Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

Cancer Review Research

Plasminogen Activator Inhibitor-1 in Cancer: Rationale and Insight for Future Therapeutic Testing Veronica R. Placencio1,2 and Yves A. DeClerck1,2,3

Abstract

Despite its function as an inhibitor of and tissue-type led to the testing of small-molecule PAI-1 inhibitors, initially (PA), PA inhibitor-1 (PAI-1) has a paradox- developed as antithrombotic agents, in animal models of cancer. ical protumorigenic role in cancer, promoting angiogenesis and The review discusses the challenges and the opportunities that lay tumor cell survival. In this review, we summarize preclinical evi- aheadtothedevelopmentofefficacious and nontoxic PAI-1 inhi- dence in support of the protumorigenic function of PAI-1 that has bitors as anticancer agents. Cancer Res; 75(15); 2969–74. 2015 AACR.

Introduction Adding to the complexity of the function of PAI-1 in cancer is the fact that it can have paracrine and autocrine effects as it is Plasminogen activator inhibitor-1 (PAI-1) is a serine prote- produced by tumor cells and nonmalignant cells, including ase inhibitor () and the main regulator of the plasmin- endothelial cells (EC), macrophage cells, or adipocytes in the ogen activation system. It acts by inhibiting tissue-type plas- tumor microenvironment. minogen activator (tPA) and urokinase-type plasminogen On the basis of the protumorigenic role of uPA in angio- activator (uPA; ref. 1). PAI-1 has a pleiotropic biologic func- genesis, tumor invasion, and , it was anticipated that tion that stems from its complex structure. Three specific PAI-1 would have an antitumorigenic function. Surprisingly, -binding domains in the molecule have been well several studies revealed a paradoxical association between characterized (Fig. 1; ref. 1): a domain in the reactive center elevated levels of PAI-1 in blood and tissue samples of cancer loop (RCL) that binds to PA, a domain in the flexible joint patients and an unfavorable clinical outcome and poor region that binds to vitronectin composed of helix D (hD), response to therapy (1, 5). The observation that the production helix E (hE), and helix F (hF), and a domain within hD and hE of PAI-1 by EC, fibroblasts, adipocytes, smooth muscle cells, that binds to low-density lipoprotein receptor-related protein andmacrophagecellsinthetumormicroenvironmentwas (LRP1). Upon binding to the RCL, PA cleaves PAI-1 at P1-P1' stimulated by protumorigenic factors such as TGFb,IL6,and inducing a dynamic conformational change that inserts the TNFa added further evidence supporting a protumorigenic role RCL as a b-strand (4A) into the core of the protein. The binding (6, 7). In this article, we review the mechanisms and evidence in of PAI-1 to PA forms a catalytically inactive complex, which is mouse models supporting a protumorigenic function for PAI-1. internalized when combined with the receptor for uPA (uPAR) We then present recent pharmacologic approaches and preclin- and LRP1 (2). Premature insertion of the RCL into the b-sheet ical studies aimed at targeting PAI-1 in cancer and discuss the is the basis for the conversion of active PAI-1 into its latent lessons learned from these studies. form. Binding of PAI-1 to LRP1 also promotes cell migration and signaling (3). Binding of the flexible joint region of PAI-1 PAI-1 Is Proangiogenic to vitronectin has multiple consequences: it masks the adjacent fi RGD-binding site for av integrins and inhibits cell attachment A rst clue explaining the paradoxical association of PAI-1 to vitronectin (2) and stabilizes PAI-1 inhibiting its conversion with more aggressive forms of cancer came from the obser- to latency and increasing its anti-uPA ability by 200-fold (4). vation by several laboratories, including our own, that PAI-1 has a proangiogenic activity through its antiprotease and vitronectin-binding functions. The activity of PAI-1 on angio- genesis is, however, dose dependent with a promoting activity 1Division of Hematology, Oncology and Blood and Bone Marrow at physiologic concentrations (8, 9) and an inhibitory activity Transplantation, Department of Pediatrics, University of Southern California, Los Angeles, California. 2The Saban Research Institute of at pharmacologic concentrations (10). This is explained by Children's Hospital Los Angeles, Los Angeles, California. 3Department the fact that at physiologic concentrations, PAI-1 inhibition of of Biochemistry and Molecular Biology, University of Southern Cali- uPA limits the cleavage by pericellular plasmin of membrane- fornia, Los Angeles, California. associated Fas-L at R144-K145 and the release of a soluble Corresponding Author: Yves A. DeClerck, Children's Hospital Los Angeles, 4650 Fas-L fragment that induces Fas-mediated apoptosis in EC Sunset Boulevard, MS#54, Los Angeles, CA 90027. Phone: 323-361-2150; Fax: (11). PAI-1 also stimulates angiogenesis through its vitronec- 323-361-4902; E-mail: [email protected] tin-binding function promoting the detachment of EC doi: 10.1158/0008-5472.CAN-15-0876 from vitronectin and their migration toward fibronectin rich 2015 American Association for Cancer Research. tissues (9).

www.aacrjournals.org 2969

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

Placencio and DeClerck

A s4A uPA/ tPA binding site (RCL insertion) RCL

sB/sC pocket

Figure 1. Structure of PAI-1 and inhibitor Active Latent targeting sites. A, structure of PAI-1 and targeted binding sites. The hD reactive center loop (RCL) is shown in Vitronectin LRP1 binding red, which inserts itself to become s4A binding site site in the latent form of PAI-1. The sB/sC (hD, hE, and hF) hE pocket is indicated by the arrow (purple). The vitronectin (hD, hE, and hF hF) and LRP1 (hD and hE) binding regions are shown in purple and blue, respectively. Molecular modeling was B carried out using Pymol software Apoptosis, Migration, Angiogenesis, 123version 1.2r1 and the PDB files 1B3K for angiogenesis signaling migration active PAI-1 (57) and 1DVN for latent PAI-1 (58). B, roles of PAI-1 in tumorigenesis: (1) the inhibitory role Plasminogen of PAI-1 on uPA action to prevent the cleavage of plasminogen into plasmin uPA Vitronectin and subsequent substrates on the cell surface, leading to angiogenesis or intracellular apoptotic signaling; (2) the ability of PAI-1 to bind to LRP1, Plasmin directly leading to intracellular signaling and cell migration; and (3) the ability of PAI-1 to bind to uPAR LRP1 Integrin vitronectin in the extracellular matrix to promote endothelial cell migration and angiogenesis.

© 2015 American Association for Cancer Research

PAI-1 Inhibits Spontaneous Apoptosis in maximum antitumor effect was shown upon PAI-1 suppression in Cancer Cells both host and tumor cells pointing to a combined role for extracellular PAI-1. Similar observations were made when PAI- PAI-1 protects tumor cells from apoptosis through multiple 1–deficient human tumor cells were xenotransplanted in immu- mechanisms. Similar to EC, it inhibits Fas-mediated apoptosis in nodeficient PAI-1 null mice. These studies typically demonstrated several human cancer cells, including brain metastasis through its that suppression of PAI-1 expression in human tumor cells and in control over pericellular plasmin activity (12, 13). PAI-1 also mouse host cells resulted in poor tumor take and slower tumor affects intrinsic apoptosis, as the absence of extracellular PAI-1 in growth (11, 12, 22, 23). However in some situations, such as in tumor cells results in higher levels of activated caspase-9 (14). skin carcinoma, the effects observed depended on the tumor grade Intracellular PAI-1 also promotes cell survival through its ability with minimal effects observed in high-grade tumors (23). to inhibit caspase-3 protecting tumor cells from chemotherapy- In contrast with the above data, experiments using genetically induced apoptosis (15). engineered mice (GEM) prone to develop cancer crossed with PAI- 1 null mice failed to demonstrate any effect of PAI-1 suppression What Have We Learned from Mouse Tumor on tumor initiation, growth, or metastasis. For example, when FVB- PymT mice prone to develop mammary tumors were crossed with Models? PAI-1 null mice, there was no effect on the development of Observations in PAI-1–deficient mice have provided additional mammary tumors, metastasis, or survival (24). Similarly, genetic and important clues on the function of PAI-1 in tumorigenesis ablation of PAI-1 had no effect on tumor development in Apc/ – – and angiogenesis. The vast majority of experiments using PAI-1– Apc1638N/PAI-1 / mice prone to develop colon cancer (25), in deficient murine tumor cells syngeneically implanted into WT or TRP-1SV40 Tag/PAI-1 / mice prone to develop ocular tumors PAI-1–deficient mice consistently demonstrated the contributory (26), and in K14-HPV16/PAI-1 / mice prone to develop skin role of host and tumor-derived PAI-1 to tumor growth, angio- cancer (27). An explanation for this difference of effect genesis, and metastasis (Table 1; refs. 10, 16–21). Consistently, between transplanted and GEM mice is the possible presence of

2970 Cancer Res; 75(15) August 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

PAI-1 Inhibition in Preclinical Cancer Models

Table 1. Outcome of the genetic ablation of PAI-1 in the host and/or implanted tumor cells in murine models of cancer Model PAI-1 host mouse Implanted cells Tumor growth Angiogenesis Metastasis Reference Syngeneic implantation þ/þ B16 melanoma No difference n/e No difference 21 / B16 melanoma No difference n/e No difference 21 / Malignant keratinocytes Decreased Decreased n/e 17 / T241 fibrosarcoma Decreased Decreased Decreased 16 þ/þ Malignant keratinocytes Decreased Decreased n/e 10 / Malignant keratinocytes Decreased Decreased n/e 10 / Spontaneously transformed Decreased n/e n/e 18 primary lung fibroblasts / Spontaneously transformed Decreased n/e n/e 19 primary fibrosarcoma / Spontaneously transformed Decreased n/e n/e 20 primary fibrosarcoma Xenograft / Human neuroblastoma Decreased Decreased n/e 11 þ/þ Human M21 melanoma Increased Increased n/e 22 / Human M21 melanoma Decreased Decreased n/e 22 / Human HaCaTII-4 and HaCaT Decreased Decreased n/e 23 A5-RT3 / Human PAI-1 KD HT1080 Decreased Decreased n/e 12 fibrosarcoma, colon cancer HCT116, breast cancer MDA- MB-231, lung cancer A549 GEMM MMTV-PymT/PAI-1/ No difference No difference No difference 24 Apc/Apc1638N/PAI-1/ No difference n/e n/e 25 TRP-1/SV40 Tag/PAI-1/ No difference No difference Decreased 26 K14-HPV16/PAI-1/ No difference No difference n/e 27 Abbreviations: GEMM, genetically engineered mouse model; þ/þ, transgenic overexpressing PAI-1 mice; /, PAI-1 deficient mice; n/e, the experimental parameter was not evaluated.

compensatory , including PAI-2, inhibitor, pro- rombotic activity in preclinical rat and canine models of acute tease nexin-1, or (28–31), whose overexpression in trans- arterial thrombosis (38, 39). Other inhibitors derived from the formed cells may have compensated for a lack of PAI-1 (27–31). structure of this inhibitor (PAI-749 and PAZ-417) then underwent human phase I clinical trials in healthy volunteers and in patients with Alzheimer's disease (40–42). To date, the results of these Pharmacologic Inhibition of PAI-1 studies have not been reported. Over the last two decades, several laboratories and pharmaceu- As an alternative approach, using computer simulation models tical companies have developed a variety of small-molecule PAI-1 mimicking the RCL insertion within the strands of b-sheet A, inhibitors using high-throughput screening (32). The vast major- conformation disrupting agents were designed. Such compounds ity of these inhibitors are molecules interfering with the molecular include a family of dimeric 2-acylamino-3-thiophenecarboxylic interactions of PAI-1 to inhibit tPA and uPA by binding to the acid derivatives (TM5001 and TM5007) developed with the reactive center loop and by inducing a conformational change that anticipation that by binding to the s4A position of the A b-sheet, promotes an irreversible conversion of PAI-1 into its latent form. they would induce the conversion of PAI-1 into its latent and To our knowledge, no inhibitors specifically interacting with the inactive form (43). These inhibitors were found active against vitronectin-binding domain of PAI-1 have been reported yet. The vascular thrombosis in a rat model and against lung fibrosis in main focus of investigations on the activity of these inhibitors in murine models. Their pharmacokinetic properties, however, were biologic processes has been in cardiovascular diseases (thrombus suboptimal due to their relatively high lipophilicity [calculated repermeabilization), lung fibrosis, Alzheimer's disease and to a octanol/water partition coefficient (ClogP) value of 5.79, above lesser degree in cancer. A first family of PAI-1 inhibitors developed the ideal 2–3 value]. A second-generation molecule (TM5275) consisted of diketopiperazine inhibitors that interfere with uPA with better oral pharmacokinetic properties and lower ClogP of binding to the -binding domain of the RCL. These inhi- 3.37 was then developed (44). TM5275 showed activity in rat and bitors (XR334, XR1853, XR5082, and XR5118) block tPA inhibi- mouse models of acute arterial thrombosis and lung fibrosis. tion by PAI-1 in vitro (33, 34). However, their in vivo efficacy is Importantly, this inhibitor had no systemic toxicity as it did not limited by their low solubility, poor oral availability, or high IC50 prolong bleeding time in nonhuman primates (45). values (in the 5 to >1,000 mmol/L range) that are unachievable in A limiting element in the activity of these inhibitors so far, has the blood upon administration in animals. Other menthol-based, been their lack of activity against the stable form of PAI-1 bound to benzothiophene, and butadiene small molecules with lower IC50 vitronectin (46). Thus, the recent focus has been on the devel- values (0.64 to 44 mmol/L) in vitro were developed (35–37); opment of inhibitors active against vitronectin-bound PAI-1. however, despite their good anti-PAI-1 activity in vitro, they were These efforts, however, have been limited by a lack of crystal either not tested or not further pursued in animal experiments for structure information on vitronectin-bound PAI-1 (47). Polyphe- reasons not reported. nolic inhibitors of PAI-1 (CDE-066 and 096) synthesized on the One indole oxoacetic acid PAI-1 inhibitor, PAI-039 (tiplaxti- basis of the structure of small molecules identified by a high- nin; ref. 38), was extensively and successfully tested for its antith- throughput screen were found to bind to the sB/sC pocket of PAI-1

www.aacrjournals.org Cancer Res; 75(15) August 1, 2015 2971

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

Placencio and DeClerck

Table 2. Outcome of pharmacologic inhibition of PAI-1 in preclinical murine models of cancer Efficacy in cancer models

Compound Inhibitor class IC50 (mmol/L) Half-life (h) Cancer model In vitro Tumor growth Angiogenesis Metastasis PAI-039 Indole oxoacetic acid 2.7 2.95–3.73 Bladder and cervical cancer Yes Yes Yes n/e TM5275 N-acylanthranilic acid 6.9 2.5 Ovarian cancer Yes n/e n/e n/e SK-116, SK-216 Not stated 35, 44 n/e ApcMin/þ GEMM Yes Yes n/e n/e SK-216 Not stated 44 n/e Lung cancer and melanoma Yes Yes Yes Yes Abbreviation: n/e, the experimental parameter was not evaluated.

with an IC50 of 32 mmol/L and 25 nmol/L, respectively (Fig. 1; cancer progression, its potential as a target for therapeutic inter- refs. 48, 49). CDE-096 induces allosteric conformational changes vention in cancer has only been recently considered and explored. affecting the flexibility of PAI-1 and preventing it from binding to These studies provide important insight on the need to develop PA and decreasing vitronectin binding (49). CDE-096 was still better inhibitors for cancer. The need for chronic administration able to interact with vitronectin-bound PAI-1, although the mag- and thus a pharmacologic profile with a prolonged plasma half- nitude of polarization was decreased 7.3-fold compared with life of orally available inhibitors is an important consideration. binding-free PAI-1. The in vivo pharmacokinetic profile of these Although much work has been done on the development of orally inhibitors is unknown. available compounds, the half-life of these inhibitors in vivo is 2 to 3 hours and therefore would be impractical in long-term cancer Pharmacologic Inhibition of PAI-1 in Cancer therapy. The fact that most inhibitors developed until now are Therapy active in vitro at concentrations in the mmol/L range represents another limitation. Such concentrations are typically difficult to Pharmacologic inhibition of PAI-1 in cardiovascular diseases reach in blood and more importantly in tumor tissues for an has been the primary goal with the objective to prevent inhibition extensive period of time. The lack of activity of most inhibitors fi of intravascular brinolysis and subsequently promote thrombus against the stable vitronectin-bound form of PAI-1 is a third repermeabilization in an acute setting. In contrast, preclinical limitation, since this is the predominant form of PAI-1 in vitro- fi evidence supporting the therapeutic ef cacy of small PAI-1 inhi- nectin-rich tumor tissues (55). A fourth limitation is the potential bitors in cancer has been limited (Table 2). In cancer, the objective systemic effect that the chronic administration of PAI-1 inhibitors is to prevent inhibition of pericellular activation of plasminogen may have on hemostasis. By suppressing the control that PAI-1 and interfere with PAI-1-vitronectin interactions. Although in exerts on plasmin-mediated fibrinolysis, small-molecule PAI-1 cardiovascular and thrombotic disease, the desired inhibition is inhibitors may impair hemostasis and cause excessive spontane- short-term; in cancer (or other chronic conditions), it is long-term ous bleeding (56). Although studies in nonhuman primates have fi requiring a pharmacologic pro le suitable for chronic adminis- shown an absence of side effects on systemic hemostasis follow- tration (32). ing the chronic administration of some PAI-1 inhibitors (44, 45), Insofar, four small-molecule PAI-1 inhibitors have been tested this aspect deserves more extensive investigation. in preclinical models of cancer. Although limited, these studies In summary, targeting PAI-1 in cancer therapy remains an have shown antitumor activity. SK-116 and SK-216 inhibitors attractive but also challenging approach that will require the administered orally for 9 weeks to Apc/Apc1638N mice that design and development of inhibitors that address some of the spontaneously developed intestinal polyps caused an almost 2- current limitations outlined above. As new inhibitors with better fold reduction in the number of small intestinal polyps (50). pharmacologic profiles are developed, they should continue to be When administered to PAI-1 producing Lewis lung carcinoma and tested in preclinical models of cancer. PAI-1–nonproducing B16 melanoma tumor-bearing mice, SK- 216 caused a 2-fold reduction in subcutaneous primary tumor fl size and inhibited angiogenesis and metastases (51). The oral Disclosure of Potential Con icts of Interest fl administration of PAI-039 to mice xenotransplanted with human No potential con icts of interest were disclosed. T24 bladder and HeLa cervical cancer cells resulted in a 2-fold reduction of tumor volume after 14 days associated with a Acknowledgments decrease in tumor cell proliferation and vascularization and an The authors thank Dr. Marta Kubala for reading through the article and increase in apoptosis (52). The TM5275 inhibitor was recently showing them how to generate the crystal structure to highlight the inhibitor- shown to increase apoptosis in vitro in ovarian lines binding sites. (53). The in vivo activity of these inhibitors in cancer models have not been reported with the exception of TM5275 and TM5441 in a Grant Support most recent study (54). This work was supported by the U.S. Department of Health and Human Services/National Institutes of Health with a grant (Y.A. DeClerck; grant 5R01 Challenges and Opportunities CA 129377).

Despite the fact that much is known on the structure of PAI-1, Received March 31, 2015; revised April 27, 2015; accepted April 28, 2015; its complex biologic function, and its protumorigenic role in published OnlineFirst July 15, 2015.

References 1. McMahon B, Kwaan HC. The plasminogen activator system and cancer. 2. Schroeck F, Arroyo de Prada N, Sperl S, Schmitt M, Viktor M. Interaction of Pathophysiol Haemost Thromb 2008;36:184–94. plasminogen activator inhibitor type-1 (PAI-1) with vitronectin (Vn):

2972 Cancer Res; 75(15) August 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

PAI-1 Inhibition in Preclinical Cancer Models

mapping the binding sites on PAI-1 and Vn. Biol Chem 2002;383: 23. Maillard C, Jost M, Romer MU, Brunner N, Houard X, Lejeune A, et al. 1143–9. Host plasminogen activator inhibitor-1 promotes human skin carcino- 3. Kwaan HC, Mazar AP, McMahon BJ. The apparent uPA/PAI-1 paradox in ma progression in a stage-dependent manner. Neoplasia 2005;7:57–66. cancer: more than meets the eye. Semin Thromb Hemost 2013;39: 24. Almholt K, Nielsen BS, Frandsen TL, Brunner N, Dano K, Johnsen M. 382–91. Metastasis of transgenic breast cancer in plasminogen activator inhibitor-1 4. Wind T, Hansen M, Jensen JK, Andreasen PA. The molecular basis for -deficient mice. 2003;22:4389–97. anti-proteolytic and non-proteolytic functions of plasminogen activator 25. Fen Li C, Kandel C, Baliko F, Nadesan P, Brunner N, Alman BA. Plasmin- inhibitor type-1: roles of the reactive centre loop, the shutter region, the ogen activator inhibitor-1 (PAI-1) modifies the formation of aggressive flexible joint region and the small serpin fragment. Biol Chem 2002; fibromatosis (desmoid tumor). Oncogene 2005;24:1615–24. 383:21–36. 26. Maillard CM, Bouquet C, Petitjean MM, Mestdagt M, Frau E, Jost M, 5. Foekens JA, Peters HA, Look MP, Portengen H, Schmitt M, Kramer MD, et al. Reduction of brain metastases in plasminogen activator inhibitor- et al. The urokinase system of plasminogen activation and prognosis in 1-deficient mice with transgenic ocular tumors. Carcinogenesis 2008; 2780 breast cancer patients. Cancer Res 2000;60:636–43. 29:2236–42. 6. Dennler S, Itoh S, Vivien D, ten Dijke P, Huet S, Gauthier JM. Direct binding 27. Masset A, Maillard C, Sounni NE, Jacobs N, Bruyere F, Delvenne P, et al. of Smad3 and Smad4 to critical TGF beta-inducible elements in the Unimpeded skin carcinogenesis in K14-HPV16 transgenic mice deficient promoter of human plasminogen activator inhibitor-type 1 gene. EMBO for plasminogen activator inhibitor. Int J Cancer 2011;128:283–93. J 1998;17:3091–100. 28. Biliran H Jr, Sheng S. Pleiotrophic inhibition of pericellular urokinase-type 7. Brown NJ. Therapeutic potential of plasminogen activator inhibitor-1 plasminogen activator system by endogenous tumor suppressive maspin. inhibitors. Ther Adv Cardiovasc Dis 2010;4:315–24. Cancer Res 2001;61:8676–82. 8. Bajou K, Masson V, Gerard RD, Schmitt PM, Albert V, Praus M, et al. The 29. Espana F, Estelles A, Fernandez PJ, Gilabert J, Sanchez-Cuenca J, GriffinJH. plasminogen activator inhibitor PAI-1 controls in vivo tumor vasculariza- Evidence for the regulation of urokinase and tissue type plasminogen tion by interaction with , not vitronectin. Implications for anti- activators by the serpin, , in semen and blood plasma. angiogenic strategies. J Cell Biol 2001;152:777–84. Thromb Haemost 1993;70:989–94. 9. Isogai C, Laug WE, Shimada H, Declerck PJ, Stins MF, Durden DL, et al. 30. Montemurro P, Barbuti G, Conese M, Gabriele S, Petio M, Colucci M, et al. Plasminogen activator inhibitor-1 promotes angiogenesis by stimulat- Retinoic acid stimulates plasminogen activator inhibitor 2 production by ing endothelial cell migration toward fibronectin. Cancer Res 2001;61: blood mononuclear cells and inhibits urokinase-induced extracellular 5587–94. proteolysis. Br J Haematol 1999;107:294–9. 10. Bajou K, Maillard C, Jost M, Lijnen RH, Gils A, Declerck P, et al. Host- 31. Scott RW, Bergman BL, Bajpai A, Hersh RT, Rodriguez H, Jones BN, et al. derived plasminogen activator inhibitor-1 (PAI-1) concentration is critical Protease nexin. Properties and a modified purification procedure. J Biol for in vivo tumoral angiogenesis and growth. Oncogene 2004;23:6986–90. Chem 1985;260:7029–34. 11. Bajou K, Peng H, Laug WE, Maillard C, Noel A, Foidart JM, et al. Plasmin- 32. Fortenberry YM. Plasminogen activator inhibitor-1 inhibitors: a patent ogen activator inhibitor-1 protects endothelial cells from FasL-mediated review (2006-present). Expert Opin Ther Pat 2013;23:801–15. apoptosis. Cancer cell 2008;14:324–34. 33. CharltonPA,FaintRW,BentF,Bryans J, Chicarelli-Robinson I, Mackie I, 12. Fang H, Placencio VR, DeClerck YA. Protumorigenic activity of plasmin- et al. Evaluation of a low molecular weight modulator of human ogen activator inhibitor-1 through an antiapoptotic function. J Natl Cancer plasminogen activator inhibitor-1 activity. Thromb Haemost 1996;75: Inst 2012;104:1470–84. 808–15. 13. Valiente M, Obenauf AC, Jin X, Chen Q, Zhang XH, Lee DJ, et al. Serpins 34. Friederich PW, Levi M, Biemond BJ, Charlton P, Templeton D, van promote cancer cell survival and vascular co-option in brain metastasis. Zonneveld AJ, et al. Novel low-molecular-weight inhibitor of PAI-1 Cell 2014;156:1002–16. (XR5118) promotes endogenous fibrinolysis and reduces postthrombo- 14. Balsara RD, Castellino FJ, Ploplis VA. A novel function of plasminogen lysis thrombus growth in rabbits. Circulation 1997;96:916–21. activator inhibitor-1 in modulation of the AKT pathway in wild-type and 35. Liang A, Wu F, Tran K, Jones SW, Deng G, Ye B, et al. Characterization of a plasminogen activator inhibitor-1-deficient endothelial cells. J Biol Chem small molecule PAI-1 inhibitor, ZK4044. Thromb Res 2005;115:341–50. 2006;281:22527–36. 36. Rupin A, Gaertner R, Mennecier P, Richard I, Benoist A, De Nanteuil G, et al. 15. Schneider DJ, Chen Y, Sobel BE. The effect of plasminogen activator S35225 is a direct inhibitor of Plasminogen Activator Inhibitor type-1 inhibitor type 1 on apoptosis. Thromb Haemost 2008;100:1037–40. activity in the blood. Thromb Res 2008;122:265–70. 16. Gutierrez LS, Schulman A, Brito-Robinson T, Noria F, Ploplis VA, Castel- 37. Miyazaki H, Ogiku T, Sai H, Ohmizu H, Murakami J, Ohtani A. Design, lino FJ. Tumor development is retarded in mice lacking the gene for synthesis, and evaluation of orally active inhibitors of plasminogen acti- urokinase-type plasminogen activator or its inhibitor, plasminogen acti- vator inhibitor-1 (PAI-1) production. Bioorg Med Chem Lett 2008;18: vator inhibitor-1. Cancer Res 2000;60:5839–47. 6419–22. 17. Bajou K, Noel A, Gerard RD, Masson V, Brunner N, Holst-Hansen C, et al. 38. Elokdah H, Abou-Gharbia M, Hennan JK, McFarlane G, Mugford CP, Absence of host plasminogen activator inhibitor 1 prevents cancer invasion Krishnamurthy G, et al. Tiplaxtinin, a novel, orally efficacious inhibitor and vascularization. Nat Med 1998;4:923–8. of plasminogen activator inhibitor-1: design, synthesis, and preclinical 18. Lademann U, Romer MU, Jensen PB, Hofland KF, Larsen L, Christensen characterization. J Med Chem 2004;47:3491–4. IJ, et al. Malignant transformation of wild-type but not plasminogen 39. Hennan JK, Morgan GA, Swillo RE, Antrilli TM, Mugford C, Vlasuk GP, et al. activator inhibitor-1 gene-deficient fibroblasts decreases cellular sensi- Effect of tiplaxtinin (PAI-039), an orally bioavailable PAI-1 antagonist, in a tivity to chemotherapy-mediated apoptosis. Eur J Cancer 2005;41: rat model of thrombosis. J Thromb Haemost 2008;6:1558–64. 1095–100. 40. Lucking AJ, Visvanathan A, Philippou H, Fraser S, Grant PJ, Connolly TM, 19. Romer MU, Kirkebjerg Due A, Knud Larsen J, Hofland KF, Christensen IJ, et al. Effect of the small molecule plasminogen activator inhibitor-1 (PAI- Buhl-Jensen P, et al. Indication of a role of plasminogen activator inhibitor 1) inhibitor, PAI-749, in clinical models of fibrinolysis. J Thromb Haemost type I in protecting murine fibrosarcoma cells against apoptosis. Thromb 2010;8:1333–9. Haemost 2005;94:859–66. 41. Jacobsen JS, Comery TA, Martone RL, Elokdah H, Crandall DL, Oganesian 20. Romer MU, Larsen L, Offenberg H, Brunner N, Lademann UA. Plasmin- A, et al. Enhanced clearance of Abeta in brain by sustaining the plasmin ogen activator inhibitor 1 protects fibrosarcoma cells from etoposide- proteolysis cascade. Proc Natl Acad Sci U S A 2008;105:8754–9. induced apoptosis through activation of the PI3K/Akt cell survival path- 42. Barker R, Kehoe PG, Love S. Activators and inhibitors of the plasminogen way. Neoplasia 2008;10:1083–91. system in Alzheimer's disease. J Cell Mol Med 2012;16:865–76. 21. Eitzman DT, Krauss JC, Shen T, Cui J, Ginsburg. Lack of plasminogen 43. Izuhara Y, Takahashi S, Nangaku M, Takizawa S, Ishida H, Kurokawa K, activator inhibitor-1 effect in a transgenic mouse model of metastatic et al. Inhibition of plasminogen activator inhibitor-1: its mechanism and melanoma. Blood 1996;87:4718–22. effectiveness on and fibrosis. Arterioscler Thromb Vasc Biol 22. McMahon GA, Petitclerc E, Stefansson S, Smith E, Wong MK, Westrick RJ, 2008;28:672–7. et al. Plasminogen activator inhibitor-1 regulates tumor growth and 44. Izuhara Y, Yamaoka N, Kodama H, Dan T, Takizawa S, Hirayama N, angiogenesis. J Biol Chem 2001;276:33964–8. et al. A novel inhibitor of plasminogen activator inhibitor-1 provides

www.aacrjournals.org Cancer Res; 75(15) August 1, 2015 2973

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

Placencio and DeClerck

antithrombotic benefits devoid of bleeding effect in nonhuman pri- 52. Gomes-Giacoia E, Miyake M, Goodison S, Rosser CJ. Targeting plas- mates. J Cereb Blood Flow Metab 2010;30:904–12. minogen activator inhibitor-1 inhibits angiogenesis and tumor 45. Yamaoka N, Kawano Y, Izuhara Y, Miyata T, Meguro K. Structure-activity growth in a human cancer xenograft model.MolCancerTher2013;12: relationships of new 2-acylamino-3-thiophenecarboxylic acid dimers as 2697–708. plasminogen activator inhibitor-1 inhibitors. Chem Pharm Bull 2010; 53. Mashiko S, Kitatani K, Toyoshima M, Ichimura A, Dan T, Usui T, et al. 58:615–9. Inhibition of plasminogen activator inhibitor-1 is a potential therapeutic 46. Gorlatova NV, Cale JM, Elokdah H, Li D, Fan K, Warnock M, et al. strategy in ovarian cancer. Cancer Biol Ther 2015:16:253–60. Mechanism of inactivation of plasminogen activator inhibitor-1 by a small 54. Placencio VR, Ichimura A, Miyata T, DeClerck YA. Small molecule inhi- molecule inhibitor. J Biol Chem 2007;282:9288–96. bitors of plasminogen activator inhibitor-1 elicit anti-tumorigenic and 47. Rouch A, Vanucci-Bacque C, Bedos-Belval F, Baltas M. Small molecules anti-angiogenic activity. PLoS ONE 2015 (manuscript submitted). inhibitors of plasminogen activator inhibitor-1 - An overview. Eur J Med 55. Declerck PJ, De Mol M, Alessi MC, Baudner S, Paques EP, Preissner KT, Chem 2015;92:619–36. et al. Purification and characterization of a plasminogen activator 48. Cale JM, Li SH, Warnock M, Su EJ, North PR, Sanders KL, et al. Charac- inhibitor 1 binding protein from human plasma. Identification as a terization of a novel class of polyphenolic inhibitors of plasminogen multimeric form of S protein (vitronectin). J Biol Chem 1988;263: activator inhibitor-1. J Biol Chem 2010;285:7892–902. 15454–61. 49. Li SH, Reinke AA, Sanders KL, Emal CD, Whisstock JC, Stuckey JA, et al. 56. Fay WP, Parker AC, Condrey LR, Shapiro AD. Human plasminogen Mechanistic characterization and crystal structure of a small molecule activator inhibitor-1 (PAI-1) deficiency: characterization of a large kindred inactivator bound to plasminogen activator inhibitor-1. Proc Natl Acad with a null mutation in the PAI-1 gene. Blood 1997;90:204–8. Sci U S A 2013;110:E4941–9. 57. Sharp AM, Stein PE, Pannu NS, Carrell RW, Berkenpas MB, Ginsburg D, 50. Mutoh M, Niho N, Komiya M, Takahashi M, Ohtsubo R, Nakatogawa K, et al. The active conformation of plasminogen activator inhibitor 1, a et al. Plasminogen activator inhibitor-1 (Pai-1) blockers suppress intestinal target for drugs to control fibrinolysis and cell adhesion. Structure polyp formation in Min mice. Carcinogenesis 2008;29:824–9. 1999;7:111–8. 51. Masuda T, Hattori N, Senoo T, Akita S, Ishikawa N, Fujitaka K, et al. SK-216, 58. Stout TJ, Graham H, Buckley DI, Matthews DJ. Structures of active and an inhibitor of plasminogen activator inhibitor-1, limits tumor progres- latent PAI-1: a possible stabilizing role for chloride ions. Biochemistry sion and angiogenesis. Mol Cancer Ther 2013;12:2378–88. 2000;39:8460–9.

2974 Cancer Res; 75(15) August 1, 2015 Cancer Research

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research. Published OnlineFirst July 15, 2015; DOI: 10.1158/0008-5472.CAN-15-0876

Plasminogen Activator Inhibitor-1 in Cancer: Rationale and Insight for Future Therapeutic Testing

Veronica R. Placencio and Yves A. DeClerck

Cancer Res 2015;75:2969-2974. Published OnlineFirst July 15, 2015.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-15-0876

Cited articles This article cites 56 articles, 19 of which you can access for free at: http://cancerres.aacrjournals.org/content/75/15/2969.full#ref-list-1

Citing articles This article has been cited by 7 HighWire-hosted articles. Access the articles at: http://cancerres.aacrjournals.org/content/75/15/2969.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at Subscriptions [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/75/15/2969. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 30, 2021. © 2015 American Association for Cancer Research.