Skip to main content
Top
Published in: Journal of Hematology & Oncology 1/2017

Open Access 01-12-2017 | Research

Phospho-Akt overexpression is prognostic and can be used to tailor the synergistic interaction of Akt inhibitors with gemcitabine in pancreatic cancer

Authors: Daniela Massihnia, Amir Avan, Niccola Funel, Mina Maftouh, Anne van Krieken, Carlotta Granchi, Rajiv Raktoe, Ugo Boggi, Babette Aicher, Filippo Minutolo, Antonio Russo, Leticia G. Leon, Godefridus J. Peters, Elisa Giovannetti

Published in: Journal of Hematology & Oncology | Issue 1/2017

Login to get access

Abstract

Background

There is increasing evidence of a constitutive activation of Akt in pancreatic ductal adenocarcinoma (PDAC), associated with poor prognosis and chemoresistance. Therefore, we evaluated the expression of phospho-Akt in PDAC tissues and cells, and investigated molecular mechanisms influencing the therapeutic potential of Akt inhibition in combination with gemcitabine.

Methods

Phospho-Akt expression was evaluated by immunohistochemistry in tissue microarrays (TMAs) with specimens tissue from radically-resected patients (n = 100). Data were analyzed by Fisher and log-rank test. In vitro studies were performed in 14 PDAC cells, including seven primary cultures, characterized for their Akt1 mRNA and phospho-Akt/Akt levels by quantitative-RT-PCR and immunocytochemistry. Growth inhibitory effects of Akt inhibitors and gemcitabine were evaluated by SRB assay, whereas modulation of Akt and phospho-Akt was investigated by Western blotting and ELISA. Cell cycle perturbation, apoptosis-induction, and anti-migratory behaviors were studied by flow cytometry, AnnexinV, membrane potential, and migration assay, while pharmacological interaction with gemcitabine was determined with combination index (CI) method.

Results

Immunohistochemistry of TMAs revealed a correlation between phospho-Akt expression and worse outcome, particularly in patients with the highest phospho-Akt levels, who had significantly shorter overall and progression-free-survival. Similar expression levels were detected in LPC028 primary cells, while LPC006 were characterized by low phospho-Akt. Remarkably, Akt inhibitors reduced cancer cell growth in monolayers and spheroids and synergistically enhanced the antiproliferative activity of gemcitabine in LPC028, while this combination was antagonistic in LPC006 cells. The synergistic effect was paralleled by a reduced expression of ribonucleotide reductase, potentially facilitating gemcitabine cytotoxicity. Inhibition of Akt decreased cell migration and invasion, which was additionally reduced by the combination with gemcitabine. This combination significantly increased apoptosis, associated with induction of caspase-3/6/8/9, PARP and BAD, and inhibition of Bcl-2 and NF-kB in LPC028, but not in LPC006 cells. However, targeting the key glucose transporter Glut1 resulted in similar apoptosis induction in LPC006 cells.

Conclusions

These data support the analysis of phospho-Akt expression as both a prognostic and a predictive biomarker, for the rational development of new combination therapies targeting the Akt pathway in PDAC. Finally, inhibition of Glut1 might overcome resistance to these therapies and warrants further studies.
Appendix
Available only for authorised users
Literature
5.
go back to reference Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.CrossRefPubMedPubMedCentral Jones S, Zhang X, Parsons DW, Lin JC, Leary RJ, Angenendt P, et al. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science. 2008;321:1801–6.CrossRefPubMedPubMedCentral
6.
go back to reference Baer R, Cintas C, Therville N, Guillermet-Guibert J. Implication of PI3K/Akt pathway in pancreatic cancer: when PI3K isoforms matter? Adv Biol Requl. 2015;59:19–35.CrossRef Baer R, Cintas C, Therville N, Guillermet-Guibert J. Implication of PI3K/Akt pathway in pancreatic cancer: when PI3K isoforms matter? Adv Biol Requl. 2015;59:19–35.CrossRef
7.
go back to reference Mirzoeva OK, Hann B, Hom YK, Debnath J, Aftab D, Shokat K, et al. Autophagy suppression promotes apoptotic cell death in response to inhibition of the PI3K-mTOR pathway in pancreatic adenocarcinoma. J Mol Med. 2011;89:877–89.CrossRefPubMed Mirzoeva OK, Hann B, Hom YK, Debnath J, Aftab D, Shokat K, et al. Autophagy suppression promotes apoptotic cell death in response to inhibition of the PI3K-mTOR pathway in pancreatic adenocarcinoma. J Mol Med. 2011;89:877–89.CrossRefPubMed
8.
go back to reference Hers I, Vincent EE, Tavaré JM. Akt signalling in health and disease. Cell Signal. 2011;23:1515–27.CrossRefPubMed Hers I, Vincent EE, Tavaré JM. Akt signalling in health and disease. Cell Signal. 2011;23:1515–27.CrossRefPubMed
9.
go back to reference Tan I, Liu P, Huang Y, Zhou LYang Y, Wang H, et al. Phosphoproteome analysis of invasion and metastasis-related factors in pancreatic cancer cells. Plos One. 2016;11, e0152280.CrossRefPubMedPubMedCentral Tan I, Liu P, Huang Y, Zhou LYang Y, Wang H, et al. Phosphoproteome analysis of invasion and metastasis-related factors in pancreatic cancer cells. Plos One. 2016;11, e0152280.CrossRefPubMedPubMedCentral
10.
go back to reference Avan A, Avan A, Le Large TY, Mambrini A, Funel N, Maftouh M, et al. AKT1 and SELP polymorphisms predict the risk of developing cachexia in pancreatic cancer patients. Plos one. 2014;9e:108057.CrossRef Avan A, Avan A, Le Large TY, Mambrini A, Funel N, Maftouh M, et al. AKT1 and SELP polymorphisms predict the risk of developing cachexia in pancreatic cancer patients. Plos one. 2014;9e:108057.CrossRef
11.
go back to reference LoRusso PM. Inhibition of the PI3K/AKT/mTOR pathway in solid tumors. J Clin Oncol. 2016. Epub ahead of print. LoRusso PM. Inhibition of the PI3K/AKT/mTOR pathway in solid tumors. J Clin Oncol. 2016. Epub ahead of print.
12.
go back to reference Warfel NA, Kraft AS. PIM kinase (and Akt) biology and signaling in tumors. Pharmacol Therm. 2015;151:41–9.CrossRef Warfel NA, Kraft AS. PIM kinase (and Akt) biology and signaling in tumors. Pharmacol Therm. 2015;151:41–9.CrossRef
13.
go back to reference Liu D, Zhang Y, Dang C, Ma Q, Lee W, Chen W. siRNA directed against TrkA sensitizes human pancreatic cancer cells to apoptosis induced by gemcitabine through an inactivation of PI3K/Akt-dependent pathway. Oncol Rep. 2007;18:673–7.PubMed Liu D, Zhang Y, Dang C, Ma Q, Lee W, Chen W. siRNA directed against TrkA sensitizes human pancreatic cancer cells to apoptosis induced by gemcitabine through an inactivation of PI3K/Akt-dependent pathway. Oncol Rep. 2007;18:673–7.PubMed
14.
go back to reference Yao J, Qian C. Inhibition of NOTCH3 enhacens sensitivity to gemcitabine in pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway. Med Oncol. 2010;1017:22. Yao J, Qian C. Inhibition of NOTCH3 enhacens sensitivity to gemcitabine in pancreatic cancer through an inactivation of PI3K/Akt-dependent pathway. Med Oncol. 2010;1017:22.
15.
go back to reference Fei HR, Chen G, Wang JM, Wang FZ. Perifosine induces cell cycle arrest and apoptosis in human hepatocellular carcinoma cell lines by blockade of Akt phosphorylation. Cytotechnology. 2010;62:449–60.CrossRefPubMedPubMedCentral Fei HR, Chen G, Wang JM, Wang FZ. Perifosine induces cell cycle arrest and apoptosis in human hepatocellular carcinoma cell lines by blockade of Akt phosphorylation. Cytotechnology. 2010;62:449–60.CrossRefPubMedPubMedCentral
16.
go back to reference Zitzmann K, Vlotides G, Brand S, Lahm H, Spöttl G, Göke B, et al. Perifosine-mediate Akt inhibition in neuroendocrine tumor cells: role of specific Akt isoforms. Endocr Relat Cancer. 2012;19:423–34.CrossRefPubMed Zitzmann K, Vlotides G, Brand S, Lahm H, Spöttl G, Göke B, et al. Perifosine-mediate Akt inhibition in neuroendocrine tumor cells: role of specific Akt isoforms. Endocr Relat Cancer. 2012;19:423–34.CrossRefPubMed
17.
go back to reference Ma J, Sawai H, Matsuo Y, Ochi N, Yasuda A, Takahashi H, et al. IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. J Surg Res. 2010;160:90–101.CrossRefPubMed Ma J, Sawai H, Matsuo Y, Ochi N, Yasuda A, Takahashi H, et al. IGF-1 mediates PTEN suppression and enhances cell invasion and proliferation via activation of the IGF-1/PI3K/Akt signaling pathway in pancreatic cancer cells. J Surg Res. 2010;160:90–101.CrossRefPubMed
18.
go back to reference Chen X, Liao J, Lu Y, Duan X, Sun W. Activation of PI3K/Akt pathway mediates bone morphogenetic protein 2-induced invasion of pancreatic cancer cell Panc-1. Pathol Oncol Res. 2011;17:257–61.CrossRefPubMed Chen X, Liao J, Lu Y, Duan X, Sun W. Activation of PI3K/Akt pathway mediates bone morphogenetic protein 2-induced invasion of pancreatic cancer cell Panc-1. Pathol Oncol Res. 2011;17:257–61.CrossRefPubMed
19.
go back to reference Fensterle J, Aicher B, Seipelt I, Teifel M, Engel J. Current view on the mechanism of action of perifosine in cancer. Anticancer Agents Med Chem. 2014;14:629–35.CrossRefPubMed Fensterle J, Aicher B, Seipelt I, Teifel M, Engel J. Current view on the mechanism of action of perifosine in cancer. Anticancer Agents Med Chem. 2014;14:629–35.CrossRefPubMed
20.
go back to reference Holohan B, Hagiopian MM, Lai TP, Huang E, Friedman DR, Wright WE, et al. Perifosine as a potential novel anti-telomerase therapy. Oncotarget. 2015;6:21816–26.CrossRefPubMedPubMedCentral Holohan B, Hagiopian MM, Lai TP, Huang E, Friedman DR, Wright WE, et al. Perifosine as a potential novel anti-telomerase therapy. Oncotarget. 2015;6:21816–26.CrossRefPubMedPubMedCentral
21.
go back to reference Chu E. An update on the current and emerging targeted agents in metastatic colorectal cancer. Clin Colorectal Cancer. 2012;11:1–13.CrossRefPubMed Chu E. An update on the current and emerging targeted agents in metastatic colorectal cancer. Clin Colorectal Cancer. 2012;11:1–13.CrossRefPubMed
22.
go back to reference Lin X, Zhang X, Wang Q, Li J, Zhang P, Zhao M, et al. Perifosine downregulates MDR1 gene expression and reverses multidrug-resistant phenotype by inhibiting PI3K/Akt/NF-kB signaling pathway in a human breast cancer cell line. Neoplasma. 2012;59:248–56.CrossRefPubMed Lin X, Zhang X, Wang Q, Li J, Zhang P, Zhao M, et al. Perifosine downregulates MDR1 gene expression and reverses multidrug-resistant phenotype by inhibiting PI3K/Akt/NF-kB signaling pathway in a human breast cancer cell line. Neoplasma. 2012;59:248–56.CrossRefPubMed
23.
go back to reference Simons AL, Parsons AD, Foster KA, Orcutt KP, Fath MA, Spitz DR. Inhibition of glutathione and thioredoxin metabolism enhances sensitivity to perifosine in head and neck cancer cells. J Oncol. 2009;2009:519563.CrossRefPubMedPubMedCentral Simons AL, Parsons AD, Foster KA, Orcutt KP, Fath MA, Spitz DR. Inhibition of glutathione and thioredoxin metabolism enhances sensitivity to perifosine in head and neck cancer cells. J Oncol. 2009;2009:519563.CrossRefPubMedPubMedCentral
24.
go back to reference Gao Y, Ishiyama H, Sun M, Brinkman KL, Wang X, Zhu J, et al. The alkylphospholipid, perifosine, radiosensitizes prostate cancer cell both in vitro and in vivo. Radiat Oncol. 2011;6:39.CrossRefPubMedPubMedCentral Gao Y, Ishiyama H, Sun M, Brinkman KL, Wang X, Zhu J, et al. The alkylphospholipid, perifosine, radiosensitizes prostate cancer cell both in vitro and in vivo. Radiat Oncol. 2011;6:39.CrossRefPubMedPubMedCentral
25.
go back to reference Xin Y, Shen XD, Cheng L, Hong DF, Chen B. Perifosine inhibit S6K1-Gli1signaling and enhances gemcitabine-induced anti-pancreatic cancer efficacy. Cancer Chemoter Pharmacol. 2014;73:711–9.CrossRef Xin Y, Shen XD, Cheng L, Hong DF, Chen B. Perifosine inhibit S6K1-Gli1signaling and enhances gemcitabine-induced anti-pancreatic cancer efficacy. Cancer Chemoter Pharmacol. 2014;73:711–9.CrossRef
26.
go back to reference Shen J, Xu L, Zhao Q. Perifosine and ABT-737 synergistically inhibit lung cancer cells in vitro and in vivo. Biochem Biophys Res Commun. 2016;473:1170–6.CrossRefPubMed Shen J, Xu L, Zhao Q. Perifosine and ABT-737 synergistically inhibit lung cancer cells in vitro and in vivo. Biochem Biophys Res Commun. 2016;473:1170–6.CrossRefPubMed
27.
go back to reference Pinton G, Manente AG, Angeli G, Mutti L, Moro L. Perifosine as a potential novel anti-cancer agent inhibits EGFR/MET-AKT axis in malignant pleural mesothelioma. PLoS One. 2012;7, e36856.CrossRefPubMedPubMedCentral Pinton G, Manente AG, Angeli G, Mutti L, Moro L. Perifosine as a potential novel anti-cancer agent inhibits EGFR/MET-AKT axis in malignant pleural mesothelioma. PLoS One. 2012;7, e36856.CrossRefPubMedPubMedCentral
28.
go back to reference Huang W, Yang L, Liang S, Liu D, Chen X, Ma Z, et al. AEG-1 is a target of perifosine and is over-expressed in gastric dysplasia and cancers. Dig Dis Sci. 2013;58:2873–80.CrossRefPubMed Huang W, Yang L, Liang S, Liu D, Chen X, Ma Z, et al. AEG-1 is a target of perifosine and is over-expressed in gastric dysplasia and cancers. Dig Dis Sci. 2013;58:2873–80.CrossRefPubMed
29.
go back to reference Chen MB, Wu XY, Tao GQ, Liu CY, Chen J, Wang LQ, et al. Perifosine sensitizes curcumin-induced anti-colorectal cancer effects by targeting multiple signaling pathways both in vivo and in vitro. Int J Cancer. 2012;131:2487–98.CrossRefPubMed Chen MB, Wu XY, Tao GQ, Liu CY, Chen J, Wang LQ, et al. Perifosine sensitizes curcumin-induced anti-colorectal cancer effects by targeting multiple signaling pathways both in vivo and in vitro. Int J Cancer. 2012;131:2487–98.CrossRefPubMed
30.
go back to reference Li X, Luwor R, Lu Y, Liang K, Fan Z. Enhancement of antitumor activity of the anti-EGF receptor monoclonal antibody cetuximab/C225 by perifosine in PTEN-deficient cancer cells. Oncogene. 2006;25:525–35.PubMed Li X, Luwor R, Lu Y, Liang K, Fan Z. Enhancement of antitumor activity of the anti-EGF receptor monoclonal antibody cetuximab/C225 by perifosine in PTEN-deficient cancer cells. Oncogene. 2006;25:525–35.PubMed
31.
go back to reference Giovannetti E, Funel N, Peters GJ, Del Chiaro M, Erozenci LA, Vasile E, et al. MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010;70:4528–38.CrossRefPubMed Giovannetti E, Funel N, Peters GJ, Del Chiaro M, Erozenci LA, Vasile E, et al. MicroRNA-21 in pancreatic cancer: correlation with clinical outcome and pharmacologic aspects underlying its role in the modulation of gemcitabine activity. Cancer Res. 2010;70:4528–38.CrossRefPubMed
32.
go back to reference Avan A, Crea F, Paolicchi E, Funel N, Galvani E, Marquez VE, et al. Molecular mechanisms involved in the synergistic interaction of the EZH2 inhibitor 3-deazaneplanocin A with gemcitabine in pancreatic cancer cells. Mol Cancer Ther. 2012;11:1735–46.CrossRefPubMedPubMedCentral Avan A, Crea F, Paolicchi E, Funel N, Galvani E, Marquez VE, et al. Molecular mechanisms involved in the synergistic interaction of the EZH2 inhibitor 3-deazaneplanocin A with gemcitabine in pancreatic cancer cells. Mol Cancer Ther. 2012;11:1735–46.CrossRefPubMedPubMedCentral
33.
go back to reference Avan A, Quint K, Nicolini F, Funel N, Frampton AE, Maftouh M, et al. Enhancement of the antiproliferative activity of gemcitabine by modulation of c-Met pathway in pancreatic cancer. Curr Pharm Des. 2013;19:940–50.CrossRefPubMed Avan A, Quint K, Nicolini F, Funel N, Frampton AE, Maftouh M, et al. Enhancement of the antiproliferative activity of gemcitabine by modulation of c-Met pathway in pancreatic cancer. Curr Pharm Des. 2013;19:940–50.CrossRefPubMed
34.
go back to reference Galvani E, Giovannetti E, Saccani F, Cavazzoni A, Leon LG, Dekker H, et al. Molecular mechanisms underlying the antitumor activity of 3-aminopropanamide irreversible inhibitors of the epidermal growth factor receptor in non-small cell lung cancer. Neoplasia. 2013;15:61–72.CrossRefPubMedPubMedCentral Galvani E, Giovannetti E, Saccani F, Cavazzoni A, Leon LG, Dekker H, et al. Molecular mechanisms underlying the antitumor activity of 3-aminopropanamide irreversible inhibitors of the epidermal growth factor receptor in non-small cell lung cancer. Neoplasia. 2013;15:61–72.CrossRefPubMedPubMedCentral
35.
go back to reference Hudson CD, Hugemann T, Mather SJ, Avril N. Resistance to the tyrosine kinase inhibitor axitinib is associated with increased glucose metabolism in pancreatic adenocarcinoma. Cell Death Dis. 2014;5, e1160.CrossRefPubMed Hudson CD, Hugemann T, Mather SJ, Avril N. Resistance to the tyrosine kinase inhibitor axitinib is associated with increased glucose metabolism in pancreatic adenocarcinoma. Cell Death Dis. 2014;5, e1160.CrossRefPubMed
36.
go back to reference Granchi C, Qian Y, Lee HY, Paterni I, Pasero C, Iegre J, et al. Salicylketoximes that target glucose transporter 1 Restrict energy supply to lung cancer cells. ChemMedChem. 2015;10:1892–900.CrossRefPubMedPubMedCentral Granchi C, Qian Y, Lee HY, Paterni I, Pasero C, Iegre J, et al. Salicylketoximes that target glucose transporter 1 Restrict energy supply to lung cancer cells. ChemMedChem. 2015;10:1892–900.CrossRefPubMedPubMedCentral
37.
go back to reference Tuccinardi T, Granchi C, Iegre J, Paterni I, Bertini S, Macchia M, et al. Oxime-based inhibitors of glucose transporter 1 displaying antiproliferative effects in cancer cells. Bioorg Med Chem. 2013;23:6923–7.CrossRef Tuccinardi T, Granchi C, Iegre J, Paterni I, Bertini S, Macchia M, et al. Oxime-based inhibitors of glucose transporter 1 displaying antiproliferative effects in cancer cells. Bioorg Med Chem. 2013;23:6923–7.CrossRef
38.
go back to reference Longati P, Jia X, Eimer J, Wagman A, Witt MR, Rehnmark S, et al. 3D pancreatic carcinoma spheroids induce a matrix-rich, chemoresistant phenotype offering a better model for drug testing. BMC Cancer. 2013;13:95.CrossRefPubMedPubMedCentral Longati P, Jia X, Eimer J, Wagman A, Witt MR, Rehnmark S, et al. 3D pancreatic carcinoma spheroids induce a matrix-rich, chemoresistant phenotype offering a better model for drug testing. BMC Cancer. 2013;13:95.CrossRefPubMedPubMedCentral
39.
go back to reference Gills JJ, Dennis PA. Perifosine: update on a novel Akt inhibitor. Curr Oncol Rep. 2009;11:102–10.CrossRefPubMed Gills JJ, Dennis PA. Perifosine: update on a novel Akt inhibitor. Curr Oncol Rep. 2009;11:102–10.CrossRefPubMed
40.
go back to reference Elnaggar M, Giovannetti E, Peters GJ. Molecular targets of gemcitabine action: rationale for development of novel drugs and drug combinations. Curr Pharm Des. 2012;18:2811–29.CrossRefPubMed Elnaggar M, Giovannetti E, Peters GJ. Molecular targets of gemcitabine action: rationale for development of novel drugs and drug combinations. Curr Pharm Des. 2012;18:2811–29.CrossRefPubMed
41.
go back to reference Lei W, Feng XH, Deng WB, Ni H, Zhang ZR, Jia B, et al. Progesterone and DNA damage encourage uterine cell proliferation and decidualization through up-regulating ribonucleotide reductase 2 expression during early pregnancy in mice. J Biol Chem. 2012;287:15174–92.CrossRefPubMedPubMedCentral Lei W, Feng XH, Deng WB, Ni H, Zhang ZR, Jia B, et al. Progesterone and DNA damage encourage uterine cell proliferation and decidualization through up-regulating ribonucleotide reductase 2 expression during early pregnancy in mice. J Biol Chem. 2012;287:15174–92.CrossRefPubMedPubMedCentral
42.
go back to reference Lau MT, Klausen C, Leung PC. E-cadherin inhibits tumor cell growth by suppressing PI3K/Akt signaling via β-catenin-Egr1-mediated PTEN expression. Oncogene. 2011;30:2753–66.CrossRefPubMed Lau MT, Klausen C, Leung PC. E-cadherin inhibits tumor cell growth by suppressing PI3K/Akt signaling via β-catenin-Egr1-mediated PTEN expression. Oncogene. 2011;30:2753–66.CrossRefPubMed
43.
go back to reference Barthel A, Okino ST, Liao J, Nakatani K, Li J, Whitlock Jr JP, et al. Regulation of GLUT1 gene transcription by the serine/threonine kinase Akt1. J Biol Chem. 1999;274:20281–6.CrossRefPubMed Barthel A, Okino ST, Liao J, Nakatani K, Li J, Whitlock Jr JP, et al. Regulation of GLUT1 gene transcription by the serine/threonine kinase Akt1. J Biol Chem. 1999;274:20281–6.CrossRefPubMed
44.
go back to reference Makinoshima H, Takita M, Saruwatari K, Umemura S, Obata Y, Ishii G, et al. Signaling through the phosphatidylinositol 3-Kinase (PI3K)/mammalian target of rapamycin (mTOR) axis is responsible for aerobic glycolysis mediated by glucose transporter in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma. J Biol Chem. 2015;290:17495–504.CrossRefPubMedPubMedCentral Makinoshima H, Takita M, Saruwatari K, Umemura S, Obata Y, Ishii G, et al. Signaling through the phosphatidylinositol 3-Kinase (PI3K)/mammalian target of rapamycin (mTOR) axis is responsible for aerobic glycolysis mediated by glucose transporter in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma. J Biol Chem. 2015;290:17495–504.CrossRefPubMedPubMedCentral
45.
go back to reference Schlieman MG, Fahy BN, Ramsamooj R, Beckett L, Bold RJ. Incidence mechanism and prognostic value of activated AKT in pancreas cancer. Br J Cancer. 2003;89:2110–5.CrossRefPubMedPubMedCentral Schlieman MG, Fahy BN, Ramsamooj R, Beckett L, Bold RJ. Incidence mechanism and prognostic value of activated AKT in pancreas cancer. Br J Cancer. 2003;89:2110–5.CrossRefPubMedPubMedCentral
46.
go back to reference Yamamoto S, Tomita Y, Hoshida Y, Morooka T, Nagano H, Dono K, et al. Prognostic significance of activated Akt expression in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2004;10:2846–50.CrossRefPubMed Yamamoto S, Tomita Y, Hoshida Y, Morooka T, Nagano H, Dono K, et al. Prognostic significance of activated Akt expression in pancreatic ductal adenocarcinoma. Clin Cancer Res. 2004;10:2846–50.CrossRefPubMed
47.
go back to reference Chadha KS, Khoury T, Yu J, Black JD, Gibbs JF, Kuvshinoff BW, et al. Activated Akt and Erk expression and survival after surgery in pancreatic carcinoma. Ann Surg Oncol. 2006;13:933–9.CrossRefPubMed Chadha KS, Khoury T, Yu J, Black JD, Gibbs JF, Kuvshinoff BW, et al. Activated Akt and Erk expression and survival after surgery in pancreatic carcinoma. Ann Surg Oncol. 2006;13:933–9.CrossRefPubMed
48.
go back to reference Jamieson NB, Carter CR, McKay CJ, Oien KA. Tissue biomarkers for prognosis in pancreatic ductal adenocarcinoma: a systematic review and meta-analysis. Clin Cancer Res. 2011;17:3316–31.CrossRefPubMed Jamieson NB, Carter CR, McKay CJ, Oien KA. Tissue biomarkers for prognosis in pancreatic ductal adenocarcinoma: a systematic review and meta-analysis. Clin Cancer Res. 2011;17:3316–31.CrossRefPubMed
49.
go back to reference Loupakis F, Pollina L, Stasi I, Ruzzo A, Scartozzi M, Santini D, et al. PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. J Clin Oncol. 2009;27:2622–9.CrossRefPubMed Loupakis F, Pollina L, Stasi I, Ruzzo A, Scartozzi M, Santini D, et al. PTEN expression and KRAS mutations on primary tumors and metastases in the prediction of benefit from cetuximab plus irinotecan for patients with metastatic colorectal cancer. J Clin Oncol. 2009;27:2622–9.CrossRefPubMed
50.
go back to reference Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.CrossRefPubMed Bailey P, Chang DK, Nones K, Johns AL, Patch AM, Gingras MC, et al. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature. 2016;531:47–52.CrossRefPubMed
51.
go back to reference Carnero A, Blanco-Aparicio C, Renner O, Link W, Leal JF. The PTEN/PI3 K/AKT signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets. 2008;8:187–98.CrossRefPubMed Carnero A, Blanco-Aparicio C, Renner O, Link W, Leal JF. The PTEN/PI3 K/AKT signalling pathway in cancer, therapeutic implications. Curr Cancer Drug Targets. 2008;8:187–98.CrossRefPubMed
52.
go back to reference Sun Y, Wu C, Ma J, Yang Y, Man X, Wu H et al, Toll-like receptor 4 promotes angiogenesis in pancreatic cancer via PI3K/AKT signaling Exp Cell Res. 2016;S0014-482730190-2. Sun Y, Wu C, Ma J, Yang Y, Man X, Wu H et al, Toll-like receptor 4 promotes angiogenesis in pancreatic cancer via PI3K/AKT signaling Exp Cell Res. 2016;S0014-482730190-2.
53.
go back to reference Rios-Marco P, Marco C, Cueto FJ, Carrasco MP, Jimenèz-Lopez JM. Pleiotropic effects of antitumour alkylphospholipids on cholesterol tran sport and metabolism. Exp Cell Res. 2016;340:81–90.CrossRefPubMed Rios-Marco P, Marco C, Cueto FJ, Carrasco MP, Jimenèz-Lopez JM. Pleiotropic effects of antitumour alkylphospholipids on cholesterol tran sport and metabolism. Exp Cell Res. 2016;340:81–90.CrossRefPubMed
54.
go back to reference Gòmez VE, Giovannetti E, Peters GJ. Unraveling the complexity of autophagy: potential therapeutic applications in pancreatic ductal adenocarcinoma. Semin Cancer Biol. 2015;35:11–9.CrossRefPubMed Gòmez VE, Giovannetti E, Peters GJ. Unraveling the complexity of autophagy: potential therapeutic applications in pancreatic ductal adenocarcinoma. Semin Cancer Biol. 2015;35:11–9.CrossRefPubMed
55.
go back to reference Xin Y, Shen XD, Cheng L, Hong DF, Chen B. Perifosine inhibits S6K1-Gli1signaling and enhances gemcitabine-induced anti-pancreatic cancer efficiency. Cancer Chemother Pharmacol. 2014;73:711–9.CrossRefPubMed Xin Y, Shen XD, Cheng L, Hong DF, Chen B. Perifosine inhibits S6K1-Gli1signaling and enhances gemcitabine-induced anti-pancreatic cancer efficiency. Cancer Chemother Pharmacol. 2014;73:711–9.CrossRefPubMed
56.
go back to reference Sharma N, Nanta R, Sharma J, Gunewardena S, Singh KP, Shankar S, et al. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget. 2015;6:32039–60.CrossRefPubMedPubMedCentral Sharma N, Nanta R, Sharma J, Gunewardena S, Singh KP, Shankar S, et al. PI3K/AKT/mTOR and sonic hedgehog pathways cooperate together to inhibit human pancreatic cancer stem cell characteristics and tumor growth. Oncotarget. 2015;6:32039–60.CrossRefPubMedPubMedCentral
57.
go back to reference Wong MH, Xue A, Baxter RC, Pavlakis N, Smith RC. Upstream and downstream co-inhibition of mitogen-activated protein kinase and PI3K/Akt/mTOR pathways in pancreatic ductal adenocarcinoma. Neoplasia. 2016;18:425–35.CrossRefPubMedPubMedCentral Wong MH, Xue A, Baxter RC, Pavlakis N, Smith RC. Upstream and downstream co-inhibition of mitogen-activated protein kinase and PI3K/Akt/mTOR pathways in pancreatic ductal adenocarcinoma. Neoplasia. 2016;18:425–35.CrossRefPubMedPubMedCentral
58.
go back to reference Sun H, Yu T, Li J. Co-administration of perifosine with paclitaxel synergistically induces apoptosis in ovarian cancer cells: more than just AKT inhibition. Cancer Lett. 2011;310:118–28.CrossRefPubMed Sun H, Yu T, Li J. Co-administration of perifosine with paclitaxel synergistically induces apoptosis in ovarian cancer cells: more than just AKT inhibition. Cancer Lett. 2011;310:118–28.CrossRefPubMed
59.
go back to reference Schmidt-Hieber M, Dabrowski R, Weimann A, Aicher B, Lohneis P, Busse A, et al. In vitro cytotoxicity of the novel antimyeloma agents perifosine, bortezomib and lenalidomide against different cell lines. Invest New Drugs. 2012;30:480–9.CrossRefPubMed Schmidt-Hieber M, Dabrowski R, Weimann A, Aicher B, Lohneis P, Busse A, et al. In vitro cytotoxicity of the novel antimyeloma agents perifosine, bortezomib and lenalidomide against different cell lines. Invest New Drugs. 2012;30:480–9.CrossRefPubMed
60.
go back to reference Ciccolini J, Serdjebi C, Peters GJ, Giovannetti E. Pharmacokinetics and pharmacogenetics of gemcitabine as a mainstay in adult and pediatric oncology: an EORTC-PAMM perspective. Cancer Chemother Pharmacol. 2016;78:1–12.CrossRefPubMedPubMedCentral Ciccolini J, Serdjebi C, Peters GJ, Giovannetti E. Pharmacokinetics and pharmacogenetics of gemcitabine as a mainstay in adult and pediatric oncology: an EORTC-PAMM perspective. Cancer Chemother Pharmacol. 2016;78:1–12.CrossRefPubMedPubMedCentral
61.
go back to reference Fujita H, Ohuchida K, Mizumoto K, Itaba S, Ito T, Nakata K, et al. Gene expression levels are predictive markers of outcome in pancreatic cancer after gemcitabine-based adjuvant chemotherapy. Neoplasia. 2010;12:807–17.CrossRefPubMedPubMedCentral Fujita H, Ohuchida K, Mizumoto K, Itaba S, Ito T, Nakata K, et al. Gene expression levels are predictive markers of outcome in pancreatic cancer after gemcitabine-based adjuvant chemotherapy. Neoplasia. 2010;12:807–17.CrossRefPubMedPubMedCentral
62.
go back to reference Irie HY, Pearline RV, Grueneberg D, Hsia M, Ravichandran P, Kothari N, et al. Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J Cell Biol. 2005;171:1023–34.CrossRefPubMedPubMedCentral Irie HY, Pearline RV, Grueneberg D, Hsia M, Ravichandran P, Kothari N, et al. Distinct roles of Akt1 and Akt2 in regulating cell migration and epithelial-mesenchymal transition. J Cell Biol. 2005;171:1023–34.CrossRefPubMedPubMedCentral
63.
go back to reference Toll AD, Dasgupta A, Potoczek M, Yeo CJ, Brody JR, et al. Implications of enhancer of zeste homologue 2 expression in pancreatic ductal adenocarcinoma. Hum Pathol. 2010;41:1205–9.CrossRefPubMed Toll AD, Dasgupta A, Potoczek M, Yeo CJ, Brody JR, et al. Implications of enhancer of zeste homologue 2 expression in pancreatic ductal adenocarcinoma. Hum Pathol. 2010;41:1205–9.CrossRefPubMed
64.
go back to reference Aicher B, Schmidt P, Teifel M, Engel J, Günther E. Perifosine in combination with antimetabolites induces synergistic effects on cytotoxicity and apoptosis in human colon, multiple myeloma, breast, renal, and liver tumor cell lines. Eur J Cancer. 2010;8:67–8.CrossRef Aicher B, Schmidt P, Teifel M, Engel J, Günther E. Perifosine in combination with antimetabolites induces synergistic effects on cytotoxicity and apoptosis in human colon, multiple myeloma, breast, renal, and liver tumor cell lines. Eur J Cancer. 2010;8:67–8.CrossRef
65.
go back to reference Mortenson MM, Galante JG, Gilad O, Schlieman MG, Virudachalam S, Kung HJ, et al. BCL-2 overexpression activates AKT and induces nuclear translocation in pancreatic cancer. J Cell Biochem. 2007;102:1171–9.CrossRefPubMed Mortenson MM, Galante JG, Gilad O, Schlieman MG, Virudachalam S, Kung HJ, et al. BCL-2 overexpression activates AKT and induces nuclear translocation in pancreatic cancer. J Cell Biochem. 2007;102:1171–9.CrossRefPubMed
66.
go back to reference Marsh Rde W, Rocha Lima CM, Levy DE, Mitchell EP, Rowland Jr KM, Benson 3rd AB. A phase II trial of perifosine in locally advanced, unresectable, or metastatic pancreatic adenocarcinoma. Am J Clin Oncol. 2007;30:26–31.CrossRefPubMed Marsh Rde W, Rocha Lima CM, Levy DE, Mitchell EP, Rowland Jr KM, Benson 3rd AB. A phase II trial of perifosine in locally advanced, unresectable, or metastatic pancreatic adenocarcinoma. Am J Clin Oncol. 2007;30:26–31.CrossRefPubMed
67.
go back to reference Soares HP, Ming M, Mellon M, Young SH, Han L, Sinnet-Smith J, et al. Dual PI3K/mTOR inhibitors induce rapid overactivation of the MEK/ERK pathway in human pancreatic cancer cells through suppression of mTORC2. Mol Cancer Ther. 2015;14:1014–23.CrossRefPubMedPubMedCentral Soares HP, Ming M, Mellon M, Young SH, Han L, Sinnet-Smith J, et al. Dual PI3K/mTOR inhibitors induce rapid overactivation of the MEK/ERK pathway in human pancreatic cancer cells through suppression of mTORC2. Mol Cancer Ther. 2015;14:1014–23.CrossRefPubMedPubMedCentral
68.
go back to reference Daemen A, Peterson D, Sahu N, McCord N, Du X, Liu B, et al. Metabolite profiling stratifies pancreatic ductal adenocarcinomas into subtypes with distinct sensitivities to metabolic inhibitors. Proc Natl Acad Sci U S A. 2015;112:E4410–7.CrossRefPubMedPubMedCentral Daemen A, Peterson D, Sahu N, McCord N, Du X, Liu B, et al. Metabolite profiling stratifies pancreatic ductal adenocarcinomas into subtypes with distinct sensitivities to metabolic inhibitors. Proc Natl Acad Sci U S A. 2015;112:E4410–7.CrossRefPubMedPubMedCentral
69.
go back to reference Granchi C, Fancelli D, Minutolo F. An update on therapeutic opportunities offered by cancer glycolytic metabolism. Bioorg Med Chem Lett. 2014;24:4915–25.CrossRefPubMed Granchi C, Fancelli D, Minutolo F. An update on therapeutic opportunities offered by cancer glycolytic metabolism. Bioorg Med Chem Lett. 2014;24:4915–25.CrossRefPubMed
70.
go back to reference Wang YD, Li SJ, Liao JX. Inhibition of glucose transporter 1 (GLUT1) chemosensitized head and neck cancer cells to cisplatin. Technol Cancer Res Treat. 2013;12:525–35.PubMed Wang YD, Li SJ, Liao JX. Inhibition of glucose transporter 1 (GLUT1) chemosensitized head and neck cancer cells to cisplatin. Technol Cancer Res Treat. 2013;12:525–35.PubMed
Metadata
Title
Phospho-Akt overexpression is prognostic and can be used to tailor the synergistic interaction of Akt inhibitors with gemcitabine in pancreatic cancer
Authors
Daniela Massihnia
Amir Avan
Niccola Funel
Mina Maftouh
Anne van Krieken
Carlotta Granchi
Rajiv Raktoe
Ugo Boggi
Babette Aicher
Filippo Minutolo
Antonio Russo
Leticia G. Leon
Godefridus J. Peters
Elisa Giovannetti
Publication date
01-12-2017
Publisher
BioMed Central
Published in
Journal of Hematology & Oncology / Issue 1/2017
Electronic ISSN: 1756-8722
DOI
https://doi.org/10.1186/s13045-016-0371-1

Other articles of this Issue 1/2017

Journal of Hematology & Oncology 1/2017 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