We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Overview and mechanism of action of mTOR inhibitors

    Stéphane Oudard

    Stéphane Oudard is Professor of Oncology and Chief of the Oncology Clinical and Translational Research Unit at the Georges Pompidou Hospital in Paris, France. He received his medical degree from Hôlactone produced by the fungustel-Dieu Hospital, University of Paris, France (1993). On completion of his residency in medical oncology in Paris, he obtained his Masters of Science at Lariboisière-Saint Louis University Hospital, Paris, France (1994) and his Doctorate at Institut Curie, Paris, France (1996). He completed a 2-year fellowship in cancer research at Georgetown University, Washington, DC, USA, before returning to Paris. He has authored more than 125 scientific articles and 30 literature reviews that have been published in various international journals. He is currently a member of the French Cancer Society, European Society for Medical Oncology and the American Society of Clinical Oncology. As a clinical researcher, he has served as a coordinator, investigator, or coinvestigator on several Phase I-III French, European and international clinical trials. He has been largely involved in the development of cabazitaxel, sorafenib, sunitinib and everolimus for treatment of uro–oncology tumors. His research interests include prostate and kidney cancers, translational research, angiogenesis, inhibition of glycolysis and drug resistance. He is the principal investigator (PI) of the Rising PSA Phase III trial and french PI of the PREDICT trial.

    Published Online:https://doi.org/10.2217/ebo.11.208
    Abstract:

    Over recent years, much work on the treatment of solid tumors has focused on the search for inhibitors that reduce tumor vascularity by inhibition of angiogenesis, in view of the well-established role of angiogenesis in the maintenance of solid tumor growth. The first types of angiogenesis inhibitor to be introduced were inhibitors of the tyrosine kinase activity of several growth factor receptors present on the surface of endothelial cells, in particular, the VEGF and PDGF receptor. Examples of tyrosine kinase activity inhibiting VEGF receptors and other growth factor receptors are sorafenib, sunitinib, pazopanib and axitinib [1–4]. Another strategy has been to develop monoclonal antibodies directed against circulating VEGF, namely bevacizumab [5]. Inhibitors that have been more recently developed, however, target the mTOR pathway. mTOR is a serine/threonine kinase discovered empirically during studies on rapamycin (sirolimus). Rapamycin is a macrocyclic lactone produced by the fungus Streptomyces hygroscopicus and has immunosuppressive, antimicrobial and antitumor properties. mTOR has a central regulatory function in the synthesis of key cellular proteins that influence several aspects of cell growth, differentiation, cell cycle progression,angiogenesis, protein degradation and apoptosis [6].It is a downstream effector of the PI3K pathway, known to be dysregulated in a wide range of malignancies, and has thus become an attractive target for inhibition of cell growth, proliferation and angiogenesis. This overview will briefly describe the PI3K/Akt/mTOR pathway; highlight some of the defects in solid tumors that have been linked with dysregulation of this pathway; explain the mechanism of action of mTOR inhibitors; describe approved mTOR inhibitors and their indications; and hint at future directions.

    References

    • Escudier B , Eisen T , Zhao Y , Stadler WM et al. Sorafenib for treatment of renal cell carcinoma: final efficacy and safety results of the Phase 3 treatment approaches in renal cancer global evaluation trial . Clin. Oncol. 27 20 , 3312 – 3318 (2009) .
    • Motzer RJ , Hutson TE , Tomczak P et al. Overall survival and updated results for sunitinib compared with interferon α in patients with metastatic renal-cell carcinoma. . J. Clin. Oncol. 27 22 , 3584 – 3590 (2009) .
    • Sternberg CN , Davis ID , Mardiak J et al. Pazopanib in locally advanced or metastatic renal cell carcinoma: results of a randomized Phase 3 trial. . J. Clin. Oncol. 28 6 , 1061 – 1068 (2010) .
    • Rixe O , Bukowski RM , Michaelson MD et al. Axitinib treatment in patients with cytokine-refractory metastatic renal-cell cancer: a Phase 2 study. . Lancet Oncol. 8 11 , 975 – 984 (2007) .
    • Escudier B , Bellmunt J , Négrier S et al. Phase 3 trial of bevacizumab plus interferon α-2a in patients with metastatic renal cell carcinoma (AVOREN): final analysis of overall survival. . J. Clin. Oncol. 28 13 , 2144 – 2150 (2010) .
    • Zoncu R , Efeyan A . Sabatini DM mTOR: from growth signal integration to cancer, diabetes and ageing . Nat. Rev. Mol. Cell Biol. 12 1 , 21 – 35 (2011) .
    • Caron E , Ghosh S , Matsuoka Y et al. . A comprehensive map of the mTOR signaling network . Mol. Syst. Biol. 6,453 , (2010) .
    • Strimpakos AS , Karapanagiotou EM , Saif WM et al. The role of mTOR in the management of solid tumors: an overview . Cancer Treat. Rev. 35 2 , 148 – 159 (2009) .
    • Brugarolas J , Lei K , Hurley RL , et al. Regulation of mTOR function in response to hypoxia by REDD1 and the TSC1/TSC2 tumor suppressor complex . Genes Dev. 18 23 , 2893 – 2904 (2004) .
    • 10  Di Nicolantonio F , Arena S , Tabernero J et al. Deregulation of the PI3K and KRAS signaling human cancer cells determines their response to everolimus . J. Clin. Invest. 120 8 , 2858 – 2866 (2010) .
    • 11  Beroukhim R , Brunet JP , Di Napoli A et al. Patterns of gene expression and copynumber alterations in von- Hippel Lindau disease associated and sporadic clear cell carcinoma of the kidney . Cancer Res. 69 11 , 4674 – 4681 (2009) .
    • 12  Faivre S , Kroemer G , Raymond E Current development of mTOR inhibitors as anticancer agents . Nat. Rev. 5 8 , 671 – 688 (2006) .
    • 13  Hudes G , Carducci M , Tomczak P et al. Temsirolimus, interferon a, or both for advanced renal-cell carcinoma . N. Engl. J. Med. 356 22 , 2271 – 2281 (2007) .
    • 14  Hess G , Herbrecht R , Romaguera J et al. Phase 3 study to evaluate temsirolimus compared with investigator’s choice therapy for the treatment of relapsed or refractory mantle cell lymphoma . J. Clin. Oncol. 27 23 , 3822 – 3829 (2009) .
    • 15  Motzer RJ , Escudier B , Oudard S et al. For the RECORD-1 study group. Phase 3 trial of everolimus for metastatic renal cell carcinoma. Final results and analysis of prognostic factors . Cancer 116 18 , 4256 – 4265 (2010) .
    • 16  Krueger DA , Care MM , Holland K et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis . N. Engl. J. Med. 363 19 , 1801 – 1811 (2010) .
    • 17  Yao JC , Shah MH , Ito T et al. Randomized, double-blind, placebo-controlled, multicenter Phase 3 trial of everolimus in patients with advanced pancreatic neuroendocrine tumors (PNET) (RADIANT-3). . Ann.Oncol. 21 (Suppl.8) S4 (2010) (Abstract LBA9). .
    • 18  Pavel M , Hainsworth JD , Baudin E et al. Randomized, double-blind, placebocontrolled, multicenter Phase III trial of everolimus + octreotide LAR versus placebo + octreotide LAR in patients with advanced neuroendocrine tumors (NET) (RADIANT-2) . Ann. Oncol. 21 (Suppl.8) S4 (2010) (Abstract LBA8) .
    • 19  Blay JY , Updating progress in sarcoma therapy with mTOR inhibitors . Ann. Oncol. 22 2 , 280 – 287 (2011) .
    • 20  Sankhala K , Mita A , Kelly K et al. The emerging safety profile of mTOR inhibitors, a novel class of anticancer agents . Target Oncol. 4 2 , 135 – 142 (2009) .
    • 21  Oudard S , Medioni J , Aylllon J et al. . Everolimus (RAD001): an mTOR inhibitor for the treatment of metastatic renal cell carcinoma . Expert Rev. Anticancer Ther. 9 6 , 705 – 717 (2009) .
    • 22  White DA , Camus P , Endo M et al. . Noninfectious pneumonitis after everolimus therapy for advanced renal cell carcinoma . Am. J. Respir. Crit. Care Med. 182 3 , 396 – 403 (2010) .
    • 23  Di Lorenzo G , Porta C , Bellmun J et al. . Toxicities of targeted therapy and their management in kidney cancer . Eur. Urol. 59 4 , 526 – 540 (2011) .
    • 24  Ravaud A . Treatment-associated adverse event management in the advanced renal cell carcinoma patient treated with targeted therapies . Oncologist 16 (Suppl.2) , S32 – S44 (2011) .
    • 25  Maroto JP , Hudes G , Dutcher JP et al. Drug-related pneumonitis in patients with advanced renal cell carcinoma treated with temsirolimus . J. Clin. Oncol. 29 13 , 1750 – 1756 (2011) .
    • 26  Cho D , Signoretti S , Dabora S et al. Potential histologic and molecular predictors of response to temsirolimus in patients with advanced renal cell carcinoma . Clin. Genitourin Can. 5 6 , 379 – 385 (2007) .
    • 27  Figlin RA , de Souza P , McDermott D et al. Analysis of PTEN and HIF-1α and correlation with efficacy in patients with advanced renal cell carcinoma treated with temsirolimus versus interferon-α . Cancer 115 16 , 3651 – 3660 (2009) .
    • 28  Tamburini J , Chapuis N , Bardet V et al. Mammalian target of rapamycin (mTOR) inhibition activates phosphatidylinositol 3-kinase/Akt by up-regulating insulinlike growth factor-1 receptor signaling in acute myeloid leukemia: rationale for therapeutic inhibition of both pathways . Blood 111 1 , 379 – 382 (2008) .
    • 29  Swanton C , Larkin JM , Gerlinger M et al. . Predictive biomarker discovery through the parallel integration of clinical trial and functional genomics datasets . Genome Med. 2 8 , 53 (2010) .
    • 30  Heng DY , MacKenzie MJ , Vaishampayan UN et al. . Primary anti-VEGF refractory metastatic renal cell carcinoma (mRCC): clinical characteristics, risk factors and subsequent therapy . J. Clin. Oncol. 29 7S , (2011) (Abstract 305) .
    • 31  Wan X , Harkavy B , Shen N et al. Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism . Oncogene 26 13 , 1932 – 1940 (2007) .
    • 32  Toshi A , Lee E , Gadir N et al. Differential dependence of hypoxia-inducible factors 1 α and 2α on mTORC1 and mTORC2 . J. Biol. Chem. 283 , 34495 – 34499 (2008) .
    • 33  Cho DC , Atkins MB . Future directions in renal cell carcinoma: 2011 and beyond . J. Biol. Chem. 238 50 , 34495 – 34499 (2011) .