USP7 inhibitors suppress tumour neoangiogenesis and promote synergy with immune checkpoint inhibitors by downregulating fibroblast VEGF

Anamarija Jurisic , Pei-Ju Sung , Mark Wappett , Julien Daubriac , Ian T. Lobb , Wei-Wei Kung , Nyree Crawford , Natalie Page , Eamon Cassidy , Stephanie Feutren-Burton , J. S. Shane Rountree , Matthew D. Helm , Colin R. O'Dowd , Richard D. Kennedy , Gerald Gavory , Aaron N. Cranston , Daniel B. Longley , Xavier Jacq , Timothy Harrison

Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (4) : e1648

PDF
Clinical and Translational Medicine ›› 2024, Vol. 14 ›› Issue (4) : e1648 DOI: 10.1002/ctm2.1648
RESEARCH ARTICLE

USP7 inhibitors suppress tumour neoangiogenesis and promote synergy with immune checkpoint inhibitors by downregulating fibroblast VEGF

Author information +
History +
PDF

Abstract

Background: Understanding how to modulate the microenvironment of tumors that are resistant to immune checkpoint inhibitors represents a major challenge in oncology.Here we investigate the ability of USP7 inhibitors to reprogram the tumor microenvironment (TME) by inhibiting secretion of vascular endothelial growth factor (VEGF) from fibroblasts.

Methods: To understand the role played by USP7 in the TME, we systematically evaluated the effects of potent, selective USP7 inhibitors on co-cultures comprising components of the TME, using human primary cells. We also evaluated the effects of USP7 inhibition on tumor growth inhibition in syngeneic models when dosed in combination with immune checkpoint inhibitors (ICIs).

Results: Abrogation of VEGF secretion from fibroblasts in response to USP7 inhibition resulted in inhibition of tumor neoangiogenesis and increased tumor recruitment of CD8-positive T-lymphocytes, leading to significantly improved sensitivity to immune checkpoint inhibitors. In syngeneic models, treatment with USP7 inhibitors led to striking tumor responses resulting in significantly improved survival.

Conclusions: USP7-mediated reprograming of the TME is not linked to its previously characterized role in modulating MDM2 but does require p53 and UHRF1 in addition to the well-characterized VEGF transcription factor, HIF-1α. This represents a function of USP7 that is unique to fibroblasts, and which is not observed in cancer cells or other components of the TME. Given the potential for USP7 inhibitors to transform “immune desert” tumors into “immune responsive” tumors, this paves the way for a novel therapeutic strategy combining USP7 inhibitors with immune checkpoint inhibitors (ICIs).

Keywords

ADC-159 / CAFs / deubiquitylating enzymes / DUBs / HAUSP / HIF-1α / immune checkpoint inhibitors / immune therapy / IO / neoangiogenesis / TME / tumour microenvironment / USP7 / VEGF

Cite this article

Download citation ▾
Anamarija Jurisic, Pei-Ju Sung, Mark Wappett, Julien Daubriac, Ian T. Lobb, Wei-Wei Kung, Nyree Crawford, Natalie Page, Eamon Cassidy, Stephanie Feutren-Burton, J. S. Shane Rountree, Matthew D. Helm, Colin R. O'Dowd, Richard D. Kennedy, Gerald Gavory, Aaron N. Cranston, Daniel B. Longley, Xavier Jacq, Timothy Harrison. USP7 inhibitors suppress tumour neoangiogenesis and promote synergy with immune checkpoint inhibitors by downregulating fibroblast VEGF. Clinical and Translational Medicine, 2024, 14(4): e1648 DOI:10.1002/ctm2.1648

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144(5):646-674.

[2]

Hanahan D, Coussens LM. Accessories to the crime: functions of cells recruited to the tumor microenvironment. Cancer Cell. 2012;21(3):309-322.

[3]

Junttila MR, de Sauvage FJ. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature. 2013;501(7467):346-354.

[4]

Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer: harnessing the T cell response. Nat Rev Immunol. 2012;12(4):269-281.

[5]

Prasad V, Kaestner V. Nivolumab and pembrolizumab: monoclonal antibodies against programmed cell death-1 (PD-1) that are interchangeable. Semin Oncol. 2017;44(2):132-135.

[6]

Sharma P, Hu-Lieskovan S, Wargo JA, Primary RibasA. Adaptive, and acquired resistance to cancer immunotherapy. Cell. 2017;168(4):707-723.

[7]

Pugh CW, Ratcliffe PJ. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat Med. 2003;9(6):677-684.

[8]

Kaelin WG. The von Hippel-Lindau protein, HIF hydroxylation, and oxygen sensing. Biochem Biophys Res Commun. 2005;338(1):627-638.

[9]

Semenza GL. Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 2003;3(10):721-732.

[10]

Wang GL, Jiang BH, Rue EA, Semenza GL. Hypoxia-inducible factor 1 is a basic-helix-loop-helix-PAS heterodimer regulated by cellular O2 tension. Proc Natl Acad Sci U S A. 1995;92(12):5510-5514.

[11]

Salceda S, Caro J. Hypoxia-inducible factor 1alpha (HIF-1alpha) protein is rapidly degraded by the ubiquitin-proteasome system under normoxic conditions. Its stabilization by hypoxia depends on redox-induced changes. J Biol Chem. 1997;272(36):22642-22647.

[12]

Maxwell PH, Wiesener MS, Chang GW, et al. The tumour suppressor protein VHL targets hypoxia-inducible factors for oxygen-dependent proteolysis. Nature. 1999;399(6733):271-275.

[13]

Forsythe JA, Jiang BH, Iyer NV, et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol Cell Biol. 1996;16(9):4604-4613.

[14]

Carmeliet P, Dor Y, Herbert JM, et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature. 1998;394(6692):485-490.

[15]

Moreira IS, Fernandes PA, Ramos MJ. Vascular endothelial growth factor (VEGF) inhibition—a critical review. Anticancer Agents Med Chem. 2007;7(2):223-245.

[16]

Poon RT, Fan ST, Wong J. Clinical implications of circulating angiogenic factors in cancer patients. J Clin Oncol. 2001;19(4):1207-1225.

[17]

Chen X, Song E. Turning foes to friends: targeting cancer-associated fibroblasts. Nat Rev Drug Discovery. 2019;18(2):99-115.

[18]

Harrigan JA, Jacq X, Martin NM, Jackson SP. Deubiquitylating enzymes and drug discovery: emerging opportunities. Nat Rev Drug Discovery. 2018;17(1):57-78.

[19]

Wertz IE, Murray JM. Structurally-defined deubiquitinase inhibitors provide opportunities to investigate disease mechanisms. Drug Discovery Today Technol. 2019;31:109-123.

[20]

Kategaya L, Di Lello P, Rouge L, et al. USP7 small-molecule inhibitors interfere with ubiquitin binding. Nature. 2017;550(7677):534-538.

[21]

Turnbull AP, Ioannidis S, Krajewski WW, et al. Molecular basis of USP7 inhibition by selective small-molecule inhibitors. Nature. 2017;550(7677):481-486.

[22]

Lamberto I, Liu X, Seo HS, et al. Structure-guided development of a potent and selective non-covalent active-site inhibitor of USP7. Cell Chem Biol. 2017;24(12):1490-1500.

[23]

O'Dowd CR, Helm MD, Rountree JSS, et al. Identification and structure-guided development of pyrimidinone based USP7 inhibitors. ACS Med Chem Lett. 2018;9(3):238-243.

[24]

Gavory G, O'Dowd CR, Helm MD, et al. Discovery and characterization of highly potent and selective allosteric USP7 inhibitors. Nat Chem Biol. 2018;14(2):118-125.

[25]

Tomasek JJ, Gabbiani G, Hinz B, Chaponnier C, Brown RA. Myofibroblasts and mechano-regulation of connective tissue remodelling. Nat Rev Mol Cell Biol. 2002;3(5):349-363.

[26]

de Jong A, Merkx R, Berlin I, et al. Ubiquitin-based probes prepared by total synthesis to profile the activity of deubiquitinating enzymes. Chembiochem. 2012;13(15):2251-2258.

[27]

Kallio PJ, Wilson WJ, O'Brien S, Makino Y, Poellinger L. Regulation of the hypoxia-inducible transcription factor 1alpha by the ubiquitin-proteasome pathway. J Biol Chem. 1999;274(10):6519-6525.

[28]

Mancini M, Magnani E, Macchi F, Bonapace IM. The multi-functionality of UHRF1: epigenome maintenance and preservation of genome integrity. Nucleic Acids Res. 2021;49(11):6053-6068.

[29]

Achour M, Jacq X, Ronde P, et al. The interaction of the SRA domain of ICBP90 with a novel domain of DNMT1 is involved in the regulation of VEGF gene expression. Oncogene. 2008;27(15):2187-2197.

[30]

Hurley JR, Balaji S, Narmoneva DA. Complex temporal regulation of capillary morphogenesis by fibroblasts. Am J Physiol Cell Physiol. 2010;299(2):C444-C453.

[31]

Kunz-Schughart LA, Schroeder JA, Wondrak M, et al. Potential of fibroblasts to regulate the formation of three-dimensional vessel-like structures from endothelial cells in vitro. Am J Physiol Cell Physiol. 2006;290(5):C1385-1398.

[32]

Berthod F, Germain L, Tremblay N, Auger FA. Extracellular matrix deposition by fibroblasts is necessary to promote capillary-like tube formation in vitro. J Cell Physiol. 2006;207(2):491-498.

[33]

Motz GT, Coukos G. Deciphering and reversing tumor immune suppression. Immunity. 2013;39(1):61-73.

[34]

Dirkx AE, Oude Egbrink MG, Kuijpers MJ, et al. Tumor angiogenesis modulates leukocyte-vessel wall interactions in vivo by reducing endothelial adhesion molecule expression. Cancer Res. 2003;63(9):2322-2329.

[35]

Guerreiro N, Jullion A, Ferretti S, Fabre C, Meille C. Translational modeling of anticancer efficacy to predict clinical outcomes in a first-in-human phase 1 study of MDM2 inhibitor HDM201. AAPS J. 2021;23(2):28.

[36]

Yang J, Yan J, Liu B. Targeting VEGF/VEGFR to modulate antitumor immunity. Front Immunol. 2018;9:978-987.

[37]

Ohm JE, Carbone DP. VEGF as a mediator of tumor-associated immunodeficiency. Immunol Res. 2001;23(2-3):263-272.

[38]

Newman AC, Nakatsu MN, Chou W, Gershon PD, Hughes CCW. The requirement for fibroblasts in angiogenesis: Fibroblast-derived matrix proteins are essential for endothelial cell lumen formation. Mol Biol Cell. 2011;22(20):3791-3800.

[39]

Ferrara N, Adamis AP. Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discovery. 2016;15(6):385-403.

[40]

Jain RK. Antiangiogenesis strategies revisited: from starving tumors to alleviating hypoxia. Cancer Cell. 2014;26(5):605-622.

[41]

Vasudev NS, Reynolds AR. Anti-angiogenic therapy for cancer: current progress, unresolved questions and future directions. Angiogenesis. 2014;17(3):471-494.

[42]

Shojaei F, Wu X, Malik AK, et al. Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+Gr1+ myeloid cells. Nat Biotechnol. 2007;25(8):911-920.

[43]

Rivera LB, Meyronet D, Hervieu V, Frederick MJ, Bergsland E, Bergers G. Intratumoral myeloid cells regulate responsiveness and resistance to antiangiogenic therapy. Cell Rep. 2015;11(4):577-591.

[44]

Rivera LB, Bergers GCANCER. Tumor angiogenesis, from foe to friend. Science. 2015;349(6249):694-695.

[45]

Bouzin C, Brouet A, De Vriese J, Dewever J, Feron O. Effects of vascular endothelial growth factor on the lymphocyte-endothelium interactions: identification of caveolin-1 and nitric oxide as control points of endothelial cell anergy. J Immunol. 2007;178(3):1505-1511.

[46]

Manegold C, Dingemans AC, Gray JE, et al. The potential of combined immunotherapy and antiangiogenesis for the synergistic treatment of advanced NSCLC. J Thorac Oncol. 2017;12(2):194-207.

[47]

Bagaev A, Kotlov N, Nomie K, et al. Conserved pan-cancer microenvironment subtypes predict response to immunotherapy. Cancer Cell. 2021;39(6):845-865.

[48]

van Loosdregt J, Fleskens V, Fu J, et al. Stabilization of the transcription factor Foxp3 by the deubiquitinase USP7 increases Treg-cell-suppressive capacity. Immunity. 2013;39(2):259-271.

[49]

Wang F, Wang L, Wu J, et al. Active site-targeted covalent irreversible inhibitors of USP7 impair the functions of Foxp3+ T-regulatory cells by promoting ubiquitination of Tip60. PLoS One. 2017;12(12):e0189744.

[50]

Wang L, Kumar S, Dahiya S, et al. Ubiquitin-specific Protease-7 inhibition impairs Tip60-dependent Foxp3+ T-regulatory cell function and promotes antitumor immunity. EBioMedicine. 2016;13:99-112.

[51]

Ritorto MS, Ewan R, Perez-Oliva AB, et al. Screening of DUB activity and specificity by MALDI-TOF mass spectrometry. Nat Commun. 2014;5:4763-4774.

[52]

Bushman JW, Donovan KA, Schauer NJ, et al. Proteomics-based identification of DUB substrates using selective inhibitors. Cell Chem Biol. 2020;28(1):78-87.

[53]

Wu HT, Kuo YC, Hung JJ, et al. K63-polyubiquitinated HAUSP deubiquitinates HIF-1alpha and dictates H3K56 acetylation promoting hypoxia-induced tumour progression. Nat Commun. 2016;7:13644-13661.

[54]

Lee JT, Gu W. The multiple levels of regulation by p53 ubiquitination. Cell Death Differ. 2010;17(1):86-92.

[55]

Cummins JM, Rago C, Kohli M, Kinzler KW, Lengauer C, Vogelstein B. Tumour suppression: disruption of HAUSP gene stabilizes p53. Nature. 2004;428(6982):1-2.

[56]

An WG, Kanekal M, Simon MC, Maltepe E, Blagosklonny MV, Neckers LM. Stabilization of wild-type p53 by hypoxia-inducible factor 1alpha. Nature. 1998;392(6674):405-408.

[57]

Chen D, Li M, Luo J, Gu W. Direct interactions between HIF-1 alpha and Mdm2 modulate p53 function. J Biol Chem. 2003;278(16):13595-13598.

[58]

Ravi R, Mookerjee B, Bhujwalla ZM, et al. Regulation of tumor angiogenesis by p53-induced degradation of hypoxia-inducible factor 1alpha. Genes Dev. 2000;14(1):34-44.

[59]

Nieminen AL, Qanungo S, Schneider EA, Jiang BH, Agani FH. Mdm2 and HIF-1alpha interaction in tumor cells during hypoxia. J Cell Physiol. 2005;204(2):364-369.

[60]

Farhang Ghahremani M, Goossens S, Nittner D, et al. p53 promotes VEGF expression and angiogenesis in the absence of an intact p21-Rb pathway. Cell Death Differ. 2013;20(7):888-897.

[61]

Bronner C, Alhosin M, Hamiche A, Mousli M. Coordinated dialogue between UHRF1 and DNMT1 to ensure faithful inheritance of methylated DNA patterns. Genes (Basel). 2019;10(1):65-79.

[62]

Su D, Wang W, Hou Y, et al. Bimodal regulation of the PRC2 complex by USP7 underlies tumorigenesis. Nucleic Acids Res. 2021;49(8):4421-4440.

[63]

Felle M, Joppien S, Nemeth A, et al. The USP7/Dnmt1 complex stimulates the DNA methylation activity of Dnmt1 and regulates the stability of UHRF1. Nucleic Acids Res. 2011;39(19):8355-8365.

[64]

Dobin A, Davis CA, Schlesinger F, et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics. 2013;29(1):15-21.

[65]

Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33(3):290-295.

[66]

Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014;15(12):550-571.

[67]

Ritchie ME, Phipson B, Wu D, et al. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43(7):e47-60.

[68]

Kuleshov MV, Jones MR, Rouillard AD, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90-97.

RIGHTS & PERMISSIONS

2024 Almac Discovery Ltd. Clinical and Translational Medicine published by John Wiley & Sons Australia, Ltd on behalf of Shanghai Institute of Clinical Bioinformatics.

AI Summary AI Mindmap
PDF

173

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/