Anti-PAI-1 Monoclonal Antibody Inhibits the Metastasis and Growth of Esophageal Squamous Cell Carcinoma

Plasminogen activator inhibitor (PAI-1) is highly expressed in esophageal squamous cell carcinoma (ESCC) and strongly contributes to metastasis, making it a potential target for ESCC therapy. However, the antibodies and inhibitors targeting PAI-1 have not shown good therapeutic effect in the in vivo experiments yet. Here, we generated a panel of novel monoclonal antibodies (mAbs) against PAI-1. Analysis of PAI-1 expression in 90 tissue specimens and 128 serum specimens from ESCC patients with these mAbs confirmed that PAI-1 levels was significantly correlated with metastasis and poor survival. In addition, we found that high PAI-1 expression contributed to the enhanced motility and invasiveness of two ESCC cell lines. Next, mAb-1E2 and mAb-2E3, which have highest affinity with PAI-1, were shown to possess strong inhibitory effects on ESCC migration and invasion. Anti-tumor and anti-metastatic effects of mAb-2E3 were further demonstrated in the experimental animal models. Finally, LRP1 was identified as key factor mediating the pro-invasive function of PAI-1 and the anti-invasive capacity of mAb-2E3 in ESCC cells. The mAb-2E3 markedly decreased STAT1 phosphorylation levels and blocked the binding between PAI-1 and LRP1-ClusterII domain. Collectively, mAb-2E3 developed by our lab may be an effective antibody drug which can be used for anti-metastatic therapy in ESCC.

[1]  Furong Huang,et al.  Dysregulated ceramides metabolism by fatty acid 2-hydroxylase exposes a metabolic vulnerability to target cancer metastasis , 2022, Signal Transduction and Targeted Therapy.

[2]  A. Laurenzana,et al.  A Possible Role for PAI-1 Blockade in Melanoma Immunotherapy. , 2021, The Journal of investigative dermatology.

[3]  G. Wang,et al.  The molecular mechanism of LRP1 in physiological vascular homeostasis and signal transduction pathways. , 2021, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[4]  W. Weichert,et al.  uPA‐PAI‐1 heteromerization promotes breast cancer progression by attracting tumorigenic neutrophils , 2021, EMBO molecular medicine.

[5]  Quanren Wang,et al.  Safety and Feasibility of Radiotherapy Plus Camrelizumab for Locally Advanced Esophageal Squamous Cell Carcinoma , 2021, The oncologist.

[6]  D. Waisman,et al.  Plasmin and Plasminogen System in the Tumor Microenvironment: Implications for Cancer Diagnosis, Prognosis, and Therapy , 2021, Cancers.

[7]  A. Cervantes,et al.  Checkpoint inhibitors for gastroesophageal cancers: Dissecting heterogeneity to better understand their role in first line and adjuvant therapy. , 2021, Annals of oncology : official journal of the European Society for Medical Oncology.

[8]  A. Jemal,et al.  Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries , 2021, CA: a cancer journal for clinicians.

[9]  T. Miyata,et al.  Structural Insight into the Two-Step Mechanism of PAI-1 Inhibition by Small Molecule TM5484 , 2021, International journal of molecular sciences.

[10]  S. Lavrentiadou,et al.  Oleuropein is a natural inhibitor of PAI-1-mediated proliferation in human ER-/PR- breast cancer cells , 2021, Breast Cancer Research and Treatment.

[11]  Y. Kakeji,et al.  PAI-1 derived from cancer-associated fibroblasts in esophageal squamous cell carcinoma promotes the invasion of cancer cells and the migration of macrophages , 2020, Laboratory Investigation.

[12]  Jian-ping Xu,et al.  Phase II clinical trial using camrelizumab combined with apatinib and chemotherapy as the first‐line treatment of advanced esophageal squamous cell carcinoma , 2020, Cancer communications.

[13]  T. Huang,et al.  PAI-1-Dependent Inactivation of SMAD4-Modulated Junction and Adhesion Complex in Obese Endometrial Cancer , 2020, Cell reports.

[14]  Sung-Bae Kim,et al.  Randomized Phase III KEYNOTE-181 Study of Pembrolizumab Versus Chemotherapy in Advanced Esophageal Cancer. , 2020, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[15]  Qing‐Yu He,et al.  Advances in targeted therapy for esophageal cancer , 2020, Signal Transduction and Targeted Therapy.

[16]  M. Yoshino,et al.  AS3288802, a highly selective antibody to active plasminogen activator inhibitor-1 (PAI-1), exhibits long efficacy duration in cynomolgus monkeys. , 2020, Biologicals : journal of the International Association of Biological Standardization.

[17]  N. Kim,et al.  Association between Five Common Plasminogen Activator Inhibitor-1 (PAI-1) Gene Polymorphisms and Colorectal Cancer Susceptibility , 2020, International journal of molecular sciences.

[18]  Mingdong Huang,et al.  Plasminogen activator inhibitor (PAI) trap3, an exocellular peptide inhibitor of PAI-1, attenuates the rearrangement of F-actin and migration of cancer cells. , 2020, Experimental cell research.

[19]  Min Zhang,et al.  PAI-1 overexpression promotes invasion and migration of esophageal squamous carcinoma cells. , 2020, Yi chuan = Hereditas.

[20]  A. Fatehi Hassanabad,et al.  Esophageal carcinoma: Towards targeted therapies , 2019, Cellular Oncology.

[21]  Y. DeClerck,et al.  The plasminogen activator inhibitor-1 paradox in cancer: a mechanistic understanding , 2019, Cancer and Metastasis Reviews.

[22]  H. Kwaan,et al.  Fibrin and Fibrinolysis in Cancer , 2019, Seminars in Thrombosis and Hemostasis.

[23]  B. Heissig,et al.  The fibrinolytic factor tPA drives LRP1‐mediated melanoma growth and metastasis , 2018, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[24]  Sung-Bae Kim,et al.  Efficacy and Safety of Pembrolizumab for Heavily Pretreated Patients With Advanced, Metastatic Adenocarcinoma or Squamous Cell Carcinoma of the Esophagus: The Phase 2 KEYNOTE-180 Study , 2019, JAMA oncology.

[25]  S. Yuan,et al.  Plasminogen activator inhibitor-1 in cancer research. , 2018, Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie.

[26]  N. Sun,et al.  Cisplatin-activated PAI-1 secretion in the cancer-associated fibroblasts with paracrine effects promoting esophageal squamous cell carcinoma progression and causing chemoresistance , 2018, Cell Death & Disease.

[27]  Kazuhiro Yoshida,et al.  Pembrolizumab versus paclitaxel for previously treated, advanced gastric or gastro-oesophageal junction cancer (KEYNOTE-061): a randomised, open-label, controlled, phase 3 trial , 2018, The Lancet.

[28]  Yue Fan,et al.  Oxymatrine inhibits the migration of human colorectal carcinoma RKO cells via inhibition of PAI-1 and the TGF-β1/Smad signaling pathway , 2016, Oncology reports.

[29]  A. Pathak,et al.  Intracellular Expression of PAI-1 Specific Aptamers Alters Breast Cancer Cell Migration, Invasion and Angiogenesis , 2016, PloS one.

[30]  L. Shen,et al.  Nimotuzumab plus paclitaxel and cisplatin as the first line treatment for advanced esophageal squamous cell cancer: A single centre prospective phase II trial , 2016, Cancer science.

[31]  P. Gettins,et al.  The High Affinity Binding Site on Plasminogen Activator Inhibitor-1 (PAI-1) for the Low Density Lipoprotein Receptor-related Protein (LRP1) Is Composed of Four Basic Residues* , 2015, The Journal of Biological Chemistry.

[32]  Y. DeClerck,et al.  Plasminogen Activator Inhibitor-1 in Cancer: Rationale and Insight for Future Therapeutic Testing. , 2015, Cancer research.

[33]  Wei Li,et al.  HIF-1α pathway: role, regulation and intervention for cancer therapy , 2015, Acta pharmaceutica Sinica. B.

[34]  M. Baltas,et al.  Small molecules inhibitors of plasminogen activator inhibitor-1 - an overview. , 2015, European journal of medicinal chemistry.

[35]  Yan Zhang,et al.  Expression of Bmi-1 and PAI-1 in esophageal squamous cell carcinoma. , 2014, World journal of gastroenterology.

[36]  J. Whisstock,et al.  Mechanistic characterization and crystal structure of a small molecule inactivator bound to plasminogen activator inhibitor-1 , 2013, Proceedings of the National Academy of Sciences.

[37]  H. Emonard,et al.  LRP-1: A Checkpoint for the Extracellular Matrix Proteolysis , 2013, BioMed research international.

[38]  A. Mazar,et al.  The Apparent uPA/PAI-1 Paradox in Cancer: More than Meets the Eye , 2013, Seminars in Thrombosis & Hemostasis.

[39]  A. Gils,et al.  The Biochemistry, Physiology and Pathological roles of PAI-1 and the requirements for PAI-1 inhibition in vivo. , 2012, Thrombosis research.

[40]  B. Sullenger,et al.  Effects of plasminogen activator inhibitor-1-specific RNA aptamers on cell adhesion, motility, and tube formation. , 2011, Nucleic acid therapeutics.

[41]  Heesang Song,et al.  Low-density lipoprotein receptor-related protein 1 promotes cancer cell migration and invasion by inducing the expression of matrix metalloproteinases 2 and 9. , 2009, Cancer research.

[42]  K. Cortese,et al.  Clathrin and LRP-1-Independent Constitutive Endocytosis and Recycling of uPAR , 2008, PloS one.

[43]  P. Andreasen PAI-1 - a potential therapeutic target in cancer. , 2007, Current drug targets.

[44]  K. Fan,et al.  Mechanism of Inactivation of Plasminogen Activator Inhibitor-1 by a Small Molecule Inhibitor* , 2007, Journal of Biological Chemistry.

[45]  I. Mikhailenko,et al.  Beyond endocytosis: LRP function in cell migration, proliferation and vascular permeability , 2005, Journal of thrombosis and haemostasis : JTH.

[46]  A. Gils,et al.  Plasminogen activator inhibitor-1. , 2004, Current medicinal chemistry.

[47]  M. Duffy,et al.  The urokinase plasminogen activator system: a rich source of tumour markers for the individualised management of patients with cancer. , 2004, Clinical biochemistry.

[48]  N. Brünner,et al.  XR5967, a novel modulator of plasminogen activator inhibitor-1 activity, suppresses tumor cell invasion and angiogenesis in vitro , 2004, Anti-cancer drugs.

[49]  P. Quax,et al.  Antibodies to PAI-1 alter the invasive and migratory properties of human tumour cells in vitro , 2004, Clinical & Experimental Metastasis.

[50]  P. Carmeliet,et al.  Hyperthermia inhibits angiogenesis by a plasminogen activator inhibitor 1-dependent mechanism. , 2003, Cancer research.

[51]  A. Gils,et al.  The Distal Hinge of the Reactive Site Loop and Its Proximity , 2001, The Journal of Biological Chemistry.

[52]  P. Declerck,et al.  Identification of a Functional Epitope in Plasminogen Activator Inhibitor-1, not Localized in the Reactive Center Loop , 1998, Thrombosis and Haemostasis.

[53]  H. Höfler,et al.  Clinical Impact of the Plasminogen Activation System in Tumor Invasion and Metastasis: Prognostic Relevance and Target for Therapy , 1997, Thrombosis and Haemostasis.

[54]  P. Declerck,et al.  Neutralization of plasminogen activator inhibitor-1 inhibitory properties: identification of two different mechanisms. , 1997, Biochimica et biophysica acta.

[55]  K. Miwa,et al.  An assay system for the modulators of plasminogen activation on the cell surface. , 1991, Thrombosis research.

[56]  T. Wun,et al.  Plasminogen activation: biochemistry, physiology, and therapeutics. , 1988, Critical reviews in biotechnology.

[57]  K. Danø,et al.  Monoclonal Antibodies to Human 54,000 Molecular Weight Plasminogen Activator Inhibitor from Fibrosarcoma Cells - Inhibitor Neutralization and One-Step Affinity Purification , 1986, Thrombosis and Haemostasis.