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

mTOR inhibitors in renal cell carcinoma

    Piotr J Wysocki

    Department of Cancer Immunology, Chair of Medical Biotechnology, University of Medical Sciences, Department of Chemotherapy, Great Poland Cancer Center, Poland.

    Published Online:https://doi.org/10.2217/ebo.11.172
    Abstract:

    Progress in the molecular biology of cancer has made possible the discovery of signaling pathways involved in the development and progression of renal cell carcinoma (RCC). One of the most important pathways in the biology of RCC is the PI3K/Akt/mTOR. mTOR is a serine–threonine kinase involved in coupling growth and metabolic signals into stimulation of proliferation and gene expression in cancer and endothelial cells, which ultimately leads to tumor progression. mTOR inhibitors have been tested in several Phase II and III clinical trials in patients with metastatic RCC. Temsirolimus has been demonstrated to significantly improve overall survival and progression-free survival (PFS) of poor-risk metastatic RCC patients compared with interferon (IFN). The efficacy of temsirolimus has been observed both in nephrectomized and non-nephrectomized patients irrespective of the histological type of RCC. Everolimus administered to metastatic RCC patients following failure of one or two anti-VEGFR tyrosine kinase inhibitor-based regimens has made a significant prolongation of PFS compared with placebo possible.

    References

    • Vivanco I , Sawyers CL . The phosphatidylinositol 3-kinase AKT pathway in human cancer . Nat. Rev. Cancer 2 (7) , 489 – 501 (2002) .
    • Rivet J , Mourah S , Murata H et al. VEGF and VEGFR-1 are coexpressed by epithelial and stromal cells of renal cell carcinoma . Cancer 112 (2) , 433 – 442 (2008) .
    • Tawfik OW , Kramer B , Shideler B , Danley M , Kimler BF , Holzbeierlein J . Prognostic significance of CD44, platelet-derived growth factor receptor α, and cyclooxygenase 2 expression in renal cell carcinoma . Arch. Pathol. Lab. Med. 131 (2) , 261 – 267 (2007) .
    • Franovic A , Gunaratnam L , Smith K , Robert I , Patten D , Lee S . Translational up-regulation of the EGFR by tumor hypoxia provides a nonmutational explanation for its overexpression in human cancer . Proc. Natl Acad. Sci. USA 104 (32) , 13092 – 13097 (2007) .
    • Xiao GH , Jeffers M , Bellacosa A , Mitsuuchi Y , Vande Woude GF , Testa JR . Anti-apoptotic signaling by hepatocyte growth factor/Met via the phosphatidylinositol 3-kinase/Akt and mitogen-activated protein kinase pathways . Proc. Natl Acad. Sci. USA 98 (1) , 247 – 252 (2001) .
    • Bellacosa A , Chan TO , Ahmed NN et al. Akt activation by growth factors is a multiple-step process: the role of the PH domain . Oncogene 17 (3) , 313 – 325 (1998) .
    • Blume-Jensen P , Hunter T . Oncogenic kinase signaling . Nature 411 (6835) , 355 – 365 (2001) .
    • Schmelzle T , Hall MN . TOR, a central controller of cell growth . Cell 103 (2) , 253 – 262 (2000) .
    • Thomas G , Hall MN . TOR signaling and control of cell growth . Curr. Opin. Cell. Biol. 9 (6) , 782 – 787 (1997) .
    • 10  Faivre S , Kroemer G , Raymond E . Current development of mTOR inhibitors as anticancer agents . Nat. Rev. Drug Discov. 5 (8) , 671 – 688 (2006) .
    • 11  Kim DH , Sarbassov DD , Ali SM et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery . Cell 110 (2) , 163 – 175 (2002) .
    • 12  Hudson CC , Liu M , Chiang GG et al. Regulation of hypoxia-inducible factor 1α expression and function by the mammalian target of rapamycin . Mol. Cell. Biol. 22 (20) , 7004 – 7014 (2002) .
    • 13  Toschi A , Lee E , Gadir N , Ohh M , Foster DA . Differential dependence of hypoxia-inducible factors 1{α} and 2{α} on mTORC1 and mTORC2 . J. Biol. Chem. 283 (50) , 34495 – 34499 (2008) .
    • 14  Adjei AA , Hidalgo M . Intracellular signal transduction pathway proteins as targets for cancer therapy . J. Clin. Oncol. 23 (23) , 5386 – 5403 (2005) .
    • 15  Sarbassov DD , Ali SM , Sengupta S et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB . Mol. Cell 22 (2) , 159 – 168 (2006) .
    • 16  Stoeltzing O , Meric-Bernstam F , Ellis LM . Intracellular signaling in tumor and endothelial cells: the expected and, yet again, the unexpected . Cancer Cell 10 (2) , 89 – 91 (2006) .
    • 17  Gerber HP , McMurtrey A , Kowalski J et al. Vascular endothelial growth factor regulates endothelial cell survival through the phosphatidylinositol 3′-kinase/Akt signal transduction pathway. Requirement for Flk-1/KDR activation . J. Biol. Chem. 273 (46) , 30336 – 30343 (1998) .
    • 18  Jain RK . Normalization of tumor vasculature: an emerging concept in antiangiogenic therapy . Science 307 (5706) , 58 – 62 (2005) .
    • 19  Balcarcel RR , Stephanopoulos G . Rapamycin reduces hybridoma cell death and enhances monoclonal antibody production . Biotechnol. Bioeng. 76 (1) , 1 – 10 (2001) .
    • 20  Yatscoff RW , LeGatt DF , Kneteman NM . Therapeutic monitoring of rapamycin: a new immunosuppressive drug . Ther. Drug Monit. 15 (6) , 478 – 482 (1993) .
    • 21  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) .
    • 22  Mekhail TM , Abou-Jawde RM , Boumerhi G et al. Validation and extension of the Memorial Sloan-Kettering prognostic factors model for survival in patients with previously untreated metastatic renal cell carcinoma . J. Clin. Oncol. 23 (4) , 832 – 841 (2005) .
    • 23  Dutcher JP , de Souza P , McDermott D et al. Effect of temsirolimus versus interferon-a on outcome of patients with advanced renal cell carcinoma of different tumor histologies . Med. Oncol. 26 (2) , 202 – 209 (2009) .
    • 24  Logan TF , McDermott D , Dutcher JP , Makhson A , Mikulas J . Exploratory analysis of the influence of nephrectomy status on temsirolimus efficacy in patients with advanced renal cell carcinoma and poor-risk features. Proceedings of ASCO Annual Meeting 2008 . J. Clin. Oncol. 26 (15S) , (2008) (Abstract 5050).
    • 25  Jac J , Giessinger SK , Khan M , Wills J , Chiang S . A Phase II trial of RAD001 in patients (pts) with metastatic renal cell carcinoma (MRCC). Proceedings of ASCO Annual Meeting 2007 . J. Clin. Oncol. 25 , (2007) (Abstract 5107) .
    • 26  Motzer RJ , Escudier B , Oudard S et al. Efficacy of everolimus in advanced renal cell carcinoma: a double-blind, randomised, placebo-controlled Phase III trial . Lancet 372 (9637) , 449 – 456 (2008) .
    • 27  Motzer RJ , Escudier B , Oudard S et al. Phase 3 trial of everolimus for metastatic renal cell carcinoma: final results and analysis of prognostic factors . Cancer 116 (18) , 4256 – 4265 (2010) .
    • 28  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. Cancer 5 (6) , 379 – 385 (2007) .