rowinsky EK, Rubin EH, Sharma S, Deutsch PJ, Mazina KE, Morrison BW, Wildonger L, Yao SL, Kohl NE (2002) Preclinical and clinical pharmacodynamic assessment of L-778, 123, a dual inhibitor of farnesyl:protein transferase and geranylgeranyl:protein transferase type-I. Mol Cancer Ther 1:747–758PubMed Lobell RB, Liu D, Buser CA, Davide JP, De Puy E, Hamilton K, Koblan KS, Lee Y, Mosser S, Motzel SL, Abbruzzese JL, Fuchs CS, Rowinsky EK, Rubin EH, Sharma S, Deutsch PJ, Mazina KE, Morrison BW, Wildonger L, Yao SL, Kohl NE (2002) Preclinical and clinical pharmacodynamic assessment of L-778, 123, a dual inhibitor of farnesyl:protein transferase and geranylgeranyl:protein transferase type-I. Mol Cancer Ther 1:747–758PubMed
62.
go back to reference Sun J, Ohkanda J, Coppola D, Yin H, Kothare M, Busciglio B, Hamilton AD, Sebti SM (2003) Geranylgeranyltransferase I inhibitor GGTI-2154 induces breast carcinoma apoptosis and tumor regression in H-Ras transgenic mice. Cancer Res 63:8922–8929PubMed Sun J, Ohkanda J, Coppola D, Yin H, Kothare M, Busciglio B, Hamilton AD, Sebti SM (2003) Geranylgeranyltransferase I inhibitor GGTI-2154 induces breast carcinoma apoptosis and tumor regression in H-Ras transgenic mice. Cancer Res 63:8922–8929PubMed
Metadata
Title
Ras-related protein 1 and the insulin-like growth factor type I receptor are associated with risk of progression in patients diagnosed with carcinoma in situ
Authors
Dana K. Furstenau
Nandita Mitra
Fei Wan
Robert Lewis
Michael D. Feldman
Douglas L. Fraker
Marina A. Guvakova
Publication date
01-09-2011
Publisher
Springer US
Published in
Breast Cancer Research and Treatment / Issue 2/2011
Print ISSN: 0167-6806
Electronic ISSN: 1573-7217
DOI
https://doi.org/10.1007/s10549-010-1227-y

Other articles of this Issue 2/2011

Breast Cancer Research and Treatment 2/2011 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine