650
Views
10
CrossRef citations to date
0
Altmetric
Review

Rho GTPase effectors and NAD metabolism in cancer immune suppression

, , , , , & show all
Pages 9-17 | Received 11 Oct 2017, Accepted 30 Nov 2017, Published online: 10 Dec 2017

References

  • Lv J, Wang J, Chang S, et al. The greedy nature of mutant RAS: a boon for drug discovery targeting cancer metabolism? Acta Biochim Biophys Sin (Shanghai). 2016;48(1):17–26.
  • Tufail S, Badrealam KF, Sherwani A, et al. Tissue specific heterogeneity in effector immune cell response. Front Immunol. 2013;4:254.
  • Kato S, Goodman A, Walavalkar V, et al. Hyperprogressors after immunotherapy: analysis of genomic alterations associated with accelerated growth rate. Clin Cancer Res. 2017;23(15):4242–4250.
  • Potter M, Newport E, Morten KJ. The Warburg effect: 80 years on. Biochem Soc Trans. 2016;44:1499–1505.
  • Jansen S, Gosens R, Wieland T, et al. Paving the Rho in cancer metastasis: Rho GTPases and beyond. Pharmacol Ther. 2017. S0163-7258(17)30236-X.
  • Gillies RJ, Gatenby RA. Metabolism and its sequelae in cancer evolution and therapy. Cancer J. 2015;21:88–96.
  • Liberti MV, Locasale JW. The Warburg effect: how does it benefit cancer cells? Trends Biochem Sci. 2016;41:211–218.
  • Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–1033.
  • Imai S. Nicotinamide phosphoribosyltransferase (Nampt): a link between NAD biology, metabolism, and diseases. Curr Pharm Des. 2009;15:20–28.
  • Grahnert A, Grahnert A, Klein C, et al. Review: NAD +: a modulator of immune functions. Innate Immun. 2011;17:212–233.
  • Joyce JA, Fearon DT. T cell exclusion, immune privilege, and the tumor microenvironment. Science. 2015;348:74–80.
  • Skelton RA, Javed A, Zheng L, et al. Overcoming the resistance of pancreatic cancer to immune checkpoint inhibitors. J Surg Oncol. 2017;116:55–62.
  • Scotton CJ, Wilson JL, Scott K, et al. Multiple actions of the chemokine CXCL12 on epithelial tumor cells in human ovarian cancer. Cancer Res. 2002;62:5930–5938.
  • Liang JJ, Zhu S, Bruggeman R, et al. High levels of expression of human stromal cell-derived factor-1 are associated with worse prognosis in patients with stage II pancreatic ductal adenocarcinoma. Cancer Epidemiol Biomarkers Prev. 2010;19:2598–2604.
  • Akishima-Fukasawa Y, Nakanishi Y, Ino Y, et al. Prognostic significance of CXCL12 expression in patients with colorectal carcinoma. Am J Clin Pathol. 2009;132:202–210. quiz 307.
  • Feig C, Jones JO, Kraman M, et al. Targeting CXCL12 from FAP-expressing carcinoma-associated fibroblasts synergizes with anti-PD-L1 immunotherapy in pancreatic cancer. Proc Natl Acad Sci U S A. 2013;110:20212–20217.
  • Sukari A, Nagasaka M, Al-Hadidi A, et al. Cancer immunology and immunotherapy. Anticancer Res. 2016;36:5593–5606.
  • Bengsch F, Knoblock DM, Liu A, et al. CTLA-4/CD80 pathway regulates T cell infiltration into pancreatic cancer. Cancer Immunol Immunother. 2017.
  • Arasanz H, Gato-Canas M, Zuazo M, et al. PD1 signal transduction pathways in T cells. Oncotarget. 2017;8:51936–51945.
  • Alsaab HO, Sau S, Alzhrani R, et al. PD-1 and PD-L1 checkpoint signaling inhibition for cancer immunotherapy: mechanism, combinations, and clinical outcome. Front Pharmacol. 2017;8:561.
  • Ji RR, Chasalow SD, Wang L, et al. An immune-active tumor microenvironment favors clinical response to ipilimumab. Cancer Immunol Immunother. 2012;61:1019–1031.
  • Harlin H, Meng Y, Peterson AC, et al. Chemokine expression in melanoma metastases associated with CD8+ T-cell recruitment. Cancer Res. 2009;69:3077–3085.
  • Morgan RG, Ridsdale J, Tonks A, et al. Factors affecting the nuclear localization of beta-catenin in normal and malignant tissue. J Cell Biochem. 2014;115:1351–1361.
  • Pai SG, Carneiro BA, Mota JM, et al. Wnt/beta-catenin pathway: modulating anticancer immune response. J Hematol Oncol. 2017;10:101.
  • Jin R, Liu W, Menezes S, et al. The metastasis suppressor NDRG1 modulates the phosphorylation and nuclear translocation of beta-catenin through mechanisms involving FRAT1 and PAK4. J Cell Sci. 2014;127:3116–3130.
  • Li Y, Shao Y, Tong Y, et al. Nucleo-cytoplasmic shuttling of PAK4 modulates beta-catenin intracellular translocation and signaling. Biochim Biophys Acta. 2012;1823:465–475.
  • Szczepaniak Sloane RA, Gopalakrishnan V, Reddy SM, et al. Interaction of molecular alterations with immune response in melanoma. Cancer. 2017;123:2130–2142.
  • Spranger S, Bao R, Gajewski TF. Melanoma-intrinsic beta-catenin signalling prevents anti-tumour immunity. Nature. 2015;523:231–235.
  • Spranger S, Gajewski TF. A new paradigm for tumor immune escape: beta-catenin-driven immune exclusion. J Immunother Cancer. 2015;3:43.
  • Spranger S, Gajewski TF. Tumor-intrinsic oncogene pathways mediating immune avoidance. Oncoimmunology. 2016;5:e1086862.
  • Gajewski TF, Louahed J, Brichard VG. Gene signature in melanoma associated with clinical activity: a potential clue to unlock cancer immunotherapy. Cancer J. 2010;16:399–403.
  • Tumeh PC, Harview CL, Yearley JH, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature. 2014;515:568–571.
  • Sweis RF, Spranger S, Bao R, et al. Molecular drivers of the non-T-cell-inflamed tumor microenvironment in urothelial bladder cancer. Cancer Immunol Res. 2016;4:563–568.
  • Rajagopalan A, Berezhnoy A, Schrand B, et al. Aptamer-targeted attenuation of IL-2 signaling in CD8+ T cells enhances antitumor immunity. Mol Ther. 2017;25:54–61.
  • Luke JJ, Riyue B, Spranger S, et al. Correlation of WNT/beta-catenin pathway activation with immune exclusion across most human cancers. Am Soc Clin Oncol (ASCO). 2016;Abstract 3004.
  • Yang H, Chen H, Luo S, et al. The correlation between programmed death-ligand 1 expression and driver gene mutations in NSCLC. Oncotarget. 2017;8:23517–23528.
  • Li D, Zhu X, Wang H, et al. Association between PD-L1 expression and driven gene status in NSCLC: a meta-analysis. Eur J Surg Oncol. 2017;43:1372–1379.
  • Kim S, Koh J, Kwon D, et al. Comparative analysis of PD-L1 expression between primary and metastatic pulmonary adenocarcinomas. Eur J Cancer. 2017;75:141–149.
  • Loi S, Dushyanthen S, Beavis PA, et al. RAS/MAPK activation is associated with reduced tumor-infiltrating lymphocytes in triple-negative breast cancer: therapeutic cooperation between MEK and PD-1/PD-L1 immune checkpoint inhibitors. Clin Cancer Res. 2016;22:1499–1509.
  • Sumimoto H, Takano A, Teramoto K, et al. RAS-mitogen-activated protein kinase signal is required for enhanced PD-L1 expression in human lung cancers. PLoS One. 2016;11:e0166626.
  • Henning S, Cleverley S. Small GTPases in lymphocyte biology: Rho proteins take center stage. Immunol Res. 1999;20:29–42.
  • Radu M, Semenova G, Kosoff R, et al. PAK signalling during development and progression of cancer. Nat Rev. 2014;14:13–25.
  • Laudanna C, Campbell JJ, Butcher EC. Role of Rho in chemoattractant-activated leukocyte adhesion through integrins. Science. 1996;271:981–983.
  • D’Souza-Schorey C, Boettner B, Van Aelst L. Rac regulates integrin-mediated spreading and increased adhesion of T lymphocytes. Mol Cell Biol. 1998;18:3936–3946.
  • Vu HL, Rosenbaum S, Purwin TJ, et al. RAC1 P29S regulates PD-L1 expression in melanoma. Pigment Cell Melanoma Res. 2015;28:590–598.
  • Marques CA, Hahnel PS, Wolfel C, et al. An immune escape screen reveals Cdc42 as regulator of cancer susceptibility to lymphocyte-mediated tumor suppression. Blood. 2008;111:1413–1419.
  • Selamat W, Tay PL, Baskaran Y, et al. The Cdc42 effector kinase PAK4 localizes to cell-cell junctions and contributes to establishing cell polarity. PLoS One. 2015;10:e0129634.
  • Scharping NE, Delgoffe GM. Tumor microenvironment metabolism: a new checkpoint for anti-tumor immunity. Vaccines (Basel). 2016;4(4):E46.
  • Ho PC, Bihuniak JD, Macintyre AN, et al. Phosphoenolpyruvate is a metabolic checkpoint of anti-tumor T cell responses. Cell. 2015;162:1217–1228.
  • Kouidhi S, Elgaaied AB, Chouaib S. Impact of metabolism on T-Cell differentiation and function and cross talk with tumor microenvironment. Front Immunol. 2017;8:270.
  • Beckermann KE, Dudzinski SO, Rathmell JC. Dysfunctional T cell metabolism in the tumor microenvironment. Cytokine Growth Factor Rev. 2017;35:7–14.
  • Renner K, Singer K, Koehl GE, et al. Metabolic hallmarks of tumor and immune cells in the tumor microenvironment. Front Immunol. 2017;8:248.
  • Goffaux G, Hammami I, Jolicoeur M. A dynamic metabolic flux analysis of myeloid-derived suppressor cells confirms immunosuppression-related metabolic plasticity. Sci Rep. 2017;7:9850.
  • Chini CCS, Tarrago MG, Chini EN. NAD and the aging process: role in life, death and everything in between. Mol Cell Endocrinol. 2017;455:62–74.
  • Wang T, Zhang X, Bheda P, et al. Structure of Nampt/PBEF/visfatin, a mammalian NAD+ biosynthetic enzyme. Nat Struct Mol Biol. 2006;13:661–662.
  • Chini CC, Guerrico AM, Nin V, et al. Targeting of NAD metabolism in pancreatic cancer cells: potential novel therapy for pancreatic tumors. Clin Cancer Res. 2014;20:120–130.
  • Li Y, Zhang Y, Dorweiler B, et al. Extracellular Nampt promotes macrophage survival via a nonenzymatic interleukin-6/STAT3 signaling mechanism. J Biol Chem. 2008;283:34833–34843.
  • Kang TS, Korber DR, Tanaka T. Glycerol and environmental factors: effects on 1,3-propanediol production and NAD(+) regeneration in Lactobacillus panis PM1. J Appl Microbiol. 2013;115:1003–1011.
  • Busso N, Karababa M, Nobile M, et al. Pharmacological inhibition of nicotinamide phosphoribosyltransferase/visfatin enzymatic activity identifies a new inflammatory pathway linked to NAD. PLoS One. 2008;3:e2267.
  • Audrito V, Serra S, Brusa D, et al. Extracellular nicotinamide phosphoribosyltransferase (NAMPT) promotes M2 macrophage polarization in chronic lymphocytic leukemia. Blood. 2015;125:111–123.
  • Rongvaux A, Galli M, Denanglaire S, et al. Nicotinamide phosphoribosyl transferase/pre-B cell colony-enhancing factor/visfatin is required for lymphocyte development and cellular resistance to genotoxic stress. J Immunol. 2008;181:4685–4695.
  • Roberts KJ, Cross A, Vasieva O, et al. Inhibition of pre-B cell colony-enhancing factor (PBEF/NAMPT/visfatin) decreases the ability of human neutrophils to generate reactive oxidants but does not impair bacterial killing. J Leukoc Biol. 2013;94:481–492.
  • Naing A, Leong S, Pishvaian MJ, et al. 374PD - a first in human phase 1 study of KPT-9274, a first in class dual inhibitor of PAK4 and NAMPT, in patients with advanced solid malignancies or NHL. Ann Oncol. 2017;28.
  • Murray BW, Guo C, Piraino J, et al. Small-molecule p21-activated kinase inhibitor PF-3758309 is a potent inhibitor of oncogenic signaling and tumor growth. Proc Natl Acad Sci U S A. 2010;107:9446–9451.
  • Hasmann M, Schemainda I. FK866, a highly specific noncompetitive inhibitor of nicotinamide phosphoribosyltransferase, represents a novel mechanism for induction of tumor cell apoptosis. Cancer Res. 2003;63:7436–7442.
  • Rosen LS, Blumenkopf TA, Breazna A, et al. Phase 1, dose-escalateion, safety, pharmacokinetic and pharmacodynamic study of single agent PF-03758309, an oral PAK inhibitor, in patients with advanced solid tumors. AACR-NCI-EORTC International Conference: Molecular Targets and Cancer Therapeutics. 2011;10:ABSTRACT A177
  • Crawford JJ, Hoeflich KP, Rudolph J. p21-Activated kinase inhibitors: a patent review. Expert Opin Ther Pat. 2012;22:293–310.
  • Zhao ZS, Manser E. Do PAKs make good drug targets? F1000 Biol Rep. 2010;2:70.
  • Abu Aboud O, Chen CH, Senapedis W, et al. Dual and specific inhibition of NAMPT and PAK4 by KPT-9274 decreases kidney cancer growth. Mol Cancer Ther. 2016;15:2119–2129.
  • Aboukameel A, Muqbil I, Senapedis W, et al. Novel p21-activated kinase 4 (PAK4) allosteric modulators overcome drug resistance and stemness in pancreatic ductal adenocarcinoma. Mol Cancer Ther. 2017;16:76–87.
  • Rane C, Senapedis W, Baloglu E, et al. A novel orally bioavailable compound KPT-9274 inhibits PAK4, and blocks triple negative breast cancer tumor growth. Sci Rep. 2017;7:42555.
  • Fulciniti M, Martinez-Lopez J, Senapedis W, et al. Functional role and therapeutic targeting of p21-associated kinase 4 (PAK4) in multiple myeloma. Blood. 2017.
  • Jiang YY, Lin DC, Mayakonda A, et al. Targeting super-enhancer-associated oncogenes in oesophageal squamous cell carcinoma. Gut. 2016;66(8):1358–1368.
  • Azmi AS, Aboukameel A, Muqbil I, et al. Abstract 1358: p21 activated kinase 4 (pak4) as a novel therapeutic target for non-hodgkin’s lymphoma. Cancer Res. 2017;77.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.