Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 1/2016

01-01-2016 | Focussed Research Review

Cholesterol metabolites and tumor microenvironment: the road towards clinical translation

Authors: Laura Raccosta, Raffaella Fontana, Gianfranca Corna, Daniela Maggioni, Marta Moresco, Vincenzo Russo

Published in: Cancer Immunology, Immunotherapy | Issue 1/2016

Login to get access

Abstract

Targeting the tumor microenvironment focusing on immune cells has recently become a standard of care for some tumors. Indeed, antibodies blocking immune checkpoints (e.g., anti-CTLA-4 and anti-PD1 mAbs) have been approved by regulatory agencies for the treatment of some solid tumors based upon successes in many clinical trials. Although tumor metabolism has always attracted the attention of tumor biologists, only recently have oncologists renewed their interest in this field of tumor biology research. This has highlighted the possibility to pharmacologically target rate-limiting enzymes along key metabolic pathways of tumor cells, such as lipogenesis and aerobic glycolysis. Altered tumor metabolism has also been shown to influence the functionality of the tumor microenvironment as a whole, particularly the immune cell component of thereof. Cholesterol, oxysterols and Liver X receptors (LXRs) have been investigated in different tumor models. Recent in vitro and in vivo results point to their involvement in tumor and immune cell biology, thus making the LXR/oxysterol axis a possible target for novel antitumor strategies. Indeed, the possibility to target both tumor cell metabolism (i.e., cholesterol metabolism) and tumor-infiltrating immune cell dysfunctions induced by oxysterols might result in a synergistic antitumor effect generating long-lasting memory responses. This review will focus on the role of cholesterol metabolism with particular emphasis on the role of the LXR/oxysterol axis in the tumor microenvironment, discussing mechanisms of action, pros and cons, and strategies to develop antitumor therapies based on the modulation of this axis.
Literature
11.
go back to reference Warburg O (1956) On respiratory impairment in cancer cells. Science 124:269–270PubMed Warburg O (1956) On respiratory impairment in cancer cells. Science 124:269–270PubMed
13.
go back to reference Elstrom RL, Bauer DE, Buzzai M et al (2004) Akt stimulates aerobic glycolysis in cancer cells. Cancer Res 64:3892–3899PubMedCrossRef Elstrom RL, Bauer DE, Buzzai M et al (2004) Akt stimulates aerobic glycolysis in cancer cells. Cancer Res 64:3892–3899PubMedCrossRef
15.
go back to reference Menendez JA, Lupu R (2007) Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer 7:763–777PubMedCrossRef Menendez JA, Lupu R (2007) Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer 7:763–777PubMedCrossRef
16.
go back to reference Rysman E, Brusselmans K, Scheys K et al (2010) De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res 70:8117–8126PubMedCrossRef Rysman E, Brusselmans K, Scheys K et al (2010) De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res 70:8117–8126PubMedCrossRef
18.
go back to reference Repa JJ, Mangelsdorf DJ (2000) The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol 16:459–481PubMedCrossRef Repa JJ, Mangelsdorf DJ (2000) The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol 16:459–481PubMedCrossRef
19.
go back to reference Chang CH, Qiu J, O’Sullivan D et al (2015) Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162:1229–1241PubMedCrossRef Chang CH, Qiu J, O’Sullivan D et al (2015) Metabolic competition in the tumor microenvironment is a driver of cancer progression. Cell 162:1229–1241PubMedCrossRef
20.
go back to reference Ho PC, Bihuniak JD, Macintyre AN et al (2015) Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 162:1217–1228PubMedCrossRef Ho PC, Bihuniak JD, Macintyre AN et al (2015) Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell 162:1217–1228PubMedCrossRef
21.
23.
go back to reference Cubillos-Ruiz JR, Silberman PC, Rutkowski MR et al (2015) ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161:1527–1538PubMedCrossRef Cubillos-Ruiz JR, Silberman PC, Rutkowski MR et al (2015) ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161:1527–1538PubMedCrossRef
24.
go back to reference Horton JD, Goldstein JL, Brown MS (2002) SREBPs: transcriptional mediators of lipid homeostasis. Cold Spring Harb Symp Quant Biol 67:491–498PubMedCrossRef Horton JD, Goldstein JL, Brown MS (2002) SREBPs: transcriptional mediators of lipid homeostasis. Cold Spring Harb Symp Quant Biol 67:491–498PubMedCrossRef
25.
go back to reference Horton JD, Shah NA, Warrington JA, Anderson NN, Park SW, Brown MS, Goldstein JL (2003) Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc Natl Acad Sci USA 100:12027–12032PubMedPubMedCentralCrossRef Horton JD, Shah NA, Warrington JA, Anderson NN, Park SW, Brown MS, Goldstein JL (2003) Combined analysis of oligonucleotide microarray data from transgenic and knockout mice identifies direct SREBP target genes. Proc Natl Acad Sci USA 100:12027–12032PubMedPubMedCentralCrossRef
26.
go back to reference Goldstein JL, Brown MS (2015) A century of cholesterol and coronaries: from plaques to genes to statins. Cell 161:161–172PubMedCrossRef Goldstein JL, Brown MS (2015) A century of cholesterol and coronaries: from plaques to genes to statins. Cell 161:161–172PubMedCrossRef
27.
go back to reference Bovenga F, Sabba C, Moschetta A (2015) Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer. Cell Metab 21:517–526PubMedCrossRef Bovenga F, Sabba C, Moschetta A (2015) Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer. Cell Metab 21:517–526PubMedCrossRef
30.
go back to reference Repa JJ, Mangelsdorf DJ (2002) The liver X receptor gene team: potential new players in atherosclerosis. Nat Med 8:1243–1248PubMedCrossRef Repa JJ, Mangelsdorf DJ (2002) The liver X receptor gene team: potential new players in atherosclerosis. Nat Med 8:1243–1248PubMedCrossRef
32.
go back to reference Janowski BA, Willy PJ, Devi TR, Falck JR, Mangelsdorf DJ (1996) An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha. Nature 383:728–731PubMedCrossRef Janowski BA, Willy PJ, Devi TR, Falck JR, Mangelsdorf DJ (1996) An oxysterol signalling pathway mediated by the nuclear receptor LXR alpha. Nature 383:728–731PubMedCrossRef
33.
go back to reference Bjorkhem I (2002) Do oxysterols control cholesterol homeostasis? J Clin Invesig. 110:725–730CrossRef Bjorkhem I (2002) Do oxysterols control cholesterol homeostasis? J Clin Invesig. 110:725–730CrossRef
35.
go back to reference Chen W, Chen G, Head DL, Mangelsdorf DJ, Russell DW (2007) Enzymatic reduction of oxysterols impairs LXR signaling in cultured cells and the livers of mice. Cell Metab 5:73–79PubMedPubMedCentralCrossRef Chen W, Chen G, Head DL, Mangelsdorf DJ, Russell DW (2007) Enzymatic reduction of oxysterols impairs LXR signaling in cultured cells and the livers of mice. Cell Metab 5:73–79PubMedPubMedCentralCrossRef
38.
go back to reference Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL (2007) Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: oxysterols block transport by binding to Insig. Proc Natl Acad Sci USA 104:6511–6518PubMedPubMedCentralCrossRef Radhakrishnan A, Ikeda Y, Kwon HJ, Brown MS, Goldstein JL (2007) Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: oxysterols block transport by binding to Insig. Proc Natl Acad Sci USA 104:6511–6518PubMedPubMedCentralCrossRef
39.
go back to reference Laffitte BA, Chao LC, Li J et al (2003) Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue. Proc Natl Acad Sci USA 100:5419–5424PubMedPubMedCentralCrossRef Laffitte BA, Chao LC, Li J et al (2003) Activation of liver X receptor improves glucose tolerance through coordinate regulation of glucose metabolism in liver and adipose tissue. Proc Natl Acad Sci USA 100:5419–5424PubMedPubMedCentralCrossRef
42.
go back to reference Fukuchi J, Kokontis JM, Hiipakka RA, Chuu CP, Liao S (2004) Antiproliferative effect of liver X receptor agonists on LNCaP human prostate cancer cells. Cancer Res 64:7686–7689PubMedCrossRef Fukuchi J, Kokontis JM, Hiipakka RA, Chuu CP, Liao S (2004) Antiproliferative effect of liver X receptor agonists on LNCaP human prostate cancer cells. Cancer Res 64:7686–7689PubMedCrossRef
49.
go back to reference Villablanca EJ, Raccosta L, Zhou D et al (2010) Tumor-mediated liver X receptor-alpha activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor responses. Nat Med 16:98–105. doi:10.1038/nm.2074 PubMedCrossRef Villablanca EJ, Raccosta L, Zhou D et al (2010) Tumor-mediated liver X receptor-alpha activation inhibits CC chemokine receptor-7 expression on dendritic cells and dampens antitumor responses. Nat Med 16:98–105. doi:10.​1038/​nm.​2074 PubMedCrossRef
50.
go back to reference Joseph SB, Castrillo A, Laffitte BA, Mangelsdorf DJ, Tontonoz P (2003) Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat Med 9:213–219PubMedCrossRef Joseph SB, Castrillo A, Laffitte BA, Mangelsdorf DJ, Tontonoz P (2003) Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat Med 9:213–219PubMedCrossRef
51.
go back to reference Bensinger SJ, Tontonoz P (2008) Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 454:470–477PubMedCrossRef Bensinger SJ, Tontonoz P (2008) Integration of metabolism and inflammation by lipid-activated nuclear receptors. Nature 454:470–477PubMedCrossRef
52.
go back to reference Joseph SB, Bradley MN, Castrillo A et al (2004) LXR-dependent gene expression is important for macrophage survival and the innate immune response. Cell 119:299–309PubMedCrossRef Joseph SB, Bradley MN, Castrillo A et al (2004) LXR-dependent gene expression is important for macrophage survival and the innate immune response. Cell 119:299–309PubMedCrossRef
53.
go back to reference Valledor AF, Hsu LC, Ogawa S, Sawka-Verhelle D, Karin M, Glass CK (2004) Activation of liver X receptors and retinoid X receptors prevents bacterial-induced macrophage apoptosis. Proc Natl Acad Sci USA 101:17813–17818PubMedPubMedCentralCrossRef Valledor AF, Hsu LC, Ogawa S, Sawka-Verhelle D, Karin M, Glass CK (2004) Activation of liver X receptors and retinoid X receptors prevents bacterial-induced macrophage apoptosis. Proc Natl Acad Sci USA 101:17813–17818PubMedPubMedCentralCrossRef
Metadata
Title
Cholesterol metabolites and tumor microenvironment: the road towards clinical translation
Authors
Laura Raccosta
Raffaella Fontana
Gianfranca Corna
Daniela Maggioni
Marta Moresco
Vincenzo Russo
Publication date
01-01-2016
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 1/2016
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-015-1779-0

Other articles of this Issue 1/2016

Cancer Immunology, Immunotherapy 1/2016 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine