Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
1.
Frontiers of Medicine ; (4): 221-231, 2021.
Article in English | WPRIM | ID: wpr-880964

ABSTRACT

The mammalian target of rapamycin (mTOR) critically regulates several essential biological functions, such as cell growth, metabolism, survival, and immune response by forming two important complexes, namely, mTOR complex 1 (mTORC1) and complex 2 (mTORC2). mTOR signaling is often dysregulated in cancers and has been considered an attractive cancer therapeutic target. Great efforts have been made to develop efficacious mTOR inhibitors, particularly mTOR kinase inhibitors, which suppress mTORC1 and mTORC2; however, major success has not been achieved. With the strong scientific rationale, the intriguing question is why cancers are insensitive or not responsive to mTOR-targeted cancer therapy in clinics. Beyond early findings on induced activation of PI3K/Akt, MEK/ERK, and Mnk/eIF4E survival signaling pathways that compromise the efficacy of rapalog-based cancer therapy, recent findings on the essential role of GSK3 in mediating cancer cell response to mTOR inhibitors and mTORC1 inhibition-induced upregulation of PD-L1 in cancer cells may provide some explanations. These new findings may also offer us the opportunity to rationally utilize mTOR inhibitors in cancer therapy. Further elucidation of the biology of complicated mTOR networks may bring us the hope to develop effective therapeutic strategies with mTOR inhibitors against cancer.


Subject(s)
Glycogen Synthase Kinase 3 , Mechanistic Target of Rapamycin Complex 2 , Neoplasms/drug therapy , Phosphatidylinositol 3-Kinases , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-akt , TOR Serine-Threonine Kinases
2.
Chinese Journal of Cancer ; (12): 270-274, 2013.
Article in English | WPRIM | ID: wpr-295796

ABSTRACT

Rapamycin and its derivatives (rapalogs), a group of allosteric inhibitors of mammalian target of rapamycin (mTOR), have been actively tested in a variety of cancer clinical trials, and some have been approved by the Food and Drug Administration for the treatment of certain types of cancers. However, the single agent activity of these compounds in many tumor types remains modest. The mTOR axis is regulated by multiple upstream signaling pathways. Because the genes (e.g., PIK3CA, KRAS, PTEN, and LKB1) that encode key components in these signaling pathways are frequently mutated in human cancers, a subset of cancer types may be addicted to a given mutation, leading to hyperactivation of the mTOR axis. Thus, efforts have been made to demonstrate the potential impact of genetic alterations on rapalog-based or mTOR-targeted cancer therapy. This review will primarily summarize research advances in this direction.


Subject(s)
Humans , Antibiotics, Antineoplastic , Therapeutic Uses , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Mutation , Neoplasms , Drug Therapy , Metabolism , PTEN Phosphohydrolase , Genetics , Metabolism , Phosphatidylinositol 3-Kinases , Genetics , Metabolism , Protein Serine-Threonine Kinases , Genetics , Metabolism , Proto-Oncogene Proteins , Genetics , Metabolism , Proto-Oncogene Proteins p21(ras) , Signal Transduction , Sirolimus , Therapeutic Uses , TOR Serine-Threonine Kinases , Metabolism , ras Proteins , Genetics , Metabolism
SELECTION OF CITATIONS
SEARCH DETAIL