PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition
Abstract
:1. Introduction
2. PTEN Pathway
3. PTEN Alterations
4. PTEN Loss can Modify the Tumor Microenvironment
4.1. PTEN Loss and Alteration of T-Cell Activity in Melanoma Patients
4.2. PTEN Loss and Tumor Microenvironment Modification in Glioblastoma
4.3. PTEN Loss and Tumor Senescent Phenotype in Prostate Cancer
4.4. PTEN Loss and Modification of T-Cell Function in Leiomyosarcoma
4.5. PTEN Loss in Lung Carcinoma
5. Correlation between PTEN and PD-L1
6. Discussions
7. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Gide, T.N.; Wilmott, J.S.; Scolyer, R.A.; Long, G.V. Primary and Acquired Resistance to Immune Checkpoint Inhibitors in Metastatic Melanoma. Clin. Cancer Res. 2018, 24, 1260–1270. [Google Scholar] [CrossRef] [PubMed]
- Conway, J.R.; Kofman, E.; Mo, S.S.; Elmarakeby, H.; Van Allen, E. Genomics of response to immune checkpoint therapies for cancer: Implications for precision medicine. Genome Med. 2018, 10, 93. [Google Scholar] [CrossRef]
- Shin, D.S.; Zaretsky, J.M.; Escuin-Ordinas, H.; Garcia-Diaz, A.; Hu-Lieskovan, S.; Kalbasi, A.; Grasso, C.S.; Hugo, W.; Sandoval, S.; Torrejon, D.Y.; et al. Primary Resistance to PD-1 Blockade Mediated by JAK1/2 Mutations. Cancer Discov. 2017, 7, 188–201. [Google Scholar] [CrossRef] [PubMed]
- Fumarola, C.; Bonelli, M.A.; Petronini, P.G.; Alfieri, R.R. Targeting PI3K/AKT/mTOR pathway in non small cell lung cancer. Biochem. Pharmacol. 2014, 90, 197–207. [Google Scholar] [CrossRef] [PubMed]
- Manning, B.D.; Toker, A. AKT/PKB Signaling: Navigating the Network. Cell 2017, 169, 381–405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Papa, A.; Pandolfi, P.P. The PTEN(-)PI3K Axis in Cancer. Biomolecules 2019, 9, 153. [Google Scholar] [CrossRef] [PubMed]
- Liu, P.; Gan, W.; Chin, Y.R.; Ogura, K.; Guo, J.; Zhang, J.; Wang, B.; Blenis, J.; Cantley, L.C.; Toker, A.; et al. PtdIns(3,4,5)P3-Dependent Activation of the mTORC2 Kinase Complex. Cancer Discov. 2015, 5, 1194–1209. [Google Scholar] [CrossRef]
- Guertin, D.A.; Sabatini, D.M. Defining the role of mTOR in cancer. Cancer Cell 2007, 12, 9–22. [Google Scholar] [CrossRef] [PubMed]
- Song, M.S.; Salmena, L.; Pandolfi, P.P. The functions and regulation of the PTEN tumour suppressor. Nat. Rev. Mol. Cell Biol. 2012, 13, 283–296. [Google Scholar] [CrossRef] [PubMed]
- Hollander, M.C.; Blumenthal, G.M.; Dennis, P.A. PTEN loss in the continuum of common cancers, rare syndromes and mouse models. Nat. Rev. Cancer 2011, 11, 289–301. [Google Scholar] [CrossRef]
- Lee, Y.R.; Chen, M.; Pandolfi, P.P. The functions and regulation of the PTEN tumour suppressor: New modes and prospects. Nat. Rev. Mol. Cell Biol. 2018, 19, 547–562. [Google Scholar] [CrossRef] [PubMed]
- Lee, J.O.; Yang, H.; Georgescu, M.M.; Di Cristofano, A.; Maehama, T.; Shi, Y.; Dixon, J.E.; Pandolfi, P.; Pavletich, N.P. Crystal structure of the PTEN tumor suppressor: Implications for its phosphoinositide phosphatase activity and membrane association. Cell 1999, 99, 323–334. [Google Scholar] [CrossRef]
- Miinea, C.P.; Sano, H.; Kane, S.; Sano, E.; Fukuda, M.; Peranen, J.; Lane, W.S.; Lienhard, G.E. AS160, the Akt substrate regulating GLUT4 translocation, has a functional Rab GTPase-activating protein domain. Biochem. J. 2005, 391, 87–93. [Google Scholar] [CrossRef] [PubMed]
- Zoncu, R.; Efeyan, A.; Sabatini, D.M. mTOR: From growth signal integration to cancer, diabetes and ageing. Nat. Rev. Mol. Cell Biol. 2011, 12, 21–35. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.L.; Liu, J.; Lei, S.; Zhang, J.; Zhou, W.; Yu, H.G. PTEN inhibits the invasion and metastasis of gastric cancer via downregulation of FAK expression. Cell. Signal. 2014, 26, 1011–1020. [Google Scholar] [CrossRef]
- Cavazzoni, A.; La Monica, S.; Alfieri, R.; Ravelli, A.; Van Der Steen, N.; Sciarrillo, R.; Madeddu, D.; Lagrasta, C.A.M.; Quaini, F.; Bonelli, M.; et al. Enhanced efficacy of AKT and FAK kinase combined inhibition in squamous cell lung carcinomas with stable reduction in PTEN. Oncotarget 2017, 8, 53068–53083. [Google Scholar] [CrossRef] [Green Version]
- Rayess, H.; Wang, M.B.; Srivatsan, E.S. Cellular senescence and tumor suppressor gene p16. Int. J. Cancer 2012, 130, 1715–1725. [Google Scholar] [CrossRef]
- Alimonti, A.; Nardella, C.; Chen, Z.; Clohessy, J.G.; Carracedo, A.; Trotman, L.C.; Cheng, K.; Varmeh, S.; Kozma, S.C.; Thomas, G.; et al. A novel type of cellular senescence that can be enhanced in mouse models and human tumor xenografts to suppress prostate tumorigenesis. J. Clin. Investig. 2010, 120, 681–693. [Google Scholar] [CrossRef]
- Pilarski, R. PTEN Hamartoma Tumor Syndrome: A Clinical Overview. Cancers 2019, 11, 844. [Google Scholar] [CrossRef]
- Orloff, M.S.; Eng, C. Genetic and phenotypic heterogeneity in the PTEN hamartoma tumour syndrome. Oncogene 2008, 27, 5387–5397. [Google Scholar] [CrossRef] [Green Version]
- Bonneau, D.; Longy, M. Mutations of the human PTEN gene. Hum. Mutat. 2000, 16, 109–122. [Google Scholar] [CrossRef]
- Milella, M.; Falcone, I.; Conciatori, F.; Cesta Incani, U.; Del Curatolo, A.; Inzerilli, N.; Nuzzo, C.M.; Vaccaro, V.; Vari, S.; Cognetti, F.; et al. PTEN: Multiple Functions in Human Malignant Tumors. Front. Oncol. 2015, 5, 24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alvarez-Garcia, V.; Tawil, Y.; Wise, H.M.; Leslie, N.R. Mechanisms of PTEN loss in cancer: It’s all about diversity. Semin. Cancer Biol. 2019. Available online: https://www.sciencedirect.com/science/article/pii/S1044579X18300592?via%3Dihub (accessed on 7 February 2019). [CrossRef] [PubMed]
- Soria, J.C.; Lee, H.Y.; Lee, J.I.; Wang, L.; Issa, J.P.; Kemp, B.L.; Liu, D.D.; Kurie, J.M.; Mao, L.; Khuri, F.R. Lack of PTEN expression in non-small cell lung cancer could be related to promoter methylation. Clin. Cancer Res. 2002, 8, 1178–1184. [Google Scholar] [PubMed]
- Mirmohammadsadegh, A.; Marini, A.; Nambiar, S.; Hassan, M.; Tannapfel, A.; Ruzicka, T.; Hengge, U.R. Epigenetic silencing of the PTEN gene in melanoma. Cancer Res. 2006, 66, 6546–6552. [Google Scholar] [CrossRef]
- Escriva, M.; Peiro, S.; Herranz, N.; Villagrasa, P.; Dave, N.; Montserrat-Sentis, B.; Murray, S.A.; Franci, C.; Gridley, T.; Virtanen, I.; et al. Repression of PTEN phosphatase by Snail1 transcriptional factor during gamma radiation-induced apoptosis. Mol. Cell Biol. 2008, 28, 1528–1540. [Google Scholar] [CrossRef] [PubMed]
- Hettinger, K.; Vikhanskaya, F.; Poh, M.K.; Lee, M.K.; de Belle, I.; Zhang, J.T.; Reddy, S.A.; Sabapathy, K. c-Jun promotes cellular survival by suppression of PTEN. Cell Death Differ. 2007, 14, 218–229. [Google Scholar] [CrossRef]
- Ciuffreda, L.; Di Sanza, C.; Cesta Incani, U.; Eramo, A.; Desideri, M.; Biagioni, F.; Passeri, D.; Falcone, I.; Sette, G.; Bergamo, P.; et al. The mitogen-activated protein kinase (MAPK) cascade controls phosphatase and tensin homolog (PTEN) expression through multiple mechanisms. J. Mol. Med. (Berl.) 2012, 90, 667–679. [Google Scholar] [CrossRef]
- Xia, D.; Srinivas, H.; Ahn, Y.H.; Sethi, G.; Sheng, X.; Yung, W.K.; Xia, Q.; Chiao, P.J.; Kim, H.; Brown, P.H.; et al. Mitogen-activated protein kinase kinase-4 promotes cell survival by decreasing PTEN expression through an NF kappa B-dependent pathway. J. Biol. Chem. 2007, 282, 3507–3519. [Google Scholar] [CrossRef]
- Stambolic, V.; MacPherson, D.; Sas, D.; Lin, Y.; Snow, B.; Jang, Y.; Benchimol, S.; Mak, T.W. Regulation of PTEN transcription by p53. Mol. Cell 2001, 8, 317–325. [Google Scholar] [CrossRef]
- Virolle, T.; Adamson, E.D.; Baron, V.; Birle, D.; Mercola, D.; Mustelin, T.; de Belle, I. The Egr-1 transcription factor directly activates PTEN during irradiation-induced signalling. Nat. Cell Biol. 2001, 3, 1124–1128. [Google Scholar] [CrossRef] [PubMed]
- Patel, L.; Pass, I.; Coxon, P.; Downes, C.P.; Smith, S.A.; Macphee, C.H. Tumor suppressor and anti-inflammatory actions of PPARgamma agonists are mediated via upregulation of PTEN. Curr. Biol. 2001, 11, 764–768. [Google Scholar] [CrossRef]
- Nakanishi, A.; Kitagishi, Y.; Ogura, Y.; Matsuda, S. The tumor suppressor PTEN interacts with p53 in hereditary cancer (Review). Int. J. Oncol. 2014, 44, 1813–1819. [Google Scholar] [CrossRef] [PubMed]
- Musilova, K.; Mraz, M. MicroRNAs in B-cell lymphomas: How a complex biology gets more complex. Leukemia 2015, 29, 1004–1017. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.G.; Wang, J.J.; Zhao, F.; Liu, Q.; Jiang, K.; Yang, G.H. MicroRNA-21 (miR-21) represses tumor suppressor PTEN and promotes growth and invasion in non-small cell lung cancer (NSCLC). Clin. Chim. Acta 2010, 411, 846–852. [Google Scholar] [CrossRef] [PubMed]
- Miller, S.J.; Lou, D.Y.; Seldin, D.C.; Lane, W.S.; Neel, B.G. Direct identification of PTEN phosphorylation sites. FEBS Lett. 2002, 528, 145–153. [Google Scholar] [CrossRef] [Green Version]
- Al-Khouri, A.M.; Ma, Y.; Togo, S.H.; Williams, S.; Mustelin, T. Cooperative phosphorylation of the tumor suppressor phosphatase and tensin homologue (PTEN) by casein kinases and glycogen synthase kinase 3beta. J. Biol. Chem. 2005, 280, 35195–35202. [Google Scholar] [CrossRef]
- Torres, J.; Pulido, R. The tumor suppressor PTEN is phosphorylated by the protein kinase CK2 at its C terminus. Implications for PTEN stability to proteasome-mediated degradation. J. Biol. Chem. 2001, 276, 993–998. [Google Scholar] [CrossRef]
- Correia, N.C.; Girio, A.; Antunes, I.; Martins, L.R.; Barata, J.T. The multiple layers of non-genetic regulation of PTEN tumour suppressor activity. Eur. J. Cancer 2014, 50, 216–225. [Google Scholar] [CrossRef]
- Okumura, K.; Mendoza, M.; Bachoo, R.M.; DePinho, R.A.; Cavenee, W.K.; Furnari, F.B. PCAF modulates PTEN activity. J. Biol. Chem. 2006, 281, 26562–26568. [Google Scholar] [CrossRef]
- Lee, S.R.; Yang, K.S.; Kwon, J.; Lee, C.; Jeong, W.; Rhee, S.G. Reversible inactivation of the tumor suppressor PTEN by H2O2. J. Biol. Chem. 2002, 277, 20336–20342. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.; Trotman, L.C.; Koppie, T.; Alimonti, A.; Chen, Z.; Gao, Z.; Wang, J.; Erdjument-Bromage, H.; Tempst, P.; Cordon-Cardo, C.; et al. NEDD4-1 is a proto-oncogenic ubiquitin ligase for PTEN. Cell 2007, 128, 129–139. [Google Scholar] [CrossRef]
- Amodio, N.; Scrima, M.; Palaia, L.; Salman, A.N.; Quintiero, A.; Franco, R.; Botti, G.; Pirozzi, P.; Rocco, G.; De Rosa, N.; et al. Oncogenic role of the E3 ubiquitin ligase NEDD4-1, a PTEN negative regulator, in non-small-cell lung carcinomas. Am. J. Pathol. 2010, 177, 2622–2634. [Google Scholar] [CrossRef]
- Wu, X.; Hepner, K.; Castelino-Prabhu, S.; Do, D.; Kaye, M.B.; Yuan, X.J.; Wood, J.; Ross, C.; Sawyers, C.L.; Whang, Y.E. Evidence for regulation of the PTEN tumor suppressor by a membrane-localized multi-PDZ domain containing scaffold protein MAGI-2. Proc. Natl. Acad. Sci. USA 2000, 97, 4233–4238. [Google Scholar] [CrossRef] [Green Version]
- Stephens, P.J.; Tarpey, P.S.; Davies, H.; Van Loo, P.; Greenman, C.; Wedge, D.C.; Nik-Zainal, S.; Martin, S.; Varela, I.; Bignell, G.R.; et al. The landscape of cancer genes and mutational processes in breast cancer. Nature 2012, 486, 400–404. [Google Scholar] [CrossRef] [PubMed]
- Pandolfi, P.P. Breast cancer--loss of PTEN predicts resistance to treatment. N. Engl. J. Med. 2004, 351, 2337–2338. [Google Scholar] [CrossRef] [PubMed]
- Nagata, Y.; Lan, K.H.; Zhou, X.; Tan, M.; Esteva, F.J.; Sahin, A.A.; Klos, K.S.; Li, P.; Monia, B.P.; Nguyen, N.T.; et al. PTEN activation contributes to tumor inhibition by trastuzumab, and loss of PTEN predicts trastuzumab resistance in patients. Cancer Cell 2004, 6, 117–127. [Google Scholar] [CrossRef] [Green Version]
- Marsit, C.J.; Zheng, S.; Aldape, K.; Hinds, P.W.; Nelson, H.H.; Wiencke, J.K.; Kelsey, K.T. PTEN expression in non-small-cell lung cancer: Evaluating its relation to tumor characteristics, allelic loss, and epigenetic alteration. Hum. Pathol. 2005, 36, 768–776. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.P.; Loukola, A.; Salovaara, R.; Nystrom-Lahti, M.; Peltomaki, P.; de la Chapelle, A.; Aaltonen, L.A.; Eng, C. PTEN mutational spectra, expression levels, and subcellular localization in microsatellite stable and unstable colorectal cancers. Am. J. Pathol. 2002, 161, 439–447. [Google Scholar] [CrossRef]
- Molinari, F.; Frattini, M. Functions and Regulation of the PTEN Gene in Colorectal Cancer. Front. Oncol. 2013, 3, 326. [Google Scholar] [CrossRef]
- Dankort, D.; Curley, D.P.; Cartlidge, R.A.; Nelson, B.; Karnezis, A.N.; Damsky, W.E., Jr.; You, M.J.; DePinho, R.A.; McMahon, M.; Bosenberg, M. Braf(V600E) cooperates with Pten loss to induce metastatic melanoma. Nat. Genet. 2009, 41, 544–552. [Google Scholar] [CrossRef] [PubMed]
- Koul, D. PTEN signaling pathways in glioblastoma. Cancer Biol. Ther. 2008, 7, 1321–1325. [Google Scholar] [CrossRef] [PubMed]
- Endersby, R.; Baker, S.J. PTEN signaling in brain: Neuropathology and tumorigenesis. Oncogene 2008, 27, 5416–5430. [Google Scholar] [CrossRef] [PubMed]
- Roychowdhury, S.; Chinnaiyan, A.M. Advancing precision medicine for prostate cancer through genomics. J. Clin. Oncol. 2013, 31, 1866–1873. [Google Scholar] [CrossRef] [PubMed]
- Taylor, B.S.; Schultz, N.; Hieronymus, H.; Gopalan, A.; Xiao, Y.; Carver, B.S.; Arora, V.K.; Kaushik, P.; Cerami, E.; Reva, B.; et al. Integrative genomic profiling of human prostate cancer. Cancer Cell 2010, 18, 11–22. [Google Scholar] [CrossRef]
- Martins, F.C.; Santiago, I.; Trinh, A.; Xian, J.; Guo, A.; Sayal, K.; Jimenez-Linan, M.; Deen, S.; Driver, K.; Mack, M.; et al. Combined image and genomic analysis of high-grade serous ovarian cancer reveals PTEN loss as a common driver event and prognostic classifier. Genome Biol. 2014, 15, 526. [Google Scholar] [CrossRef] [PubMed]
- Kurose, K.; Zhou, X.P.; Araki, T.; Cannistra, S.A.; Maher, E.R.; Eng, C. Frequent loss of PTEN expression is linked to elevated phosphorylated Akt levels, but not associated with p27 and cyclin D1 expression, in primary epithelial ovarian carcinomas. Am. J. Pathol. 2001, 158, 2097–2106. [Google Scholar] [CrossRef]
- Wang, Y.; Zhao, S.; Zhu, L.; Zhang, Q.; Ren, Y. MiR-19a negatively regulated the expression of PTEN and promoted the growth of ovarian cancer cells. Gene 2018, 670, 166–173. [Google Scholar] [CrossRef]
- Aguirre, A.J.; Brennan, C.; Bailey, G.; Sinha, R.; Feng, B.; Leo, C.; Zhang, Y.; Zhang, J.; Gans, J.D.; Bardeesy, N.; et al. High-resolution characterization of the pancreatic adenocarcinoma genome. Proc. Natl. Acad. Sci. USA 2004, 101, 9067–9072. [Google Scholar] [CrossRef] [Green Version]
- Perren, A.; Komminoth, P.; Saremaslani, P.; Matter, C.; Feurer, S.; Lees, J.A.; Heitz, P.U.; Eng, C. Mutation and expression analyses reveal differential subcellular compartmentalization of PTEN in endocrine pancreatic tumors compared to normal islet cells. Am. J. Pathol. 2000, 157, 1097–1103. [Google Scholar] [CrossRef]
- Durinck, S.; Stawiski, E.W.; Pavia-Jimenez, A.; Modrusan, Z.; Kapur, P.; Jaiswal, B.S.; Zhang, N.; Toffessi-Tcheuyap, V.; Nguyen, T.T.; Pahuja, K.B.; et al. Spectrum of diverse genomic alterations define non-clear cell renal carcinoma subtypes. Nat. Genet. 2015, 47, 13–21. [Google Scholar] [CrossRef] [PubMed]
- Dellas, A.; Jundt, G.; Sartorius, G.; Schneider, M.; Moch, H. Combined PTEN and p27kip1 protein expression patterns are associated with obesity and prognosis in endometrial carcinomas. Clin. Cancer Res. 2009, 15, 2456–2462. [Google Scholar] [CrossRef]
- Roh, W.; Chen, P.L.; Reuben, A.; Spencer, C.N.; Prieto, P.A.; Miller, J.P.; Gopalakrishnan, V.; Wang, F.; Cooper, Z.A.; Reddy, S.M.; et al. Integrated molecular analysis of tumor biopsies on sequential CTLA-4 and PD-1 blockade reveals markers of response and resistance. Sci. Transl. Med. 2017, 9, eaah3560. [Google Scholar] [CrossRef] [PubMed]
- Aguissa-Toure, A.H.; Li, G. Genetic alterations of PTEN in human melanoma. Cell. Mol. Life Sci. 2012, 69, 1475–1491. [Google Scholar] [CrossRef] [PubMed]
- Bucheit, A.D.; Chen, G.; Siroy, A.; Tetzlaff, M.; Broaddus, R.; Milton, D.; Fox, P.; Bassett, R.; Hwu, P.; Gershenwald, J.E.; et al. Complete loss of PTEN protein expression correlates with shorter time to brain metastasis and survival in stage IIIB/C melanoma patients with BRAFV600 mutations. Clin. Cancer Res. 2014, 20, 5527–5536. [Google Scholar] [CrossRef]
- Peng, W.; Chen, J.Q.; Liu, C.; Malu, S.; Creasy, C.; Tetzlaff, M.T.; Xu, C.; McKenzie, J.A.; Zhang, C.; Liang, X.; et al. Loss of PTEN Promotes Resistance to T Cell-Mediated Immunotherapy. Cancer Discov. 2016, 6, 202–216. [Google Scholar] [CrossRef] [PubMed]
- Shrimali, R.K.; Yu, Z.; Theoret, M.R.; Chinnasamy, D.; Restifo, N.P.; Rosenberg, S.A. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res. 2010, 70, 6171–6180. [Google Scholar] [CrossRef]
- Gilbert, M.R.; Dignam, J.J.; Armstrong, T.S.; Wefel, J.S.; Blumenthal, D.T.; Vogelbaum, M.A.; Colman, H.; Chakravarti, A.; Pugh, S.; Won, M.; et al. A randomized trial of bevacizumab for newly diagnosed glioblastoma. N. Engl. J. Med. 2014, 370, 699–708. [Google Scholar] [CrossRef]
- Filley, A.C.; Henriquez, M.; Dey, M. Recurrent glioma clinical trial, CheckMate-143: The game is not over yet. Oncotarget 2017, 8, 91779–91794. [Google Scholar] [CrossRef]
- Parsa, A.T.; Waldron, J.S.; Panner, A.; Crane, C.A.; Parney, I.F.; Barry, J.J.; Cachola, K.E.; Murray, J.C.; Tihan, T.; Jensen, M.C.; et al. Loss of tumor suppressor PTEN function increases B7-H1 expression and immunoresistance in glioma. Nat. Med. 2007, 13, 84–88. [Google Scholar] [CrossRef]
- Zhao, J.; Chen, A.X.; Gartrell, R.D.; Silverman, A.M.; Aparicio, L.; Chu, T.; Bordbar, D.; Shan, D.; Samanamud, J.; Mahajan, A.; et al. Immune and genomic correlates of response to anti-PD-1 immunotherapy in glioblastoma. Nat. Med. 2019, 25, 462–469. [Google Scholar] [CrossRef] [PubMed]
- Yang, L.; Zhang, Y. Tumor-associated macrophages: From basic research to clinical application. J. Hematol. Oncol. 2017, 10, 58. [Google Scholar] [CrossRef] [PubMed]
- Pearson, H.B.; Li, J.; Meniel, V.S.; Fennell, C.M.; Waring, P.; Montgomery, K.G.; Rebello, R.J.; Macpherson, A.A.; Koushyar, S.; Furic, L.; et al. Identification of Pik3ca Mutation as a Genetic Driver of Prostate Cancer That Cooperates with Pten Loss to Accelerate Progression and Castration-Resistant Growth. Cancer Discov. 2018, 8, 764–779. [Google Scholar] [CrossRef] [PubMed]
- Grasso, C.S.; Wu, Y.M.; Robinson, D.R.; Cao, X.; Dhanasekaran, S.M.; Khan, A.P.; Quist, M.J.; Jing, X.; Lonigro, R.J.; Brenner, J.C.; et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature 2012, 487, 239–243. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toso, A.; Revandkar, A.; Di Mitri, D.; Guccini, I.; Proietti, M.; Sarti, M.; Pinton, S.; Zhang, J.; Kalathur, M.; Civenni, G.; et al. Enhancing chemotherapy efficacy in Pten-deficient prostate tumors by activating the senescence-associated antitumor immunity. Cell Rep. 2014, 9, 75–89. [Google Scholar] [CrossRef] [PubMed]
- Antonarakis, E.S.; Keizman, D.; Zhang, Z.; Gurel, B.; Lotan, T.L.; Hicks, J.L.; Fedor, H.L.; Carducci, M.A.; De Marzo, A.M.; Eisenberger, M.A. An immunohistochemical signature comprising PTEN, MYC, and Ki67 predicts progression in prostate cancer patients receiving adjuvant docetaxel after prostatectomy. Cancer 2012, 118, 6063–6071. [Google Scholar] [CrossRef] [PubMed]
- Bard-Chapeau, E.A.; Li, S.; Ding, J.; Zhang, S.S.; Zhu, H.H.; Princen, F.; Fang, D.D.; Han, T.; Bailly-Maitre, B.; Poli, V.; et al. Ptpn11/Shp2 acts as a tumor suppressor in hepatocellular carcinogenesis. Cancer Cell 2011, 19, 629–639. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.J.; Li, Y.; Chen, W.S.; Liang, Y.; Wang, G.; Zong, M.; Kaneko, K.; Xu, R.; Karin, M.; Feng, G.S. Shp2 deletion in hepatocytes suppresses hepatocarcinogenesis driven by oncogenic beta-Catenin, PIK3CA and MET. J. Hepatol. 2018, 69, 79–88. [Google Scholar] [CrossRef]
- Maki, R.G.; Jungbluth, A.A.; Gnjatic, S.; Schwartz, G.K.; D’Adamo, D.R.; Keohan, M.L.; Wagner, M.J.; Scheu, K.; Chiu, R.; Ritter, E.; et al. A Pilot Study of Anti-CTLA4 Antibody Ipilimumab in Patients with Synovial Sarcoma. Sarcoma 2013, 2013, 168145. [Google Scholar] [CrossRef]
- George, S.; Miao, D.; Demetri, G.D.; Adeegbe, D.; Rodig, S.J.; Shukla, S.; Lipschitz, M.; Amin-Mansour, A.; Raut, C.P.; Carter, S.L.; et al. Loss of PTEN Is Associated with Resistance to Anti-PD-1 Checkpoint Blockade Therapy in Metastatic Uterine Leiomyosarcoma. Immunity 2017, 46, 197–204. [Google Scholar] [CrossRef] [Green Version]
- Salmena, L.; Carracedo, A.; Pandolfi, P.P. Tenets of PTEN tumor suppression. Cell 2008, 133, 403–414. [Google Scholar] [CrossRef] [PubMed]
- Cancer Genome Atlas Research Network. Comprehensive genomic characterization of squamous cell lung cancers. Nature 2012, 489, 519–525. [Google Scholar] [CrossRef] [PubMed]
- Xu, C.; Fillmore, C.M.; Koyama, S.; Wu, H.; Zhao, Y.; Chen, Z.; Herter-Sprie, G.S.; Akbay, E.A.; Tchaicha, J.H.; Altabef, A.; et al. Loss of Lkb1 and Pten leads to lung squamous cell carcinoma with elevated PD-L1 expression. Cancer Cell 2014, 25, 590–604. [Google Scholar] [CrossRef]
- Wu, L.; Saxena, S.; Awaji, M.; Singh, R.K. Tumor-Associated Neutrophils in Cancer: Going Pro. Cancers 2019, 11, 564. [Google Scholar] [CrossRef]
- Batlle, E.; Massague, J. Transforming Growth Factor-beta Signaling in Immunity and Cancer. Immunity 2019, 50, 924–940. [Google Scholar] [CrossRef] [PubMed]
- Jones, S.A.; Jenkins, B.J. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef]
- Kerr, K.M.; Thunnissen, E.; Dafni, U.; Finn, S.P.; Bubendorf, L.; Soltermann, A.; Verbeken, E.; Biernat, W.; Warth, A.; Marchetti, A.; et al. A retrospective cohort study of PD-L1 prevalence, molecular associations and clinical outcomes in patients with NSCLC: Results from the European Thoracic Oncology Platform (ETOP) Lungscape Project. Lung Cancer 2019, 131, 95–103. [Google Scholar] [CrossRef]
- Hlaing, A.M.; Furusato, B.; Udo, E.; Kitamura, Y.; Souda, M.; Masutani, M.; Fukuoka, J. Expression of phosphatase and tensin homolog and programmed cell death ligand 1 in adenosquamous carcinoma of the lung. Biochem. Biophys. Res. Commun. 2018, 503, 2764–2769. [Google Scholar] [CrossRef]
- Parikh, A.R.; Ali, S.M.; Schrock, A.B.; Albacker, L.A.; Miller, V.A.; Stephens, P.J.; Crilley, P.; Markman, M. Response to rapamycin analogs but not PD-1 inhibitors in PTEN-mutated metastatic non-small-cell lung cancer with high tumor mutational burden. Lung Cancer (Auckl.) 2018, 9, 45–47. [Google Scholar] [CrossRef]
- Sun, C.; Mezzadra, R.; Schumacher, T.N. Regulation and Function of the PD-L1 Checkpoint. Immunity 2018, 48, 434–452. [Google Scholar] [CrossRef] [Green Version]
- Lastwika, K.J.; Wilson, W., III; Li, Q.K.; Norris, J.; Xu, H.; Ghazarian, S.R.; Kitagawa, H.; Kawabata, S.; Taube, J.M.; Yao, S.; et al. Control of PD-L1 Expression by Oncogenic Activation of the AKT-mTOR Pathway in Non-Small Cell Lung Cancer. Cancer Res. 2016, 76, 227–238. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Chen, D.; Lu, B.; Wang, C.; Zhang, J.; Huang, L.; Wang, X.; Timmons, C.L.; Hu, J.; Liu, B.; et al. PTEN loss increases PD-L1 protein expression and affects the correlation between PD-L1 expression and clinical parameters in colorectal cancer. PLoS ONE 2013, 8, e65821. [Google Scholar] [CrossRef] [PubMed]
- Oki, E.; Okano, S.; Saeki, H.; Umemoto, Y.; Teraishi, K.; Nakaji, Y.; Ando, K.; Zaitsu, Y.; Yamashita, N.; Sugiyama, M.; et al. Protein Expression of Programmed Death 1 Ligand 1 and HER2 in Gastric Carcinoma. Oncology 2017, 93, 387–394. [Google Scholar] [CrossRef]
- Cedres, S.; Ponce-Aix, S.; Pardo-Aranda, N.; Navarro-Mendivil, A.; Martinez-Marti, A.; Zugazagoitia, J.; Sansano, I.; Montoro, M.A.; Enguita, A.; Felip, E. Analysis of expression of PTEN/PI3K pathway and programmed cell death ligand 1 (PD-L1) in malignant pleural mesothelioma (MPM). Lung Cancer 2016, 96, 1–6. [Google Scholar] [CrossRef]
- Gyori, D.; Chessa, T.; Hawkins, P.T.; Stephens, L.R. Class (I) Phosphoinositide 3-Kinases in the Tumor Microenvironment. Cancers 2017, 9, 24. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Zhang, S.; Yao, J.; Lowery, F.J.; Zhang, Q.; Huang, W.C.; Li, P.; Li, M.; Wang, X.; Zhang, C.; et al. Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature 2015, 527, 100–104. [Google Scholar] [CrossRef] [PubMed]
- Gonzalez-Angulo, A.M.; Ferrer-Lozano, J.; Stemke-Hale, K.; Sahin, A.; Liu, S.; Barrera, J.A.; Burgues, O.; Lluch, A.M.; Chen, H.; Hortobagyi, G.N.; et al. PI3K pathway mutations and PTEN levels in primary and metastatic breast cancer. Mol. Cancer Ther. 2011, 10, 1093–1101. [Google Scholar] [CrossRef]
- Wikman, H.; Lamszus, K.; Detels, N.; Uslar, L.; Wrage, M.; Benner, C.; Hohensee, I.; Ylstra, B.; Eylmann, K.; Zapatka, M.; et al. Relevance of PTEN loss in brain metastasis formation in breast cancer patients. Breast Cancer Res. 2012, 14, R49. [Google Scholar] [CrossRef] [PubMed]
- Janku, F. Phosphoinositide 3-kinase (PI3K) pathway inhibitors in solid tumors: From laboratory to patients. Cancer Treat. Rev. 2017, 59, 93–101. [Google Scholar] [CrossRef] [Green Version]
- Janku, F.; Hong, D.S.; Fu, S.; Piha-Paul, S.A.; Naing, A.; Falchook, G.S.; Tsimberidou, A.M.; Stepanek, V.M.; Moulder, S.L.; Lee, J.J.; et al. Assessing PIK3CA and PTEN in early-phase trials with PI3K/AKT/mTOR inhibitors. Cell Rep. 2014, 6, 377–387. [Google Scholar] [CrossRef] [PubMed]
- Brown, K.K.; Toker, A. The phosphoinositide 3-kinase pathway and therapy resistance in cancer. F1000Prime Rep. 2015, 7, 13. [Google Scholar] [CrossRef] [PubMed]
- Wee, S.; Wiederschain, D.; Maira, S.M.; Loo, A.; Miller, C.; de Beaumont, R.; Stegmeier, F.; Yao, Y.M.; Lengauer, C. PTEN-deficient cancers depend on PIK3CB. Proc. Natl. Acad. Sci. USA 2008, 105, 13057–13062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jia, S.; Liu, Z.; Zhang, S.; Liu, P.; Zhang, L.; Lee, S.H.; Zhang, J.; Signoretti, S.; Loda, M.; Roberts, T.M.; et al. Essential roles of PI(3)K-p110beta in cell growth, metabolism and tumorigenesis. Nature 2008, 454, 776–779. [Google Scholar] [CrossRef] [PubMed]
- Lim, E.L.; Okkenhaug, K. Phosphoinositide 3-kinase delta is a regulatory T-cell target in cancer immunotherapy. Immunology 2019, 157, 210–218. [Google Scholar] [CrossRef] [PubMed]
- Coussens, L.M.; Zitvogel, L.; Palucka, A.K. Neutralizing tumor-promoting chronic inflammation: A magic bullet? Science 2013, 339, 286–291. [Google Scholar] [CrossRef] [PubMed]
- De Henau, O.; Rausch, M.; Winkler, D.; Campesato, L.F.; Liu, C.; Cymerman, D.H.; Budhu, S.; Ghosh, A.; Pink, M.; Tchaicha, J.; et al. Overcoming resistance to checkpoint blockade therapy by targeting PI3Kgamma in myeloid cells. Nature 2016, 539, 443–447. [Google Scholar] [CrossRef] [PubMed]
- Davis, R.J.; Moore, E.C.; Clavijo, P.E.; Friedman, J.; Cash, H.; Chen, Z.; Silvin, C.; Van Waes, C.; Allen, C. Anti-PD-L1 Efficacy Can Be Enhanced by Inhibition of Myeloid-Derived Suppressor Cells with a Selective Inhibitor of PI3Kdelta/gamma. Cancer Res. 2017, 77, 2607–2619. [Google Scholar] [CrossRef] [PubMed]
- Soler, A.; Angulo-Urarte, A.; Graupera, M. PI3K at the crossroads of tumor angiogenesis signaling pathways. Mol. Cell. Oncol. 2015, 2, e975624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stanczuk, L.; Martinez-Corral, I.; Ulvmar, M.H.; Zhang, Y.; Lavina, B.; Fruttiger, M.; Adams, R.H.; Saur, D.; Betsholtz, C.; Ortega, S.; et al. cKit Lineage Hemogenic Endothelium-Derived Cells Contribute to Mesenteric Lymphatic Vessels. Cell Rep. 2015, 10, 1708–1721. [Google Scholar] [CrossRef] [PubMed]
- Soler, A.; Serra, H.; Pearce, W.; Angulo, A.; Guillermet-Guibert, J.; Friedman, L.S.; Vinals, F.; Gerhardt, H.; Casanovas, O.; Graupera, M.; et al. Inhibition of the p110alpha isoform of PI 3-kinase stimulates nonfunctional tumor angiogenesis. J. Exp. Med. 2013, 210, 1937–1945. [Google Scholar] [CrossRef]
- Trimboli, A.J.; Cantemir-Stone, C.Z.; Li, F.; Wallace, J.A.; Merchant, A.; Creasap, N.; Thompson, J.C.; Caserta, E.; Wang, H.; Chong, J.L.; et al. Pten in stromal fibroblasts suppresses mammary epithelial tumours. Nature 2009, 461, 1084–1091. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sai, J.; Owens, P.; Novitskiy, S.V.; Hawkins, O.E.; Vilgelm, A.E.; Yang, J.; Sobolik, T.; Lavender, N.; Johnson, A.C.; McClain, C.; et al. PI3K Inhibition Reduces Mammary Tumor Growth and Facilitates Antitumor Immunity and Anti-PD1 Responses. Clin. Cancer Res. 2017, 23, 3371–3384. [Google Scholar] [CrossRef] [PubMed]
- Marshall, N.A.; Galvin, K.C.; Corcoran, A.M.; Boon, L.; Higgs, R.; Mills, K.H. Immunotherapy with PI3K inhibitor and Toll-like receptor agonist induces IFN-gamma+IL-17+ polyfunctional T cells that mediate rejection of murine tumors. Cancer Res. 2012, 72, 581–591. [Google Scholar] [CrossRef] [PubMed]
- Alfieri, R.; Giovannetti, E.; Bonelli, M.; Cavazzoni, A. New Treatment Opportunities in Phosphatase and Tensin Homolog (PTEN)-Deficient Tumors: Focus on PTEN/Focal Adhesion Kinase Pathway. Front. Oncol. 2017, 7, 170. [Google Scholar] [CrossRef] [PubMed]
- Serrels, A.; Lund, T.; Serrels, B.; Byron, A.; McPherson, R.C.; von Kriegsheim, A.; Gomez-Cuadrado, L.; Canel, M.; Muir, M.; Ring, J.E.; et al. Nuclear FAK controls chemokine transcription, Tregs, and evasion of anti-tumor immunity. Cell 2015, 163, 160–173. [Google Scholar] [CrossRef] [PubMed]
- Serrels, B.; McGivern, N.; Canel, M.; Byron, A.; Johnson, S.C.; McSorley, H.J.; Quinn, N.; Taggart, D.; Von Kreigsheim, A.; Anderton, S.M.; et al. IL-33 and ST2 mediate FAK-dependent antitumor immune evasion through transcriptional networks. Sci. Signal. 2017, 10, eaan8355. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, R.W.; Barbie, D.A.; Flaherty, K.T. Mechanisms of resistance to immune checkpoint inhibitors. Br. J. Cancer 2018, 118, 9–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Tumor Type | Mutation (%) | Deletion (%) | LOH (%) | Promoter Methylation (%) | miRNA Alteration | Reference |
---|---|---|---|---|---|---|
Breast cancer | 3–5 | 20–40 | 30–40 | 30–50 | miR-106b, -93 up-regulation | [45,46,47] |
Lung cancer | 6–9 predominantly in SQCLC | 20–40 | 20–40 | 20–30 | miR-21 up-regulation | [10,22,24,45,48] |
Colorectal cancer | 10–20 | 5–15 | 10–20 | 10–20 | miR-21, -32 up-regulation | [10,22,49,50] |
Melanoma | 10–20 | 5–10 | 20–40 | 30–50 | miR-25, -221, -222 up-regulation | [10,22,25,28,51] |
Glioma | 30–40 | 40–70 | 50–70 | 5–10 | miR-26a up-regulation | [10,22,52,53] |
Prostate cancer | 5–15 | 10–20 | 20–50 | 1–5 | miR-22 up-regulation | [10,22,54,55] |
Ovarian | 1–5 | 30–40 | 30–50 | 5–10 | miR-19a, -21, -214 up-regulation | [10,22,56,57,58] |
Pancreatic cancer | <1 | 15–20 | 30–50 | <1 | miR-21 up-regulation | [10,22,59,60] |
Kidney cancer | <1 | 1–5 | 20–30 | <1 | miR-23b-5p up-regulation | [10,22,61,62] |
Agent | Target | in Combination with | Tumors | Phase | Reference |
---|---|---|---|---|---|
Capivasertib (AZD5363) | AKT | Olaparib + Durvalumab | Advanced solid tumors | I | NCT03772561 |
Copanlisib (BAY80–6946) | PI3K | Nivolumab | Colorectal | I/II | NCT03711058 |
Olaparib + Durvalumab | Solid tumors | 1b | NCT03842228 | ||
Nivolumab | Solid tumors and lymphomas | 1b | NCT03502733 | ||
IPI-549 | PI3K-γ | Atezolizumab + Nab-Paclitaxel Atezolizumab + Bevacizumab | Breast cancer Renal cell carcinoma | II | NCT03961698 |
Nivolumab | Solid tumors | I | NCT02637531 | ||
GSK2636771 | PI3K-β | Pembrolizumab | Melanoma | I/II | NCT03131908 |
Ipatasertib (RG7440) | AKT | Atezolizumab | Solid tumors | I | NCT03673787 |
Atezolizumab | Breast cancer | 1b | NCT03800836 | ||
Idelalisib (GS-1101) | PI3K-δ | Pembrolizumab | Lung | 1b/2 | NCT03257722 |
SF1126 | PI3 Kinase and Bromodomain Inhibitor | Nivolumab | HCC | I | NCT03059147 |
Tenalisib (RP6530) | PI3K-δ/γ | Pembrolizumab | cHL | I/II | NCT03471351 |
Copanlisib (BAY80–6946) | PI3K | Nivolumab | Colorectal | I/II | NCT03711058 |
Olaparib + Durvalumab | Solid tumors | 1b | NCT03842228 | ||
Nivolumab | Solid tumors and lymphomas | 1b | NCT03502733 |
© 2019 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Cretella, D.; Digiacomo, G.; Giovannetti, E.; Cavazzoni, A. PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition. Cancers 2019, 11, 1318. https://doi.org/10.3390/cancers11091318
Cretella D, Digiacomo G, Giovannetti E, Cavazzoni A. PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition. Cancers. 2019; 11(9):1318. https://doi.org/10.3390/cancers11091318
Chicago/Turabian StyleCretella, Daniele, Graziana Digiacomo, Elisa Giovannetti, and Andrea Cavazzoni. 2019. "PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition" Cancers 11, no. 9: 1318. https://doi.org/10.3390/cancers11091318
APA StyleCretella, D., Digiacomo, G., Giovannetti, E., & Cavazzoni, A. (2019). PTEN Alterations as a Potential Mechanism for Tumor Cell Escape from PD-1/PD-L1 Inhibition. Cancers, 11(9), 1318. https://doi.org/10.3390/cancers11091318