Skip to main content
Top
Published in: Molecular Cancer 1/2011

Open Access 01-12-2011 | Research

Targeting notch pathway enhances rapamycin antitumor activity in pancreas cancers through PTEN phosphorylation

Authors: Kevin Vo, Barushi Amarasinghe, Kay Washington, Adriana Gonzalez, Jordan Berlin, Thao P Dang

Published in: Molecular Cancer | Issue 1/2011

Login to get access

Abstract

Background

Pancreas cancer is one of most aggressive human cancers with the survival rate for patients with metastatic pancreas cancer at 5-6 months. The poor survival demonstrates a clear need for better target identification, drug development and new therapeutic strategies. Recent discoveries have shown that the role for Notch pathway is important in both development and cancer. Its contribution to oncogenesis also involves crosstalks with other growth factor pathways, such as Akt and its modulator, PTEN. The mounting evidence supporting a role for Notch in cancer promotion and survival suggests that targeting this pathway alone or in combination with other therapeutics represents a promising therapeutic strategy.

Results

Using a pancreas cancer tissue microarray, we noted that Jagged1, Notch3 and Notch4 are overexpressed in pancreas tumors (26%, 84% and 31% respectively), whereas Notch1 is expressed in blood vessels. While there was no correlation between Notch receptor expression and survival, stage or tumor grade, Notch3 was associated with Jagged1 and EGFR expression, suggesting a unique relationship between Notch3 and Jagged1. Inhibition of the Notch pathway genetically and with gamma-secretase inhibitor (GSI) resulted in tumor suppression and enhanced cell death. The observed anti-tumor activity appeared to be through Akt and modulation of PTEN phosphorylation. We discovered that transcriptional regulation of RhoA by Notch is important for PTEN phosphorylation. Finally, the mTOR inhibitor Rapamycin enhanced the effect of GSI on RhoA expression, resulting in down regulation of phospho-Akt and increased in vitro tumor cytotoxity.

Conclusions

Notch pathway plays an important role in maintaining pancreas tumor phenotype. Targeting this pathway represents a reasonable strategy for the treatment of pancreas cancers. Notch modulates the Akt pathway through regulation of PTEN phosphorylation, an observation that has not been made previously. Furthermore, we discovered that this regulation is dependent on RhoA/Rock1 activation. Enhanced phospho-Akt suppression when GSI is combined with rapamycin suggests that targeting both pathways will lead to a greater efficacy in the treatment of patients with pancreas cancer.
Appendix
Available only for authorised users
Literature
1.
go back to reference Kopan R, Ilagan MXG: The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009, 137 (2): 216-33. 10.1016/j.cell.2009.03.045PubMedCentralCrossRefPubMed Kopan R, Ilagan MXG: The canonical Notch signaling pathway: unfolding the activation mechanism. Cell. 2009, 137 (2): 216-33. 10.1016/j.cell.2009.03.045PubMedCentralCrossRefPubMed
2.
go back to reference Amarir S, Marx M, Calothy G: Notch signaling activation suppresses v-Src-induced transformation of neural cells by restoring TGF-beta-mediated differentiation. PLoS One. 2010, 5 (10): e13572- 10.1371/journal.pone.0013572PubMedCentralCrossRefPubMed Amarir S, Marx M, Calothy G: Notch signaling activation suppresses v-Src-induced transformation of neural cells by restoring TGF-beta-mediated differentiation. PLoS One. 2010, 5 (10): e13572- 10.1371/journal.pone.0013572PubMedCentralCrossRefPubMed
3.
go back to reference Gutierrez A, Look AT: NOTCH and PI3K-AKT pathways intertwined. Cancer Cell. 2007, 12 (5): 411-3. 10.1016/j.ccr.2007.10.027CrossRefPubMed Gutierrez A, Look AT: NOTCH and PI3K-AKT pathways intertwined. Cancer Cell. 2007, 12 (5): 411-3. 10.1016/j.ccr.2007.10.027CrossRefPubMed
4.
go back to reference Zavadil J, Cermak L, Soto-Nieves N, B"ottinger EP: Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J. 2004, 23 (5): 1155-65. 10.1038/sj.emboj.7600069PubMedCentralCrossRefPubMed Zavadil J, Cermak L, Soto-Nieves N, B"ottinger EP: Integration of TGF-beta/Smad and Jagged1/Notch signalling in epithelial-to-mesenchymal transition. EMBO J. 2004, 23 (5): 1155-65. 10.1038/sj.emboj.7600069PubMedCentralCrossRefPubMed
5.
go back to reference Sade H, Krishna S, Sarin A: The anti-apoptotic effect of Notch-1 requires p56lck-dependent, Akt/PKB-mediated signaling in T cells. J Biol Chem. 2004, 279 (4): 2937-44.CrossRefPubMed Sade H, Krishna S, Sarin A: The anti-apoptotic effect of Notch-1 requires p56lck-dependent, Akt/PKB-mediated signaling in T cells. J Biol Chem. 2004, 279 (4): 2937-44.CrossRefPubMed
6.
go back to reference Jemal A, Siegel R, Xu J, Ward E: Cancer statistics, 2010. CA Cancer J Clin. 2010, 60 (5): 277-300. 10.3322/caac.20073CrossRefPubMed Jemal A, Siegel R, Xu J, Ward E: Cancer statistics, 2010. CA Cancer J Clin. 2010, 60 (5): 277-300. 10.3322/caac.20073CrossRefPubMed
7.
go back to reference Cunningham D, Chau I, Stocken DD, Valle JW, Smith D, Steward W, Harper PG, Dunn J, Tudur-Smith C, West J, Falk S, Crellin A, Adab F, Thompson J, Leonard P, Ostrowski J, Eatock M, Scheithauer W, Her-rmann R, Neoptolemos JP: Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009, 27 (33): 5513-8. 10.1200/JCO.2009.24.2446CrossRefPubMed Cunningham D, Chau I, Stocken DD, Valle JW, Smith D, Steward W, Harper PG, Dunn J, Tudur-Smith C, West J, Falk S, Crellin A, Adab F, Thompson J, Leonard P, Ostrowski J, Eatock M, Scheithauer W, Her-rmann R, Neoptolemos JP: Phase III randomized comparison of gemcitabine versus gemcitabine plus capecitabine in patients with advanced pancreatic cancer. J Clin Oncol. 2009, 27 (33): 5513-8. 10.1200/JCO.2009.24.2446CrossRefPubMed
8.
go back to reference Jensen J, Pedersen EE, Galante P, Hald J, Heller RS, Ishibashi M, Kageyama R, Guillemot F, Serup P, Madsen OD: Control of endodermal endocrine development by Hes-1. Nat Genet. 2000, 24: 36-44. 10.1038/71657CrossRefPubMed Jensen J, Pedersen EE, Galante P, Hald J, Heller RS, Ishibashi M, Kageyama R, Guillemot F, Serup P, Madsen OD: Control of endodermal endocrine development by Hes-1. Nat Genet. 2000, 24: 36-44. 10.1038/71657CrossRefPubMed
9.
go back to reference Mazur PK, Einwächter H, Lee M, Sipos B, Nakhai H, Rad R, Zimber-Strobl U, Strobl LJ, Radtke F, Kl"oppel G, Schmid RM, Siveke JT: Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma. Proc Natl Acad Sci USA. 2010, 107 (30): 13438-43. 10.1073/pnas.1002423107PubMedCentralCrossRefPubMed Mazur PK, Einwächter H, Lee M, Sipos B, Nakhai H, Rad R, Zimber-Strobl U, Strobl LJ, Radtke F, Kl"oppel G, Schmid RM, Siveke JT: Notch2 is required for progression of pancreatic intraepithelial neoplasia and development of pancreatic ductal adenocarcinoma. Proc Natl Acad Sci USA. 2010, 107 (30): 13438-43. 10.1073/pnas.1002423107PubMedCentralCrossRefPubMed
10.
go back to reference Miyamoto Y, Maitra A, Ghosh B, Zechner U, Argani P, Iacobuzio-Donahue CA, Sriuranpong V, Iso T, Meszoely IM, Wolfe MS, Hruban RH, Ball DW, Schmid RM, Leach SD: Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003, 3 (6): 565-76. 10.1016/S1535-6108(03)00140-5CrossRefPubMed Miyamoto Y, Maitra A, Ghosh B, Zechner U, Argani P, Iacobuzio-Donahue CA, Sriuranpong V, Iso T, Meszoely IM, Wolfe MS, Hruban RH, Ball DW, Schmid RM, Leach SD: Notch mediates TGF alpha-induced changes in epithelial differentiation during pancreatic tumorigenesis. Cancer Cell. 2003, 3 (6): 565-76. 10.1016/S1535-6108(03)00140-5CrossRefPubMed
11.
go back to reference Choi J, Park J, Davidson B, Morin P, Shih I, Wang T: Jagged-1 and Notch3 juxtacrine loop regulates ovarian tumor growth and adhesion. Cancer Res. 2008, 68 (14): 5716-5723. 10.1158/0008-5472.CAN-08-0001PubMedCentralCrossRefPubMed Choi J, Park J, Davidson B, Morin P, Shih I, Wang T: Jagged-1 and Notch3 juxtacrine loop regulates ovarian tumor growth and adhesion. Cancer Res. 2008, 68 (14): 5716-5723. 10.1158/0008-5472.CAN-08-0001PubMedCentralCrossRefPubMed
12.
go back to reference Haruki N, Kawaguchi KS, Eichenberger S, Massion PP, Olson S, Gonzalez A, Carbone DP, Dang TP: Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res. 2005, 65 (9): 3555-61. 10.1158/0008-5472.CAN-04-3132CrossRefPubMed Haruki N, Kawaguchi KS, Eichenberger S, Massion PP, Olson S, Gonzalez A, Carbone DP, Dang TP: Dominant-negative Notch3 receptor inhibits mitogen-activated protein kinase pathway and the growth of human lung cancers. Cancer Res. 2005, 65 (9): 3555-61. 10.1158/0008-5472.CAN-04-3132CrossRefPubMed
13.
go back to reference Ijichi H, Chytil A, Gorska AE, Aakre ME, Fujitani Y, Fujitani S, Wright CVE, Moses HL: Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes Dev. 2006, 20 (22): 3147-60. 10.1101/gad.1475506PubMedCentralCrossRefPubMed Ijichi H, Chytil A, Gorska AE, Aakre ME, Fujitani Y, Fujitani S, Wright CVE, Moses HL: Aggressive pancreatic ductal adenocarcinoma in mice caused by pancreas-specific blockade of transforming growth factor-beta signaling in cooperation with active Kras expression. Genes Dev. 2006, 20 (22): 3147-60. 10.1101/gad.1475506PubMedCentralCrossRefPubMed
14.
go back to reference Konishi J, Yi F, Chen X, Vo H, Carbone DP, Dang TP: Notch3 cooperates with the EGFR pathway to modulate apoptosis through the induction of bim. Oncogene. 2010, 29 (4): 589-96. 10.1038/onc.2009.366PubMedCentralCrossRefPubMed Konishi J, Yi F, Chen X, Vo H, Carbone DP, Dang TP: Notch3 cooperates with the EGFR pathway to modulate apoptosis through the induction of bim. Oncogene. 2010, 29 (4): 589-96. 10.1038/onc.2009.366PubMedCentralCrossRefPubMed
15.
go back to reference Eliasz S, Liang S, Chen Y, De Marco MA, Machek O, Skucha S, Miele L, Bocchetta M: Notch-1 stimulates survival of lung adenocarcinoma cells during hypoxia by activating the IGF-1R pathway. Oncogene. 2010, 29 (17): 2488-98. 10.1038/onc.2010.7PubMedCentralCrossRefPubMed Eliasz S, Liang S, Chen Y, De Marco MA, Machek O, Skucha S, Miele L, Bocchetta M: Notch-1 stimulates survival of lung adenocarcinoma cells during hypoxia by activating the IGF-1R pathway. Oncogene. 2010, 29 (17): 2488-98. 10.1038/onc.2010.7PubMedCentralCrossRefPubMed
16.
go back to reference Vazquez F, Ramaswamy S, Nakamura N, Sellers WR: Phosphorylation of the PTEN tail regulates protein stability and function. Mol Cell Biol. 2000, 20 (14): 5010-8. 10.1128/MCB.20.14.5010-5018.2000PubMedCentralCrossRefPubMed Vazquez F, Ramaswamy S, Nakamura N, Sellers WR: Phosphorylation of the PTEN tail regulates protein stability and function. Mol Cell Biol. 2000, 20 (14): 5010-8. 10.1128/MCB.20.14.5010-5018.2000PubMedCentralCrossRefPubMed
17.
go back to reference Vemula S, Shi J, Hanneman P, Wei L, Kapur R: ROCK1 functions as a suppressor of inflammatory cell migration by regulating PTEN phosphorylation and stability. Blood. 2010, 115 (9): 1785-96. 10.1182/blood-2009-08-237222PubMedCentralCrossRefPubMed Vemula S, Shi J, Hanneman P, Wei L, Kapur R: ROCK1 functions as a suppressor of inflammatory cell migration by regulating PTEN phosphorylation and stability. Blood. 2010, 115 (9): 1785-96. 10.1182/blood-2009-08-237222PubMedCentralCrossRefPubMed
18.
go back to reference Kikuchi K, Li X, Zheng Y, Takano Y: Invasion of breast cancer cells into collagen matrix requires TGF-β and Cdc42 signaling. FEBS Lett. 2011, 585 (2): 286-290. 10.1016/j.febslet.2010.12.010CrossRefPubMed Kikuchi K, Li X, Zheng Y, Takano Y: Invasion of breast cancer cells into collagen matrix requires TGF-β and Cdc42 signaling. FEBS Lett. 2011, 585 (2): 286-290. 10.1016/j.febslet.2010.12.010CrossRefPubMed
19.
go back to reference Liliental J, Moon SY, Lesche R, Mamillapalli R, Li D, Zheng Y, Sun H, Wu H: Genetic deletion of the Pten tumor suppressor gene promotes cell motility by activation of Rac1 and Cdc42 GTPases. Curr Biol. 2000, 10 (7): 401-4. 10.1016/S0960-9822(00)00417-6CrossRefPubMed Liliental J, Moon SY, Lesche R, Mamillapalli R, Li D, Zheng Y, Sun H, Wu H: Genetic deletion of the Pten tumor suppressor gene promotes cell motility by activation of Rac1 and Cdc42 GTPases. Curr Biol. 2000, 10 (7): 401-4. 10.1016/S0960-9822(00)00417-6CrossRefPubMed
20.
go back to reference Li Z, Dong X, Dong X, Wang Z, Liu W, Deng N, Ding Y, Tang L, Hla T, Zeng R, Li L, Wu D: Regulation of PTEN by Rho small GTPases. Nat Cell Biol. 2005, 7 (4): 399-404. 10.1038/ncb1236CrossRefPubMed Li Z, Dong X, Dong X, Wang Z, Liu W, Deng N, Ding Y, Tang L, Hla T, Zeng R, Li L, Wu D: Regulation of PTEN by Rho small GTPases. Nat Cell Biol. 2005, 7 (4): 399-404. 10.1038/ncb1236CrossRefPubMed
21.
22.
go back to reference O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, Lane H, Hofmann F, Hicklin DJ, Ludwig DL, Baselga J, Rosen N: mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006, 66 (3): 1500-8. 10.1158/0008-5472.CAN-05-2925PubMedCentralCrossRefPubMed O'Reilly KE, Rojo F, She QB, Solit D, Mills GB, Smith D, Lane H, Hofmann F, Hicklin DJ, Ludwig DL, Baselga J, Rosen N: mTOR inhibition induces upstream receptor tyrosine kinase signaling and activates Akt. Cancer Res. 2006, 66 (3): 1500-8. 10.1158/0008-5472.CAN-05-2925PubMedCentralCrossRefPubMed
23.
go back to reference Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP, Fuchs CS: Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 2009, 27 (2): 193-8. 10.1200/JCO.2008.18.9514PubMedCentralCrossRefPubMed Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP, Fuchs CS: Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 2009, 27 (2): 193-8. 10.1200/JCO.2008.18.9514PubMedCentralCrossRefPubMed
24.
go back to reference Williams CK, Li JL, Murga M, Harris AL, Tosato G: Up-regulation of the Notch ligand Delta-like 4 inhibits VEGF-induced endothelial cell function. Blood. 2006, 107 (3): 931-9.PubMedCentralCrossRefPubMed Williams CK, Li JL, Murga M, Harris AL, Tosato G: Up-regulation of the Notch ligand Delta-like 4 inhibits VEGF-induced endothelial cell function. Blood. 2006, 107 (3): 931-9.PubMedCentralCrossRefPubMed
25.
go back to reference Liu P, Cheng H, Roberts TM, Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 2009, 8 (8): 627-44. 10.1038/nrd2926PubMedCentralCrossRefPubMed Liu P, Cheng H, Roberts TM, Zhao JJ: Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov. 2009, 8 (8): 627-44. 10.1038/nrd2926PubMedCentralCrossRefPubMed
26.
27.
go back to reference Vazquez F, Grossman SR, Takahashi Y, Rokas MV, Nakamura N, Sellers WR: Phosphorylation of the PTEN tail acts as an inhibitory switch by preventing its recruitment into a protein complex. J Biol Chem. 2001, 276 (52): 48627-30. 10.1074/jbc.C100556200CrossRefPubMed Vazquez F, Grossman SR, Takahashi Y, Rokas MV, Nakamura N, Sellers WR: Phosphorylation of the PTEN tail acts as an inhibitory switch by preventing its recruitment into a protein complex. J Biol Chem. 2001, 276 (52): 48627-30. 10.1074/jbc.C100556200CrossRefPubMed
28.
go back to reference Lewis HD, Leveridge M, Strack PR, Haldon CD, O'neil J, Kim H, Madin A, Hannam JC, Look AT, Kohl N, Draetta G, Harrison T, Kerby JA, Shearman MS, Beher D: Apoptosis in T cell acute lymphoblastic leukemia cells after cell cycle arrest induced by pharmacological inhibition of notch signaling. Chem Biol. 2007, 14 (2): 209-19. 10.1016/j.chembiol.2006.12.010CrossRefPubMed Lewis HD, Leveridge M, Strack PR, Haldon CD, O'neil J, Kim H, Madin A, Hannam JC, Look AT, Kohl N, Draetta G, Harrison T, Kerby JA, Shearman MS, Beher D: Apoptosis in T cell acute lymphoblastic leukemia cells after cell cycle arrest induced by pharmacological inhibition of notch signaling. Chem Biol. 2007, 14 (2): 209-19. 10.1016/j.chembiol.2006.12.010CrossRefPubMed
29.
go back to reference Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N, Piga N, Chapon F, Godfrain C, Tournier-Lasserve E: The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest. 2000, 105 (5): 597-605. 10.1172/JCI8047PubMedCentralCrossRefPubMed Joutel A, Andreux F, Gaulis S, Domenga V, Cecillon M, Battail N, Piga N, Chapon F, Godfrain C, Tournier-Lasserve E: The ectodomain of the Notch3 receptor accumulates within the cerebrovasculature of CADASIL patients. J Clin Invest. 2000, 105 (5): 597-605. 10.1172/JCI8047PubMedCentralCrossRefPubMed
Metadata
Title
Targeting notch pathway enhances rapamycin antitumor activity in pancreas cancers through PTEN phosphorylation
Authors
Kevin Vo
Barushi Amarasinghe
Kay Washington
Adriana Gonzalez
Jordan Berlin
Thao P Dang
Publication date
01-12-2011
Publisher
BioMed Central
Published in
Molecular Cancer / Issue 1/2011
Electronic ISSN: 1476-4598
DOI
https://doi.org/10.1186/1476-4598-10-138

Other articles of this Issue 1/2011

Molecular Cancer 1/2011 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