Introduction: The mTOR Pathway and its clinical importance
In 1994 a protein was discovered that was determined to be a target for the small molecule inhibitor Rapamycin, it was named mTOR [1]. The acronym mTOR stands for the “mammalian Target for Rapamycin” and this protein was demonstrated to be essential for embryonic development, mutated null murine models did not survive in utero [2;3]. Subsequent investigation revealed that the mTOR protein is like the “Grand central station” of multiple regulatory pathways dealing mostly with gene transcription and cell growth. The unique aspect of the mTOR protein is that despite being more related to lipid kinases it also has functions similar to those of the serine/threonine kinase family phosphoinositide 3-kinase related kinases (PIKK) [4]. The mTOR protein contains multiple binding domains that interact with several different ligands that initiate scaffolding processes, adapt other mechanism or regulate mTOR’s activity. mTOR’s activation is more complex in that signals directly from the plasma in relation to either nutritional status or stress conditions can trigger mTOR activation. In addition signals from growth factors, insulin and phorbol 12-myristate 13-acetate (PMA) trigger signaling cascades such as PCK/PLD/mTOR or PI3K/PIP/TSC/Rheb/mTOR that effect the function of mTOR [5]. The pathways effected are EGFR [6;7] or AKT [8], effects are involved in the organization of actin and initiation of the start codon (AUG) in gene transcription [9]. Due to the nature of the activity of mTOR the concept of an mTOR pathway inhibitor was considered an attractive approach to the treatment of various cancers. With the knowledge that mTOR inhibition by rapamycin, the core molecular structure of this molecule could be used to design alternative mTOR kinase inhibitors [10]. Examples of mTOR pathway inhibitors are Rapamycin, chrysorphanic acid, deforolimus, Everolimus and Temsirolimus are just a few of the 20 plus more commonly known molecules. Clinical mTOR selective inhibitors have been used in the treatment of cancer, diabetes, obesity, transplant rejection, glycogen storage diseases and in age related disease.
mTOR inhibitors: FDA approval and clinical status
The determination of the mTOR structure as a complex with a ligand called raptor led to the discovery that the complex was actually a dimer formation of interlocking mTOR and raptor subunits surrounding a central cavity (mTORC1or 2). Inhibitors form complexes with FKBP12 which in turn disrupt the cohesion of the dimer complex mTORC leading to inhibition effects. include Everolimus (
Sirolimus (Rapamycin) is an mTOR antagonist FDA approved in the maintenance treatment after kidney transplants. In addition over fifty clinical trials are currently in progress in a range of disease conditions covering immunotherapy after bone marrow, kidney, liver, pancreases and stem cell transplants. Sirolimus mTOR inhibitor in the treatment of cancer is being investigated for lymphoma, glioblastoma, NSCLC, renal carcinoma, advanced solid tumors, diabetes (focusing on Macular disorders), metastatic cancer and osteosarcoma [11-18]. The range of investigation indicates the possibilities of mTOR inhibitor drugs in the treatment of metabolic and nutritional disorders. Other mTOR inhibitor in clinical trials include Everolimus (with 60 + trials covering breast, prostate, pancreatic, gastric, endometrial, renal colorectal and leukemia’s of various different types) [19-30]; the mTOR agonist Temsirolimus with 60+ trials covering similar cancer type as to Sirolimus and Everolimus [31-41]; the mTOR specific inhibitor Deforolimus with 30 + trials covering, in addition to those all ready mentioned, NSCLC and ovarian [42-44].
With the treatment of cancer nutritional status and individual stress are compromised, mTOR is directly affected by these factors and many others that tumors or treatment itself change. Activity of mTOR function can therefore be different in the various stages of tumor development. The mTOR inhibitor mechanism is difficult to predict and the mechanism of resistance to treatment are difficult to isolate [45-49]. Results of easy to use mTOR activity assay enable researchers to monitor mTOR activtity during treatment and the various stages of tumor development or necrosis. To buy mTOR inhibitors or an mTOR assay kit can be costly and prices vary significantly between suppliers.
References
1. Sabatini DD. In awe of subcellular complexity: 50 years of trespassing boundaries within the cell. Annu Rev Cell Dev Biol 2005; 21:1-33.
2. Gangloff YG, Mueller M et al. Disruption of the mouse mTOR gene leads to early postimplantation lethality and prohibits embryonic stem cell development. Mol Cell Biol 2004; 24(21):9508-9516.
3. Murakami M, Ichisaka T et al. mTOR is essential for growth and proliferation in early mouse embryos and embryonic stem cells. Mol Cell Biol 2004; 24(15):6710-6718.
4. Harris TE, Lawrence JC, Jr. TOR signaling. Sci STKE 2003; 2003(212):re15.
5. Caron E, Ghosh S et al. A comprehensive map of the mTOR signaling network. Mol Syst Biol 2010; 6:453.
6. Steelman LS, Chappell WH et al. Roles of the Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR pathways in controlling growth and sensitivity to therapy-implications for cancer and aging. Aging (Albany NY) 2011; 3(3):192-222.
7. Chappell WH, Steelman LS et al. Ras/Raf/MEK/ERK and PI3K/PTEN/Akt/mTOR inhibitors: rationale and importance to inhibiting these pathways in human health. Oncotarget 2011; 2(3):135-164.
8. Karar J, Maity A. PI3K/AKT/mTOR Pathway in Angiogenesis. Front Mol Neurosci 2011; 4:51.
9. Liu L, Parent CA. Review series: TOR kinase complexes and cell migration. J Cell Biol 2011; 194(6):815-824.
10. Carraway H, Hidalgo M. New targets for therapy in breast cancer: mammalian target of rapamycin (mTOR) antagonists. Breast Cancer Res 2004; 6(5):219-224.
11. Buhaescu I, Izzedine H et al. Sirolimus--challenging current perspectives. Ther Drug Monit 2006; 28(5):577-584.
12. Rangan GK. Sirolimus-associated proteinuria and renal dysfunction. Drug Saf 2006; 29(12):1153-1161.
13. Kneteman NM, Oberholzer J et al. Sirolimus-based immunosuppression for liver transplantation in the presence of extended criteria for hepatocellular carcinoma. Liver Transpl 2004; 10(10):1301-1311.
14. Paghdal KV, Schwartz RA. Sirolimus (rapamycin): from the soil of Easter Island to a bright future. J Am Acad Dermatol 2007; 57(6):1046-1050.
15. Campsen J, Zimmerman MA et al. Sirolimus and liver transplantation: clinical implications for hepatocellular carcinoma. Expert Opin Pharmacother 2007; 8(9):1275-1282.
16. Cutler C, Antin JH. Sirolimus immunosuppression for graft-versus-host disease prophylaxis and therapy: an update. Curr Opin Hematol 2010; 17(6):500-504.
17. Sanchez-Plumed JA, Gonzalez MM et al. Sirolimus, the first mTOR inhibitor. Nefrologia 2006; 26 Suppl 2:21-32.
18. Lee VW, Chapman JR. Sirolimus: its role in nephrology. Nephrology (Carlton ) 2005; 10(6):606-614.
19. Houghton PJ. Everolimus. Clin Cancer Res 2010; 16(5):1368-1372.
20. Anandappa G, Hollingdale A et al. Everolimus - a new approach in the treatment of renal cell carcinoma. Cancer Manag Res 2010; 2:61-70.
21. Agarwala SS, Case S. Everolimus (RAD001) in the treatment of advanced renal cell carcinoma: a review. Oncologist 2010; 15(3):236-245.
22. Oudard S, Medioni J et al. Everolimus (RAD001): an mTOR inhibitor for the treatment of metastatic renal cell carcinoma. Expert Rev Anticancer Ther 2009; 9(6):705-717.
23. Krueger DA, Care MM et al. Everolimus for subependymal giant-cell astrocytomas in tuberous sclerosis. N Engl J Med 2010; 363(19):1801-1811.
24. Amato R. Everolimus for the treatment of advanced renal cell carcinoma. Expert Opin Pharmacother 2011; 12(7):1143-1155.
25. Pavel ME, Hainsworth JD et al. Everolimus plus octreotide long-acting repeatable for the treatment of advanced neuroendocrine tumours associated with carcinoid syndrome (RADIANT-2): a randomised, placebo-controlled, phase 3 study. Lancet 2011; 378(9808):2005-2012.
26. Gabardi S, Baroletti SA. Everolimus: a proliferation signal inhibitor with clinical applications in organ transplantation, oncology, and cardiology. Pharmacotherapy 2010; 30(10):1044-1056.
27. Patel JK, Kobashigawa JA. Everolimus: an immunosuppressive agent in transplantation. Expert Opin Pharmacother 2006; 7(10):1347-1355.
28. Franz DN. Everolimus: an mTOR inhibitor for the treatment of tuberous sclerosis. Expert Rev Anticancer Ther 2011; 11(8):1181-1192.
29. Sanchez-Fructuoso AI. Everolimus: an update on the mechanism of action, pharmacokinetics and recent clinical trials. Expert Opin Drug Metab Toxicol 2008; 4(6):807-819.
30. Schaffer SA, Ross HJ. Everolimus: efficacy and safety in cardiac transplantation. Expert Opin Drug Saf 2010; 9(5):843-854.
31. Stock C, Zaccagnini M et al. Temsirolimus. Recent Results Cancer Res 2010; 184:189-197.
32. Hess G. Temsirolimus for the treatment of mantle cell lymphoma. Expert Rev Hematol 2009; 2(6):631-640.
33. Hess G, Smith SM et al. Temsirolimus in mantle cell lymphoma and other non-Hodgkin lymphoma subtypes. Semin Oncol 2009; 36 Suppl 3:S37-S45.
34. Bhatia S, Thompson JA. Temsirolimus in patients with advanced renal cell carcinoma: an overview. Adv Ther 2009; 26(1):55-67.
35. Gore ME. Temsirolimus in the treatment of advanced renal cell carcinoma. Ann Oncol 2007; 18 Suppl 9:ix87-ix88.
36. Mackenzie MJ, Rini BI et al. Temsirolimus in VEGF-refractory metastatic renal cell carcinoma. Ann Oncol 2011; 22(1):145-148.
37. Rini BI. Temsirolimus, an inhibitor of mammalian target of rapamycin. Clin Cancer Res 2008; 14(5):1286-1290.
38. Malizzia LJ, Hsu A. Temsirolimus, an mTOR inhibitor for treatment of patients with advanced renal cell carcinoma. Clin J Oncol Nurs 2008; 12(4):639-646.
39. Temsirolimus: CCI 779, CCI-779, cell cycle inhibitor-779. Drugs R D 2004; 5(6):363-367.
40. Simpson D, Curran MP. Temsirolimus: in advanced renal cell carcinoma. Drugs 2008; 68(5):631-638.
41. Hoy SM, McKeage K. Temsirolimus: In relapsed and/or refractory mantle cell lymphoma. Drugs 2010; 70(14):1819-1829.
42. Mita M, Sankhala K et al. Deforolimus (AP23573) a novel mTOR inhibitor in clinical development. Expert Opin Investig Drugs 2008; 17(12):1947-1954.
43. Ridaforolimus. Drugs R D 2010; 10(3):165-178.
44. Dancey JE, Monzon J. Ridaforolimus: a promising drug in the treatment of soft-tissue sarcoma and other malignancies. Future Oncol 2011; 7(7):827-839.
45. Xing D, Orsulic S. Modeling resistance to pathway-targeted therapy in ovarian cancer. Cell Cycle 2005; 4(8):1004-1006.
46. Rini BI. New strategies in kidney cancer: therapeutic advances through understanding the molecular basis of response and resistance. Clin Cancer Res 2010; 16(5):1348-1354.
47. Lefranc F, Brotchi J et al. Possible future issues in the treatment of glioblastomas: special emphasis on cell migration and the resistance of migrating glioblastoma cells to apoptosis. J Clin Oncol 2005; 23(10):2411-2422.
48. Rini BI, Atkins MB. Resistance to targeted therapy in renal-cell carcinoma. Lancet Oncol 2009; 10(10):992-1000.
49. Jiang BH, Liu LZ. Role of mTOR in anticancer drug resistance: perspectives for improved drug treatment. Drug Resist Updat 2008; 11(3):63-76.
Related Products
Cat.No. |
Product Name |
Information |
S2406
|
Chrysophanic Acid
|
Chrysophanic Acid (Chrysophanol) a natural anthraquinone isolated from Dianella longifolia, is a EGFR/mTOR pathway inhibitor. |
Related Targets
mTOR