uPAR antibody (huATN-658) and Zometa reduce breast cancer growth and skeletal lesions

Niaz Mahmood , Ani Arakelian , Haseeb Ahmed Khan , Imrana Tanvir , Andrew P. Mazar , Shafaat A. Rabbani

Bone Research ›› 2020, Vol. 8 ›› Issue (1) : 18

PDF
Bone Research ›› 2020, Vol. 8 ›› Issue (1) : 18 DOI: 10.1038/s41413-020-0094-3
Article

uPAR antibody (huATN-658) and Zometa reduce breast cancer growth and skeletal lesions

Author information +
History +
PDF

Abstract

Urokinase plasminogen activator receptor (uPAR) is implicated in tumor growth and metastasis due to its ability to activate latent growth factors, proteases, and different oncogenic signaling pathways upon binding to different ligands. Elevated uPAR expression is correlated with the increased aggressiveness of cancer cells, which led to its credentialing as an attractive diagnostic and therapeutic target in advanced solid cancer. Here, we examine the antitumor effects of a humanized anti-uPAR antibody (huATN-658) alone and in combination with the approved bisphosphonate Zometa (Zoledronic acid) on skeletal lesion through a series of studies in vitro and in vivo. Treatment with huATN-658 or Zometa alone significantly decreased human MDA-MB-231 cell proliferation and invasion in vitro, effects which were more pronounced when huATN-658 was combined with Zometa. In vivo studies demonstrated that huATN-658 treatment significantly reduced MDA-MB-231 primary tumor growth compared with controls. In a model of breast tumor-induced bone disease, huATN-658 and Zometa were equally effective in reducing skeletal lesions. The skeletal lesions were significantly reduced in animals receiving the combination of huATN-658 + Zometa compared with monotherapy treatment. These effects were due to a significant decrease in osteoclastic activity and tumor cell proliferation in the combination treatment group. Transcriptome analysis revealed that combination treatment significantly changes the expression of genes from signaling pathways implicated in tumor progression and bone remodeling. Results from these studies provide a rationale for the continued development of huATN-658 as a monotherapy and in combination with currently approved agents such as Zometa in patients with metastatic breast cancer.

Cite this article

Download citation ▾
Niaz Mahmood, Ani Arakelian, Haseeb Ahmed Khan, Imrana Tanvir, Andrew P. Mazar, Shafaat A. Rabbani. uPAR antibody (huATN-658) and Zometa reduce breast cancer growth and skeletal lesions. Bone Research, 2020, 8(1): 18 DOI:10.1038/s41413-020-0094-3

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Redig AJ, McAllister SS. Breast cancer as a systemic disease: a view of metastasis. J. Intern. Med., 2013, 274:113-126

[2]

Chung CT, Carlson RW. Goals and objectives in the management of metastatic breast cancer. Oncologist, 2003, 8:514-520

[3]

Bernard-Marty C, Cardoso F, Piccart MJ. Facts and controversies in systemic treatment of metastatic breast cancer. ncologist, 2004, 9:617-632

[4]

Roché H, Vahdat LT. Treatment of metastatic breast cancer: second line and beyond. Ann. Oncol., 2010, 22:1000-1010

[5]

Coleman RE, Rubens RD. The clinical course of bone metastases from breast cancer. Br. J. Cancer, 1987, 55:61-66

[6]

Coleman RE. Skeletal complications of malignancy. Cancer, 1997, 80:1588-1594

[7]

Coleman RE. Metastatic bone disease: clinical features, pathophysiology and treatment strategies. Cancer Treat. Rev., 2001, 27:165-176

[8]

Mahmood N, Mihalcioiu C, Rabbani SA. Multifaceted role of the urokinase-type plasminogen activator (uPA) and its receptor (uPAR): diagnostic, prognostic, and therapeutic applications. Front Oncol, 2018, 8:24

[9]

Degryse B et al. Src-dependence and pertussis-toxin sensitivity of urokinase receptor-dependent chemotaxis and cytoskeleton reorganization in rat smooth muscle cells. Blood, 1999, 94:649-662

[10]

Nicholl SM, Roztocil E, Davies MG. Urokinase-induced smooth muscle cell responses require distinct signaling pathways: a role for the epidermal growth factor receptor. J. Vasc. Surg., 2005, 41:672-681

[11]

Kobayashi H et al. Inhibition of metastasis of Lewis lung carcinoma by a synthetic peptide within growth factor-like domain of urokinase in the experimental and spontaneous metastasis model. Int J. Cancer, 1994, 57:727-733

[12]

Bürgle M et al. Inhibition of the interaction of urokinase-type plasminogen activator (uPA) with its receptor (uPAR) by synthetic peptides. Biol. Chem., 1997, 378:231-238

[13]

Crowley CW et al. Prevention of metastasis by inhibition of the urokinase receptor. Proc. Natl. Acad. Sci., 1993, 90:5021-5025

[14]

Tang C-H, Hill ML, Brumwell AN, Chapman HA, Wei Y. Signaling through urokinase and urokinase receptor in lung cancer cells requires interactions with β1 integrins. J. Cell Sci., 2008, 121:3747-3756

[15]

Okumura Y et al. Kinetic analysis of the interaction between vitronectin and the urokinase receptor. J. Biol. Chem., 2002, 277:9395-9404

[16]

Smith HW, Marshall CJ. Regulation of cell signalling by uPAR. Nat. Rev. Mol. Cell Biol., 2010, 11:23-36

[17]

Margheri F et al. Effects of blocking urokinase receptor signaling by antisense oligonucleotides in a mouse model of experimental prostate cancer bone metastases. Gene Ther., 2005, 12:702

[18]

Gondi CS et al. RNAi-mediated inhibition of cathepsin B and uPAR leads to decreased cell invasion, angiogenesis and tumor growth in gliomas. Oncogene, 2004, 23:8486-8496

[19]

Wang K et al. Targeting uPAR by CRISPR/Cas9 system attenuates cancer malignancy and multidrug resistance. Front Oncol., 2019, 9:80

[20]

Rysenkova KD et al. CRISPR/Cas9 nickase mediated targeting of urokinase receptor gene inhibits neuroblastoma cell proliferation. Oncotarget, 2018, 9:29414-29430

[21]

P Mazar A, W Ahn R, V O’Halloran T. Development of novel therapeutics targeting the urokinase plasminogen activator receptor (uPAR) and their translation toward the clinic. Curr. Pharm. Des., 2011, 17:1970-1978

[22]

Rabbani SA et al. An anti-urokinase plasminogen activator receptor antibody (ATN-658) blocks prostate cancer invasion, migration, growth, and experimental skeletal metastasis in vitro and in vivo. Neoplasia, 2010, 12:778-788

[23]

Kenny HA et al. Targeting the urokinase plasminogen activator receptor inhibits ovarian cancer metastasis. Clin. Cancer Res., 2011, 17:459-471

[24]

Xu X et al. Identification of a new epitope in uPAR as a target for the cancer therapeutic monoclonal antibody ATN-658, a structural homolog of the uPAR binding integrin CD11b (αM). PLoS ONE, 2014, 9

[25]

Pierga JY et al. Real-time quantitative PCR determination of urokinase-type plasminogen activator receptor (uPAR) expression of isolated micrometastatic cells from bone marrow of breast cancer patients. Int J. Cancer, 2005, 114:291-298

[26]

Hildenbrand R, Wolf G, Böhme B, Bleyl U, Steinborn A. Urokinase plasminogen activator receptor (CD87) expression of tumor‐associated macrophages in ductal carcinoma in situ, breast cancer, and resident macrophages of normal breast tissue. J. Leukoc. Biol., 1999, 66:40-49

[27]

Kalinichenko SV et al. Pdcd4 protein and mRNA level alterations do not correlate in human lung tumors. Lung Cancer, 2008, 62:173-180

[28]

Mahmood N et al. Methyl donor S-adenosylmethionine (SAM) supplementation attenuates breast cancer growth, invasion, and metastasis in vivo; therapeutic and chemopreventive applications. Oncotarget, 2018, 9:5169-5183

[29]

Ory B et al. Zoledronic acid activates the DNA S-phase checkpoint and induces osteosarcoma cell death characterized by apoptosis-inducing factor and endonuclease-G translocation independently of p53 and retinoblastoma status. Mol. Pharmacol., 2007, 71:333-343

[30]

Nienhuis HH, Arjaans M, Timmer-Bosscha H, de Vries EGE, Schröder CP. Human stromal cells are required for an anti-breast cancer effect of zoledronic acid. Oncotarget, 2015, 6:24436-24447

[31]

Aapro M, Saad F, Costa L. Optimizing clinical benefits of bisphosphonates in cancer patients with bone metastases. Oncologist, 2010, 15:1147-1158

[32]

Wan Q et al. BioXpress: an integrated RNA-seq-derived gene expression database for pan-cancer analysis. Database, 2015, 2015:bav019

[33]

Coleman RE. Management of bone metastases. Oncologist, 2000, 5:463-470

[34]

De Felice F, Piccioli A, Musio D, Tombolini V. The role of radiation therapy in bone metastases management. Oncotarget, 2017, 8:25691-25699

[35]

Clemons M, Gelmon KA, Pritchard KI, Paterson AHG. Bone-targeted agents and skeletal-related events in breast cancer patients with bone metastases: the state of the art. Curr. Oncol., 2012, 19:259-268

[36]

Previdi S et al. Combination of the c-Met inhibitor tivantinib and zoledronic acid prevents tumor bone engraftment and inhibits progression of established bone metastases in a breast xenograft model. PLoS ONE, 2013, 8

[37]

Huang J-M et al. Ulinastatin inhibits osteoclastogenesis and suppresses ovariectomy-induced bone loss by downregulating uPAR. Front. Pharmacol., 2018, 9:1016-1016

[38]

Kimmel DB. Mechanism of action, pharmacokinetic and pharmacodynamic profile, and clinical applications of nitrogen-containing bisphosphonates. J. Dent. Res., 2007, 86:1022-1033

[39]

Harvey HA. Issues concerning the role of chemotherapy and hormonal therapy of bone metastases from breast carcinoma. Cancer, 1997, 80:1646-1651

[40]

Wright LE et al. Murine models of breast cancer bone metastasis. Bonekey Rep., 2016, 5:804-804

[41]

Rosset EM, Bradshaw AD. SPARC/osteonectin in mineralized tissue. Matrix Biol., 2016, 52:78-87

[42]

Van Buren G 2nd et al. Targeting the urokinase plasminogen activator receptor with a monoclonal antibody impairs the growth of human colorectal cancer in the liver. Cancer, 2009, 115:3360-3368

[43]

Tang Z et al. GEPIA: a web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res., 2017, 45:W98-W102

[44]

Rhodes DR et al. ONCOMINE: a cancer microarray database and integrated data-mining platform. Neoplasia, 2004, 6:1-6

[45]

Chandrashekar DS et al. UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses. Neoplasia, 2017, 19:649-658

[46]

Györffy B et al. An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients. Breast Cancer Res. Treat., 2010, 123:725-731

[47]

Neve RM et al. A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell, 2006, 10:515-527

[48]

Ringnér M, Fredlund E, Häkkinen J, Borg Å, Staaf J. GOBO: gene expression-based outcome for breast cancer online. PLoS ONE, 2011, 6

[49]

Prichard MN, Shipman Jr C. A three-dimensional model to analyze drug-drug interactions. Antivir. Res., 1990, 14:181-205

[50]

Soica C et al. The synergistic biologic activity of oleanolic and ursolic acids in complex with hydroxypropyl-γ-cyclodextrin. Molecules, 2014, 19:4924-4940

[51]

Tuominen VJ, Ruotoistenmäki S, Viitanen A, Jumppanen M, Isola J. ImmunoRatio: a publicly available web application for quantitative image analysis of estrogen receptor (ER), progesterone receptor (PR), and Ki-67. Breast Cancer Res., 2010, 12:R56-R56

[52]

Dobin A et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics, 2013, 29:15-21

[53]

Trapnell C et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc., 2012, 7:562-578

[54]

Kamburov A, Stelzl U, Lehrach H, Herwig R. The ConsensusPathDB interaction database: 2013 update. Nucleic Acids Res., 2013, 41:D793-D800

[55]

Parashar S et al. S‐adenosylmethionine blocks osteosarcoma cells proliferation and invasion in vitro and tumor metastasis in vivo: therapeutic and diagnostic clinical applications. Cancer Med., 2015, 4:732-744

Funding

Gouvernement du Canada | Canadian Institutes of Health Research (Instituts de Recherche en Santé du Canada)(PJT-156225)

AI Summary AI Mindmap
PDF

139

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/