Cargando…

Targeting mTOR and Metabolism in Cancer: Lessons and Innovations

Cancer cells support their growth and proliferation by reprogramming their metabolism in order to gain access to nutrients. Despite the heterogeneity in genetic mutations that lead to tumorigenesis, a common alteration in tumors occurs in pathways that upregulate nutrient acquisition. A central sign...

Descripción completa

Detalles Bibliográficos
Autores principales: Magaway, Cedric, Kim, Eugene, Jacinto, Estela
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6952948/
https://www.ncbi.nlm.nih.gov/pubmed/31817676
http://dx.doi.org/10.3390/cells8121584
_version_ 1783486537872179200
author Magaway, Cedric
Kim, Eugene
Jacinto, Estela
author_facet Magaway, Cedric
Kim, Eugene
Jacinto, Estela
author_sort Magaway, Cedric
collection PubMed
description Cancer cells support their growth and proliferation by reprogramming their metabolism in order to gain access to nutrients. Despite the heterogeneity in genetic mutations that lead to tumorigenesis, a common alteration in tumors occurs in pathways that upregulate nutrient acquisition. A central signaling pathway that controls metabolic processes is the mTOR pathway. The elucidation of the regulation and functions of mTOR can be traced to the discovery of the natural compound, rapamycin. Studies using rapamycin have unraveled the role of mTOR in the control of cell growth and metabolism. By sensing the intracellular nutrient status, mTOR orchestrates metabolic reprogramming by controlling nutrient uptake and flux through various metabolic pathways. The central role of mTOR in metabolic rewiring makes it a promising target for cancer therapy. Numerous clinical trials are ongoing to evaluate the efficacy of mTOR inhibition for cancer treatment. Rapamycin analogs have been approved to treat specific types of cancer. Since rapamycin does not fully inhibit mTOR activity, new compounds have been engineered to inhibit the catalytic activity of mTOR to more potently block its functions. Despite highly promising pre-clinical studies, early clinical trial results of these second generation mTOR inhibitors revealed increased toxicity and modest antitumor activity. The plasticity of metabolic processes and seemingly enormous capacity of malignant cells to salvage nutrients through various mechanisms make cancer therapy extremely challenging. Therefore, identifying metabolic vulnerabilities in different types of tumors would present opportunities for rational therapeutic strategies. Understanding how the different sources of nutrients are metabolized not just by the growing tumor but also by other cells from the microenvironment, in particular, immune cells, will also facilitate the design of more sophisticated and effective therapeutic regimen. In this review, we discuss the functions of mTOR in cancer metabolism that have been illuminated from pre-clinical studies. We then review key findings from clinical trials that target mTOR and the lessons we have learned from both pre-clinical and clinical studies that could provide insights on innovative therapeutic strategies, including immunotherapy to target mTOR signaling and the metabolic network in cancer.
format Online
Article
Text
id pubmed-6952948
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-69529482020-01-23 Targeting mTOR and Metabolism in Cancer: Lessons and Innovations Magaway, Cedric Kim, Eugene Jacinto, Estela Cells Review Cancer cells support their growth and proliferation by reprogramming their metabolism in order to gain access to nutrients. Despite the heterogeneity in genetic mutations that lead to tumorigenesis, a common alteration in tumors occurs in pathways that upregulate nutrient acquisition. A central signaling pathway that controls metabolic processes is the mTOR pathway. The elucidation of the regulation and functions of mTOR can be traced to the discovery of the natural compound, rapamycin. Studies using rapamycin have unraveled the role of mTOR in the control of cell growth and metabolism. By sensing the intracellular nutrient status, mTOR orchestrates metabolic reprogramming by controlling nutrient uptake and flux through various metabolic pathways. The central role of mTOR in metabolic rewiring makes it a promising target for cancer therapy. Numerous clinical trials are ongoing to evaluate the efficacy of mTOR inhibition for cancer treatment. Rapamycin analogs have been approved to treat specific types of cancer. Since rapamycin does not fully inhibit mTOR activity, new compounds have been engineered to inhibit the catalytic activity of mTOR to more potently block its functions. Despite highly promising pre-clinical studies, early clinical trial results of these second generation mTOR inhibitors revealed increased toxicity and modest antitumor activity. The plasticity of metabolic processes and seemingly enormous capacity of malignant cells to salvage nutrients through various mechanisms make cancer therapy extremely challenging. Therefore, identifying metabolic vulnerabilities in different types of tumors would present opportunities for rational therapeutic strategies. Understanding how the different sources of nutrients are metabolized not just by the growing tumor but also by other cells from the microenvironment, in particular, immune cells, will also facilitate the design of more sophisticated and effective therapeutic regimen. In this review, we discuss the functions of mTOR in cancer metabolism that have been illuminated from pre-clinical studies. We then review key findings from clinical trials that target mTOR and the lessons we have learned from both pre-clinical and clinical studies that could provide insights on innovative therapeutic strategies, including immunotherapy to target mTOR signaling and the metabolic network in cancer. MDPI 2019-12-06 /pmc/articles/PMC6952948/ /pubmed/31817676 http://dx.doi.org/10.3390/cells8121584 Text en © 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/).
spellingShingle Review
Magaway, Cedric
Kim, Eugene
Jacinto, Estela
Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
title Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
title_full Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
title_fullStr Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
title_full_unstemmed Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
title_short Targeting mTOR and Metabolism in Cancer: Lessons and Innovations
title_sort targeting mtor and metabolism in cancer: lessons and innovations
topic Review
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6952948/
https://www.ncbi.nlm.nih.gov/pubmed/31817676
http://dx.doi.org/10.3390/cells8121584
work_keys_str_mv AT magawaycedric targetingmtorandmetabolismincancerlessonsandinnovations
AT kimeugene targetingmtorandmetabolismincancerlessonsandinnovations
AT jacintoestela targetingmtorandmetabolismincancerlessonsandinnovations