cancers

Article New Heparanase-Inhibiting Triazolo-Thiadiazoles Attenuate Primary Tumor Growth and Metastasis

Uri Barash 1,† , Shobith Rangappa 2,†, Chakrabhavi Dhananjaya Mohan 3, Divakar Vishwanath 4 , Ilanit Boyango 1, Basappa Basappa 4 , Israel Vlodavsky 1,* and Kanchugarakoppal S. Rangappa 5,*

1 Technion Integrated Cancer Center (TICC), the Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel; [email protected] (U.B.); [email protected] (I.B.) 2 Adichunchanagiri Institute for Molecular Medicine, BG Nagara, Nagamangala Taluk 571448, India; [email protected] 3 Department of Studies in Molecular Biology, University of Mysore, Manasagangotri, Mysore 570006, India; [email protected] 4 Laboratory of Chemical Biology, Department of Studies in Organic Chemistry, University of Mysore, Manasagangotri, Mysore 570006, India; [email protected] (D.V.); [email protected] (B.B.) 5 Institution of Excellence, Vijnana Bhavan, University of Mysore, Manasagangotri, Mysore 570006, India * Correspondence: [email protected] (I.V.); [email protected] (K.S.R.) † Equal contribution.

Simple Summary: Heparanase is an endoglycosidase that plays a critical role in tumor progression

 and metastasis. The expression of heparanase in the tumor microenvironment is positively correlated  with the aggressiveness of the tumor and is associated with poor prognosis. In this study, we have demonstrated that a new triazole–thiadiazole-bearing small molecule showed good heparanase Citation: Barash, U.; Rangappa, S.; Mohan, C.D.; Vishwanath, D.; inhibition along with attenuation of tumor growth and metastasis. To the best of our knowledge, this Boyango, I.; Basappa, B.; Vlodavsky, is the first report showing a marked decrease in primary tumor growth in mice treated with a small I.; Rangappa, K.S. New molecule that inhibits heparanase enzymatic activity. Given these encouraging results, studies are Heparanase-Inhibiting underway to better elucidate the mode of action and clinical significance of triazolo–thiadiazoles. Triazolo-Thiadiazoles Attenuate Primary Tumor Growth and Abstract: Compelling evidence ties heparanase, an endoglycosidase that cleaves Metastasis. Cancers 2021, 13, 2959. side (HS) chains of , with all steps of tumor development, including tumor initiation, https://doi.org/10.3390/cancers angiogenesis, growth, metastasis, and chemoresistance. Moreover, heparanase levels correlate with 13122959 shorter postoperative survival of cancer patients, encouraging the development of heparanase in- hibitors as anti-cancer drugs. Heparanase-inhibiting /heparan sulfate-mimicking compounds Academic Editor: Ian R. Hardcastle and neutralizing antibodies are highly effective in animal models of cancer progression, yet none of the compounds reached the stage of approval for clinical use. The present study focused on Received: 27 April 2021 Accepted: 22 May 2021 newly synthesized triazolo–thiadiazoles, of which compound 4-iodo-2-(3-(p-tolyl)-[1,2,4]triazolo[3,4- Published: 13 June 2021 b][1,3,4]thiadiazol-6-yl)phenol (4-MMI) was identified as a potent inhibitor of heparanase enzymatic activity, cell invasion, experimental metastasis, and tumor growth in mouse models. To the best

Publisher’s Note: MDPI stays neutral of our knowledge, this is the first report showing a marked decrease in primary tumor growth in with regard to jurisdictional claims in mice treated with small molecules that inhibit heparanase enzymatic activity. This result encourages published maps and institutional affil- the optimization of 4-MMI for preclinical and clinical studies primarily in cancer but also other iations. indications (i.e., colitis, pancreatitis, diabetic nephropathy, tissue fibrosis) involving heparanase, including viral infection and COVID-19.

Keywords: triazolo–thiadiazoles; small molecules; heparanase-inhibiting compounds; primary Copyright: © 2021 by the authors. tumor growth; cell invasion; metastasis Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons 1. Introduction Attribution (CC BY) license (https:// The (ECM) is a supramolecular network of macromolecules (i.e., creativecommons.org/licenses/by/ collagens, glycoproteins proteoglycans) that provide structural and biochemical support to 4.0/).

Cancers 2021, 13, 2959. https://doi.org/10.3390/cancers13122959 https://www.mdpi.com/journal/cancers Cancers 2021, 13, 2959 2 of 17

the cellular compartment of tissues [1,2]. Heparan sulfate (HS) is a linear glycosaminogly- can polysaccharide made up of tandem disaccharide units of glucuronic acid or iduronic acid and D-glucosamine, which are covalently attached to a core protein (i.e., , glypican, perlecan) together forming heparan sulfate proteoglycans (HSPG) [3,4]. Although there are only two repeating sugar units, they exhibit broad diversity in structure generated by a complex pattern of deacetylation, sulfation, and epimerization [3]. Importantly, the sulfated regions of HS serve as docking sites for ECM components, extrinsic proteins, growth-promoting factors (i.e., VEGF, FGF, HGF, TGFβ), chemokines, and cytokines [4,5], indicating that HSPG is crucial in maintaining tissue/cell architecture, growth, and dif- ferentiation [3,6]. Heparanase is the only β-endoglycosidase that partially degrades the HS saccharide side chains of HSPG to generate fragments of 4–7 kDa in length [7–9]. The degradation products of HS and, even more so, the liberated biologically active molecules, serve as a bioavailable source of HS-bound proteins that activate signal transduction and promote tissue remodeling, repair, neovascularization, and growth [8,9]. Compelling ev- idence gathered during the last two decades tie heparanase levels not only with tumor metastasis but with all steps of tumor development, including tumor initiation, angiogene- sis, growth, metastasis, and chemoresistance [8,9]. Moreover, heparanase levels correlate with shorter postoperative survival of cancer patients [10–13] altogether indicating that heparanase is causally involved in cancer progression, encouraging the development of heparanase inhibitors as anti-cancer drugs. Heparanase-inhibiting heparin/HS-mimicking compounds, small molecules, and antibodies are being developed [14–16], of which sev- eral saccharide-based compounds were and/or are being examined in cancer clinical trials [17,18], yet none of the compounds reached the stage of approval for clinical use. The present study is a continuation of our previously reported in vitro screening of a library of small molecules characterized by a variety of scaffolds [19]. It focuses on newly developed triazolo–thiadiazole small molecules selected out of nearly 150 compounds [19], including 10 newly synthesized triazole amino thiol derivatives of substituted [1,2,4]triazolo[3,4- b][1,3,4]thiadiazoles. Our compound of choice {4-iodo-2-(3-(p-tolyl)-[1,2,4]triazolo[3,4- b][1,3,4]thiadiazol-6-yl)phenol = 4-MMI} [19] was found to efficiently inhibit heparanase enzymatic activity, cell invasion, experimental metastasis, and tumor growth in mouse models. To the best of our knowledge, this is the first report showing a marked decrease in primary tumor growth in mice treated with small molecules that inhibit heparanase enzymatic activity.

2. Materials and Methods All chemicals and solvents were purchased from Sigma-Aldrich (Bangalore, India). The completion of the reaction was monitored by pre-coated silica gel TLC plates. An Agi- lent mass spectrophotometer was used to record the mass of the synthesized compounds. 1H and 13C NMR were recorded on Agilent and Jeol NMR spectrophotometers (400 MHz). TMS was used as an internal standard, and DMSO was used as a solvent. Chemical shifts are expressed as ppm.

2.1. General Procedure for the Synthesis of 4-amino-5-substituted phenyl-4H-1,2,4- triazole-3-thiol (4a–e) A mixture of ethyl benzoate (1 mmol) in 15 mL of ethanol, as well as hydrazine hydrate (1 mmol), was added and refluxed for 5 h. Completion of the reaction was monitored by TLC followed by a water wash to remove traces of hydrazine hydrate, and acid hydrazide was obtained as a white solid. The acid hydrazide (1 mmol) was added to a stirred solution of ethanol (15 mL) containing KOH (2 mmol), and CS2 (1.3 mmol) was added and refluxed for 10 h. After completion of the reaction as monitored by TLC, diethyl ether was added and stirred for 1 h. The potassium salt of hydrazine carbothioate was obtained as a solid, filtered, and washed with diethyl ether. The above potassium salt (1 mmol) was dissolved in 15 mL of water, and hydrazine hydrate (2 mmol) was added and refluxed at 100 ◦C for 4 h, during which H2S evolved and the color of the reaction mass turned to green. Then, the reaction mass was cooled and acidified with concentrated HCl. The solid obtained Cancers 2021, 13, 2959 3 of 17

was recrystallized in ethanol to obtain pure triazole. The reaction progress was monitored using TLC.

2.2. General Procedure for the Synthesis of 6-substituted-(3-substituted phenyl-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazole) To a mixture of 4-amino-5-substituted phenyl-4H-1,2,4-triazole-3-thiol (1 mmol) and ◦ substituted salicylic acid (1 mmol), 6 mL of POCl3 was added and refluxed at 80 C for 8 h. The completion of the reaction was monitored by TLC. The reaction mass was quenched with crushed ice and neutralized by potassium carbonate to pH 8. The solid obtained was filtered, washed with water, and recrystallized with an appropriate solvent to obtain 6-substituted-(3-substituted-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazole).

2.3. 2,4-diiodo-6-(3-(4-nitrophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)phenol (4-NDI) Yellow solid. Yield: 70%. 1H NMR (DMSO, 400 MHz): δ 9.04 (s, 1H), 8.73 (s, 1H), 8.54–8.36 (m, 2H), 8.19 (s, 1H), 7.96 (s, 1H). 13C NMR (DMSO, 100 MHz): δ 164.14, 163.23, 150.27, 148.23, 138.30, 131.76, 131.15, 126.79, 125.24, 124.71, 120.15, 95.80, 90.81. LCMS: 590.8359 (m/z), 591.7344 [M+1]+.

2.4. 2-(3-(3-bromophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)-4,6-diiodophenol (3-BDI) Brown solid. Yield: 81%. 1H NMR (DMSO, 400 MHz): δ 8.41(s, 1H), 8.33 (d, J = 8 Hz, 1H), 8.09 (d, J = 1.6 Hz, 1H), 7.85 (d, J = 1.6 Hz, 1H), 7.69 (d, J = 7.6 Hz, 1H), 7.64–7.50 (m, 1H). 13C NMR (DMSO, 100 MHz): δ 166.49, 165.79, 150.41, 147.60 (2C), 134.22, 132.74, 131.99, 129.09, 128.33, 124.95, 122.76, 116.01, 98.33, 70.09. LCMS: 623.7613 (m/z), 624.5974 [M+1]+.

2.5. 2-(3-(3-chlorophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)-4,6-diiodophenol (3-CDI) Pale yellow solid. Yield: 75%. 1H NMR (DMSO, 400 MHz): δ 8.27(s, 2H), 7.76 (s, 1H), 7.68 (d, J = 7.6 Hz, 1H), 7.61 (d, J = 7.6 Hz, 1H), 7.56 (d, J = 7.6 Hz, 1H). 13C NMR (DMSO, 100 MHz): δ 168.66, 167.62, 159.54, 151.40, 149.52, 136.42, 134.36, 131.86, 131.75, 127.45, 126.80, 126.35, 116.0, 90.50, 82.69. LCMS: 579.8118 (m/z), 580.6468 [M+1]+.

2.6. 2,4-diiodo-6-(3-(4-chlorophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)phenol (4-CDI) Yellow solid. Yield: 78%. 1H NMR (DMSO, 400 MHz): δ 8.30 (d, J = 8.4 Hz, 2H), 8.23 (s, 1H), 8.06 (s, 1H), 7.68 (d, J = 8 Hz, 2H). 13C NMR (DMSO, 100 MHz): δ 163.50, 160.67, 148.17 (2C), 134.38 (2C), 129.24, 127.53, 127.32, 124.78, 117.98, 95.15, 80.83. LCMS: 579.8118 (m/z), 580.6678 [M+1]+.

2.7. 2,4-diiodo-6-(3-(p-tolyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)phenol (4-MDI) Yellow solid. Yield: 80%. 1H NMR (DMSO, 400 MHz): δ 8.18 (d, J = 8 Hz, 2H), 8.08 (s, 1H), 7.81 (s, 1H), 7.39 (d, J = 7.2 Hz, 2H), 2.36 (s, 3H). 13C NMR (DMSO, 100 MHz): δ 166.94, 165.46, 147.37, 139.75, 134.09, 130.20, 128.11, 126.43, 126.11, 124.28, 115.97, 98.61, 69.22, 21.65. LCMS: 559.8665 (m/z), 558.8661 [M-1]+.

2.8. 4-iodo-2-(3-(4-nitrophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)phenol (4-NMI) Off-white solid. Yield: 76%. 1H NMR (DMSO, 400 MHz): δ 9.00 (s, 1H), 8.62 (d, J = 7.6 Hz, 1H), 8.39–8.25 (m, 2H), 7.96–7.82 (m, 2H), 7.67 (d, J = 7.2 Hz, 1H). 13C NMR (DMSO, 100 MHz): δ 161.88, 161.08, 156.03, 148.04, 141.86, 134.76, 131.41, 130.97, 126.95, 124.30, 119.85, 119.26, 82.00. LCMS: 464.9393 (m/z), 465.9448 [M+1]+.

2.9. 2-(3-(3-bromophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)-4-iodophenol (3-BMI) Off-white solid. Yield: 84%. 1H NMR (DMSO, 400 MHz): δ 8.30 (d, J = 8.8 Hz, 2H), 8.20 (d, J = 7.6 Hz, 1H), 7.85 (d, J = 8 Hz, 1H), 7.70 (d, J = 8 Hz, 1H), 7.43 (d, J = 8.4 Hz, 1H), 7.61– 7.47 (m, 1H). 13C NMR (DMSO, 100 MHz): δ 160.85, 156.12, 150.53, 141.74, 135.21, 132.72, 131.24, 127.92, 127.47, 124.49, 122.31, 122.19, 121.12, 119.36, 87.18. LCMS: 497.8647 (m/z), 498.8651 [M+1]+. Cancers 2021, 13, 2959 4 of 17

2.10. 2-(3-(3-chlorophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)-4-iodophenol (3-CMI) Yellow solid. Yield: 81%. 1H NMR (DMSO, 400 MHz): δ 8.38–8.23 (m, 2H), 8.09 (d, J = 2.4 Hz, 1H), 7.75–7.59 (m, 1H), 7.58 (d, J = 7.2 Hz, 1H), 7.34–7.22 (m, 1H), 6.35 (d, J = 8.8 Hz, 1H). 13C NMR (DMSO, 100 MHz): δ 168.55, 167.57, 159.48, 151.38, 149.47, 136.31, 134.32, 131.87, 131.75, 130.28, 127.41, 126.69, 126.31, 115.85, 90.63. LCMS: 453.9152 (m/z), 454.9253 [M+1]+.

2.11. 2-(3-(4-chlorophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)-4-iodophenol (4-CMI) Yellow solid. Yield: 80%. 1H NMR (DMSO, 400 MHz): δ 8.27 (d, J = 8.4 Hz, 1H), 7.98 (d, J = 2.4 Hz, 1H), 7.77 (d, J = 8.8 Hz, 1H), 7.75–7.63 (m, 2H), 6.83 (d, J = 8.8 Hz, 1H), 7.55 (d, J = 8.4 Hz, 1H). 13C NMR (DMSO, 100 MHz): δ 168.43, 164.73, 157.79, 150.08, 142.76, 138.69, 136.34, 129.66, 128.07, 124.24, 120.28, 81.56. LCMS: 453.9152 (m/z), 454.9253 [M+1]+.

2.12. 4-iodo-2-(3-(p-tolyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl)phenol (4-MMI) Yellow solid. Yield: 84%. 1H NMR (DMSO, 400 MHz): δ 8.22 (s, 1H), 8.19–8.00 (m, 2H), 7.71 (d, J = 9.2 Hz, 1H), 7.52 (d, J = 8.8 Hz, 1H), 7.46–7.29 (m, 2H), 2.35 (s, 3H). 13C NMR (DMSO, 100 MHz): δ 161.14, 158.33, 150.56, 148.22, 140.24, 135.20, 130.15, 130.05, 126.26, 126.19, 123.28, 121.42, 84.01, 21.51. LCMS: 433.9698 (m/z), 434.8964 [M+1]+.

2.13. Cells and Cell Culture U87 human glioma cells were purchased from the American Type Culture Collection. (ATCC). MPC-11 mouse myeloma cells were kindly provided by Dr. Ralph Sanderson (the University of Alabama at Birmingham, Birmingham, AL, USA) [20] and luciferase- labeled 4T1 mouse breast carcinoma cells were kindly provided by Dr. Yuval Shaked (TICC, Technion, Haifa, Israel) [21]. U87 and 4T1cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM) and the MPC-11 cells in RPMI medium, supplemented with glutamine, pyruvate, antibiotics, and 10% fetal calf serum (Biological Industries, Beit Haemek, Israel) ◦ in a humidified atmosphere containing 5% CO2 at 37 C.

2.14. Colorimetric Heparanase Assay The assay was carried out in a 96-well plate in which the formation of the disaccharide product upon cleavage of fondaparinux (Arixtra, GlaxoSmithKline, Brentford, UK) was estimated using WST-1 tetrazolium salt as described earlier [22,23]. In brief, the reaction system (100 µL) comprised sodium acetate buffer (pH 5.0, 40 mM) and fondaparinux (100 mM) with or without different doses of chemicals under investigation. The assay was initiated by the addition of recombinant heparanase to make the final concentration of 140 pM. The reaction vessels were placed at 37 ◦C for 18 h, followed by the addition of stop solution (100 µL). WST-1 prepared in 0.1 M NaOH served as a stop solution. Thereafter, the plates were incubated at 60 ◦C for 60 min, and absorbance was measured at 584 nm. N-(4-([4-(1H-Benzoimidazol-2-yl)-arylamino]-methyl)-phenyl)-benzamide (compound #98) was used as a reference compound. The title compounds were examined for their inhibitory efficacy towards heparanase at 10 and 50 µg/mL. Compounds that yielded inhibition at these concentrations were further tested at lower concentrations to determine their IC50.

2.15. ECM Degradation Heparanase Assay ECM degradation heparanase assay was carried out as reported earlier [24–26]. Briefly, ECM labeled with sulfate [35S] [25] and coating the surface of 35-mm cultures plates was treated with heparanase (200 ng/mL) with or without compounds under investigation. The medium was collected to examine the cleavage of HS proteoglycans and subjected to molecular sieving on Sepharose 6B columns. The radioactivity was measured in the eluted fractions (0.2 mL). Blue dextran and phenol red were used to mark the excluded volume (Vo) and included volume (Vt), respectively. The cleaved HS chains were eluted at 0.5 < Kav < 0.8 (fractions 16–32), and the nearly intact was eluted with the Cancers 2021, 13, 2959 5 of 17

void volume [24]. Each experiment was carried out thrice and variation in elution positions did not exceed ±15%.

2.16. Cell Invasion Invasion assay was performed using Boyden chambers as reported previously [27,28]. Briefly, U87 cells (5 × 105) were seeded into the upper chamber of the invasion chamber in a medium devoid of serum, and the lower chamber contained media supplemented with serum. The cell invasion on treatment with title compounds (4-MMI, 4-CMI; 10 mg/mL) was analyzed by staining with 0.5% crystal violet (Sigma-Aldrich) and photographed.

2.17. Experimental Metastasis 4TI mouse breast cancer cells tagged with luciferase (1.5 × 105/0.1 mL/BALB/c mouse) were inoculated intravenously [21,29]. Test compounds (i.p., 500 µg/mouse) or DMSO (0.1 mL/mouse) were administered 20 min before cell injection. The mice were analyzed using IVIS bioluminescent on day 7 and day 13 post-inoculation. These cells metastasize primarily to the lungs.

2.18. IVIS Imaging The bioluminescence imaging of tumors generated from luciferase-expressing can- cer cells was carried out using a highly sensitive, cooled charge-coupled device camera assembled in a specimen box (IVIS; Xenogen Corp, Hopkinton, MA, USA). The imaging has advantages such as real-time capture, non-invasive, and quantitative values. For this, the tumor-bearing mice were intraperitoneally administered with a luciferase substrate D-luciferin at a dose of 150 mg/kg body weight. The experimental animal was anesthetized and kept in a camera box followed by continuous exposure to isoflurane (EZAnesthesia, Palmer, PA, USA). The bioluminescence from the tumor mass was detected by the IVIS system, and, subsequently, the tumor burden was quantified using Living Image software (Xenogen) [30,31].

2.19. MPC-11 Tumor Growth The mouse myeloma (MPC-11) cells were gently washed with PBS and the cell number was adjusted to 5 × 106 cells/mL. Thereafter, six-week-old female BALB/c mice (n = 6) were subcutaneously injected with washed MPC-11 cells (5 × 105/0.1 mL). The experi- mental animals were treated with either DMSO alone (0.1 mL/mouse) or 4-MMI (daily 200 µg/mouse, i.p.) starting from day 2 after the implantation of tumor cells, for 12 days. The tumor size was measured using calipers on day 8 and day 12. The tumor volume (V) was determined using the equation V = L × W2 × 0.52, where L is the length and W is the width of the tumor. Lastly, the mice were euthanized and xenograft tumors were removed, weighed, and fixed using formalin for further studies [32].

2.20. Statistics Data are presented as means ± SE. Statistical significance was analyzed by the 2- tailed Student’s t-test. Values of p < 0.05 were considered significant. Data sets passed D’Agostino–Pearson normality (GraphPad Prism 5 utility software, San Diego, CA, USA). All experiments were repeated at least twice with similar results.

3. Results 3.1. Synthesis of Title Compounds The synthesis of triazolo–thiadiazoles was initiated by the conversion of substituted ethyl benzoates (Figure1A (1a–e)) to their corresponding hydrazides (2a–e). The esters (1a–e) were refluxed with hydrazine hydrate in ethanol, which resulted in the formation of hydrazides (2a–e) as the intermediates. Compounds 2a–e were treated with carbon disulphide under basic conditions using potassium hydroxide resulted in the formation of potassium salts (3a–e). The potassium salts were further treated with hydrazine to generate CancersCancersCancers 2021 2021 2021, 13, ,13 13, x, , x x 6 66of of of18 1818

2.20.2.20.2.20. St St Statisticsatisticsatistics DataDataData are are are presented presented presented as as as means means means ± ± SE.± SE. SE. Statistical Statistical Statistical significance significance significance was was was analyzed analyzed analyzed by by by the the the 2 2- 2tailed--tailedtailed Student’sStudent’sStudent’s t - ttest.t--test.test. Values ValuesValues of ofof p pp< <<0 .0500.05.05 were werewere considered consideredconsidered significant. significant.significant. Data DataData sets setssets passed passedpassed D DD’Ago-’’Ago-Ago- stinostinostino–Pearson––PearsonPearson normality normalitynormality ( GraphPad ((GraphPadGraphPad Prism PrismPrism 5 55utility utilityutility software softwaresoftware, ,San , SanSan Diego, Diego,Diego, CA CACA, ,USA , USAUSA).) ).All . AllAll experimentsexperimentsexperiments were were were repeated repeated repeated at at at least least least twice twice twice with with with similar similar similar results. results. results.

3.3. 3.Results Results Results 3.1.3.1.3.1. Synthesis Synthesis Synthesis of of ofTitle Title Title Compounds Compounds Compounds TheTheThe synthesis synthesissynthesis of ofof triazolo triazolotriazolo––thiadiazoles–thiadiazolesthiadiazoles was waswas initiated initiatedinitiated by byby the thethe conversion conversionconversion of ofof substituted substitutedsubstituted ethylethylethyl benzoates benzoatesbenzoates ( Figure ((FigureFigure 1A 1A1A ( 1a ((1a1a––e–)ee)) ))to) toto their theirtheir corr corrcorrespondingespondingesponding hydrazides hydrazideshydrazides ( 2a ((2a2a–e––)ee.) ).The . TheThe esters estersesters (1a((1a1a––e–)ee )were) werewere refluxed refluxedrefluxed with withwith hydrazine hydrazinehydrazine hydrate hydratehydrate in inin ethanol ethanolethanol, ,which , whichwhich resulted resultedresulted in inin the thethe formation formationformation ofofof hydrazides hydrazideshydrazides ( 2a ((2a2a––e–)ee )as) asas the thethe intermediates. intermediates.intermediates. Compounds CompoundsCompounds 2a 2a2a––e– eweree werewere treated treatedtreated with withwith carbon carboncarbon di- di-di- sulphidesulphidesulphide under underunder basic basicbasic conditions conditionsconditions using usingusing potas potaspotassiumsiumsium hydroxide hydroxidehydroxide resulted resultedresulted in inin the thethe formation formationformation of ofof potassiumpotassiumpotassium salts salts salts ( 3a( (3a3a–e––)ee.) )The. .The The potassium potassium potassium salts salts salts were were were further further further treated treated treated with with with hydrazine hydrazine hydrazine to to to gener- gener- gener- ateateate 4 4- 4amino--aminoamino-5---55substituted--substitutedsubstituted phenyl phenyl phenyl-4H--4H4H-1,2,4--1,2,41,2,4-triazole--triazoletriazole-3--3-3thiols--thiolsthiols ( 4a( (4a4a–e––)ee.) )C. .C ompoundsCompoundsompounds 4a 4a 4a––e– eweree were were reactedreactedreacted with withwith substituted substituted substituted salicylic salicylicsalicylic a acidacidcid in inin the thethe presence presencepresence of ofof phosphorous phosphorousphosphorous oxychloride oxychlorideoxychloride under underunder refluxingrefluxingrefluxing conditions conditions conditions to to to obtain obtain obtain triazolo triazolo triazolo––thiadiazoles–thiadiazolesthiadiazoles, as, ,as as shown shown shown in in in Figure Figure Figure 1 1B 1B.B The. .The The structures structures structures ofofof all allall the thethe target targettarget compounds compoundscompounds presented presentedpresented in inin Table TableTable 1 11were werewere characterized characterizedcharacterized by byby LCMS, LCMS,LCMS, 1 H 11HH NMR, NMR,NMR, andandand 13 13 C13C CNMR NMR NMR spectrometry. spectrometry. spectrometry.

Cancers 2021, 13, 2959 6 of 17

4-amino-5-substituted phenyl-4H-1,2,4-triazole-3-thiols (4a–e). Compounds 4a–e were reacted with substituted salicylic acid in the presence of phosphorous oxychloride under refluxing conditions to obtain triazolo–thiadiazoles, as shown in Figure1B. The structures FigureFigureFigure 1. 1. 1.( A( (A)A )Synthesis) Synthesis Synthesis of of of 4 -4 amino4--aminoamino-5--5substituted5--substitutedsubstituted phenyl phenyl phenyl-4H--4H4H-1,2,4--1,2,41,2,4-triazole--triazoletriazole-3--3thiol.3--thiol.thiol. ( B( (B)B )Synthesis) Synthesis Synthesis1 of of of substitutedsubstitutedof all the target[1,2,4]triazolo[3,4 [1,2,4]triazolo[3,4 compounds--b]b] presented[1,3,4]thiadiazole [1,3,4]thiadiazole in Table (w (w1herehere were R1 R1 characterized= = 4 4--NO2,NO2, 3 3--Br,Br, 3 3 by--Cl,Cl, LCMS, 4 4--Cl,Cl, 4 4--CH3CH3H NMR,and and substituted13 [1,2,4]triazolo[3,4-b] [1,3,4]thiadiazole (where R1 = 4-NO2, 3-Br, 3-Cl, 4-Cl, 4-CH3 and R2R2R2and = = 3,5= 3,5 3,5-diiodoC--diiododiiodo NMR ( 5a( (5a spectrometry.5a) )and) and and 5 -5 iodo5--iodoiodo ( 5b( (5b5b))).) ). .

TableTableTable 1. 1. 1.1.List List List of of ofsynthesized synthesized synthesized substituted substituted substituted [1,2,4]triazolo[3,4 [1,2,4]triazolo[3,4 [1,2,4]triazolo[3,4-b][1,2,4]triazolo[3,4-b]--b]b] [1,3,4]thiadiazoles. [1,3,4]thiadiazoles. [1,3,4]thiadiazoles.[1,3,4]thiadiazoles.

TriazoleTriazoleTriazole Amino AminoAmino Amino Thiol Thiol Thiol AcidAcidAcidAcid ProductsProducts Products DerivativesDerivativesDerivatives

CancersCancersCancers 2021 20212021, ,13, 1313, ,x, x x 77 7 of ofof 18 1818 CancersCancersCancers 2021 2021 2021, ,13 ,13 13, ,x ,x x 77 7 of ofof 18 1818 Cancers Cancers Cancers 2021 2021 2021,, , 13 ,13, 13,, , x x,x, x x 5a5a5a 77 7 of of of 18 1818 4a4a4a4a 5a 44-4-NDI4NDI--NDINDI

4b4b 5a5a 4b4b4b 5a5a5a 4b4b4b 5a5a5a

333-33-BDI-BDI--BDIBDIBDI 333--3-BDI-BDI-BDIBDI

4c4c 5a5a 4c4c4c 5a5a5a 4c4c4c 5a5a5a

333-33-CDI-CDI--CDICDICDI 333--3-CDI-CDI-CDICDI

5a5a 4d4d4d 5a5a5a 4d4d4d 5a5a5a5a 4d 444-4-CDI--CDICDICDI 444--44-CDI-CDI-CDI-CDICDI

4e4e 5a5a 4e4e4e 5a5a5a 4e4e4e 5a5a5a5a

444-4-MDI--MDIMDIMDI 444--4-4-MDIMDI-MDI-MDIMDI

4a4a4a 5b5b5b5b 4a4a4a4a 5b5b5b5b 4a4a 444-4-NMI--NMINMINMI 444--4-NMI-NMI-NMINMI

4b4b4b4b 5b5b5b5b 4b4b4b4b 5b5b5b5b

333-3-BMI--BMIBMIBMI 333--3-BMI-BMI-BMIBMI

CancersCancersCancers 2021 2021 2021, 13, ,13 13, x, ,x x 77 7of of of18 1818 Cancers Cancers Cancers 2021 20212021, ,13, 1313, ,x, x x 77 7 of ofof 18 1818

4b4b4b 5a5a5a 4b4b4b 5a5a5a

3-BDI 33-33-BDI-BDI-BDIBDI

4c4c4c 5a5a5a 4c4c4c 5a5a5a

3-CDI 33-33-CDI-CDI-CDICDI

5a5a5a 4d4d4d 5a5a5a 4d4d 4-CDI 44-44-CDI-CDI-CDICDI

Cancers 2021, 13, 2959 7 of 17

Table 1. Cont.

4e4e4e 5a5a5a Triazole Amino4e4e4e Thiol 5a5a5a Acid Products Derivatives 4-MDI 44-44-MDI-MDI-MDIMDI

5b5b 4a4a4a4a 5b5b5b5b 4a4a4a 4-NMI 44-44-NMI4-NMI-NMI-NMINMI

CancersCancersCancers 2021 2021 2021,, , 13 ,,13 13,, , x ,x,x x x 88 8 of ofof 18 1818 Cancers Cancers Cancers 2021 2021 2021, ,13 ,13 13, ,x ,x x 4b4b4b 5b5b5b 88 8 of ofof 18 1818 4b4b4b 5b5b5b

3-BMI 33-33-BMI3-BMI-BMI-BMIBMI

4c4c4c 5b5b5b 4c4c4c 5b5b5b 4c4c 5b5b 333-3-CMI-CMI-CMICMI 333-3-CMI-CMI-CMICMI

5b5b5b 4d4d4d 5b5b5b 4d4d4d4d 5b5b5b 444--CMI-CMICMI 444-44-CMI-CMI-CMI-CMICMI

4e4e4e 5b5b5b 4e4e4e4e 5b5b5b5b 5b 444--MMI-MMIMMI 444-44-MMI-MMI--MMIMMIMMI 3.2.3.2.3.2. NewlyNewly Newly SynthesizedSynthesized Synthesized TriazoloTriazolo Triazolo–––ThiadiazolesThiadiazolesThiadiazoles InhibitInhibit Inhibit HeparanaseHeparanase Heparanase EnzymaticEnzymatic Enzymatic ActivityActivity Activity 3.2.3.2.3.2.3.2. Newly Newly NewlyNewly Synthesized Synthesized SynthesizedSynthesized Triazolo Triazolo TriazoloTriazolo–––Thiadiazoles–ThiadiazolesThiadiazolesThiadiazoles Inhibit Inhibit InhibitInhibit Heparanase Heparanase HeparanaseHeparanase Enzymatic Enzymatic EnzymaticEnzymatic ActivityActivity ActivityActivity TheTheThe newly newly newly synthesized synthesized synthesized compounds compounds compounds presented presented presented in inin Table Table Table 1 1 1 were werewere examined examined examined for for for their theirtheir TheTheTheThe newly newly newly newly synthesized synthesized synthesized synthesized compounds compounds compounds compounds presented presented presented presented in ininin Table Table Table Table 1 1 1 1 were werewerewere examined examined examined examined for for for for their theirtheirtheir abilityabilityability toto to inhibitinhibit inhibit thethe the enzymaticenzymatic enzymatic activityactivity activity ofof of recombinantrecombinant recombinant heparanase.heparanase. heparanase. ForFor For thethe the initialinitial initial screen-screen- screen- abilityabilityabilityability to to toto inhibit inhibit inhibitinhibit the the thethe enzymatic enzymatic enzymaticenzymatic activity activity activityactivity of of ofof recombinant recombinant recombinantrecombinant heparanase. heparanase. heparanase.heparanase. For For ForFor the the thethe initial initial initialinitial screen- screen- screen-screen- inginging ofof of compoundscompounds compounds,, we,we we appliedapplied applied aa a colorimetriccolorimetric colorimetric assayassay assay basedbased based onon on thethe the cleavagecleavage cleavage ofof of thethe the syntheticsynthetic synthetic inginginging of of ofof compounds compounds compoundscompounds,, ,we,we wewe appliedapplied appliedapplied aa a a colorimetric colorimetric colorimetriccolorimetric assayassay assayassay basedbased basedbased onon onon thethe thethe cleavagecleavage cleavagecleavage ofof ofof the the thethe syntheticsynthetic syntheticsynthetic heparinheparinheparin pentasaccharidepentasaccharide pentasaccharide fondaparinuxfondaparinux fondaparinux (( Arixtra(ArixtraArixtra)) ) [[ 23[2323]].]. The.The The assayassay assay measuresmeasures measures thethe the appearanceappearance appearance heparinheparinheparinheparin pentasaccharidepentasaccharide pentasaccharidepentasaccharide fondaparinuxfondaparinux fondaparinuxfondaparinux (( Arixtra(Arixtra(ArixtraArixtra)) ) )[[ 23[23[2323]].].] .The.The TheThe assayassay assayassay measuresmeasures measuresmeasures thethe thethe appearanceappearance appearanceappearance ofofof a aa disaccharide disaccharidedisaccharide product productproduct of ofof heparanase heparanaseheparanase--catalyzed-catalyzedcatalyzed fondaparinux fondaparinuxfondaparinux cleavage, cleavage,cleavage, using usingusing the thethe te- te-te- ofofofof a a a a disaccharide disaccharide disaccharidedisaccharide product product productproduct of of ofof heparanase heparanase heparanaseheparanase--catalyzed-catalyzed-catalyzedcatalyzed fondaparinux fondaparinux fondaparinuxfondaparinux cleavage, cleavage, cleavage,cleavage, using using usingusing the the thethe te- te- te-te- trazoliumtrazoliumtrazolium saltsalt salt WSTWST WST--1-11 [[ 23[2323]].]. All.All All compoundscompounds compounds werewere were firstfirst first testedtested tested forfor for inhibitioninhibition inhibition ofof of heparanaseheparanase heparanase atat at trazoliumtrazoliumtrazoliumtrazolium saltsalt saltsalt WSTWST WSTWST--1-1-1 1 [[ 23[23[2323]].].] . All.All AllAll compoundscompounds compoundscompounds werewere werewere firstfirst firstfirst testedtested testedtested forfor forfor inhibitioninhibition inhibitioninhibition ofof ofof heparanaseheparanase heparanaseheparanase atat atat 101010 andand and 50 5050 μg μgμg/mL/mL/mL ( (not(notnot shown shownshown)).). .Compounds CompoundsCompounds that thatthat yielded yieldedyielded inhibition inhibitioninhibition at atat these thesethese high highhigh concen- concen-concen- 10101010 and and andand 50 50 5050 μg μg μgμg/mL/mL/mL/mL ( ( not(not(notnot shown shown shownshown)).).) . .Compounds Compounds CompoundsCompounds that that thatthat yielded yielded yieldedyielded inhibition inhibition inhibitioninhibition at at atat these these thesethese high high highhigh concen- concen- concen-concen- trationstrationstrations were werewere furtherfurther further examinedexamined examined at atat lowerlower lower concentrationsconcentrations concentrations to toto determine determinedetermine their theirtheir IC ICIC50505050 ( (Figure(FigureFigure 22 2))..). . trationstrationstrations werewere were furtherfurther further examinedexamined examined atat at lowerlower lower concentrationsconcentrations concentrations toto to determinedetermine determine theirtheir their ICIC IC505050 (( Figure(FigureFigure 22 )2).). . AmongAmongAmong these these these compounds, compounds, compounds, 2 2 2--(-(3(33--(-(4(4-4-chloro-chlorochlorophenylphenylphenyl))-)-[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4[1,2,4]triazolo[3,4--b][1,3,4]thiadiazol-b][1,3,4]thiadiazolb][1,3,4]thiadiazol--6-66--- AmongAmongAmongAmong these these these these compounds, compounds, compounds, compounds, 2 2 2- 2-(-(-3(3(3-3-(-(-4(4(4-4-chloro-chloro-chlorochlorophenylphenylphenylphenyl))-)-)[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4[1,2,4]triazolo[3,4--b][1,3,4]thiadiazol-b][1,3,4]thiadiazol-b][1,3,4]thiadiazolb][1,3,4]thiadiazol--6-6-6-6--- ylylyl))-)-4-4-4-iodophenol-iodophenoliodophenol ((4(4-4-CMI-CMICMI)),), , 2,4 2,4 2,4--diiodo-diiododiiodo--6-6-6-(-(3(33--(-(p(pp--tolyl-tolyltolyl))-)-[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4[1,2,4]triazolo[3,4--b][1,3,4]thiadiazol-b][1,3,4]thiadiazolb][1,3,4]thiadiazol--- ylylylyl))-)-)4-4-4-4-iodophenol-iodophenol-iodophenoliodophenol ( (4(4(4-4-CMI-CMI-CMICMI)),),) ,, 2,4 2,4 2,4 2,4--diiodo-diiodo-diiododiiodo--6-6-6-6-(-(-3(3(3-3-(-(-p(p(pp--tolyl-tolyl-tolyltolyl))-)-)[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4[1,2,4]triazolo[3,4--b][1,3,4]thiadiazol-b][1,3,4]thiadiazol-b][1,3,4]thiadiazolb][1,3,4]thiadiazol---- 666--yl-ylyl))phenol)phenolphenol ((4(4-4-MDI-MDIMDI)),), , and and and 4 4 4--iodo-iodoiodo--2-22--(-(3(33--(-(p(pp--tolyl-tolyltolyl))-)-[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4[1,2,4]triazolo[3,4--b][1,3,4]thiadiazol-b][1,3,4]thiadiazolb][1,3,4]thiadiazol--6-66--- 666-6-yl-yl-ylyl))phenol)phenol)phenolphenol ((4(4(4-4-MDI-MDI-MDIMDI)),),) , , and and and and 4 4 4- 4-iodo-iodo-iodoiodo--2-2-2-2-(-(-3(3(3-3-(-(-p(p(pp--tolyl-tolyl-tolyltolyl))-)-)[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4-[1,2,4]triazolo[3,4[1,2,4]triazolo[3,4--b][1,3,4]thiadiazol-b][1,3,4]thiadiazol-b][1,3,4]thiadiazolb][1,3,4]thiadiazol--6-6-6-6--- ylylyl))phenol)phenolphenol (( 4(44--MMI-MMIMMI)) ) showedshowed showed goodgood good heparanaseheparanase heparanase inhibitoryinhibitory inhibitory activityactivity activity withwith with ICIC IC50505050 valuesvalues values ofof of 3,3, 3, 12.5,12.5, 12.5, ylylyl))phenol)phenolphenol ( ( 4(4-4-MMI-MMIMMI)) )showed showed showed goodgood good heparanaseheparanase heparanase inhibitory inhibitory inhibitory activityactivity activity withwith with ICIC IC505050 valuesvalues values ofof of 3, 3, 3, 12.5,12.5, 12.5, andandand 3.13.1 3.1 µg/mL,µg/mL, µg/mL, respectively. respectively.respectively. CompoundCompound Compound #98,#98, #98, usedused used asas as aa a positive positivepositive control,control, control, displayeddisplayed displayed anan an ICIC IC50505050 andandand 3.13.1 3.1 µg/mL,µg/mL, µg/mL, respectively.respectively. respectively. CompoundCompound Compound #98,#98, #98, usedused used asas as aa a positivepositive positive control,control, control, displayeddisplayed displayed anan an ICIC IC505050 valuevaluevalue ofof of 0.70.7 0.7 µg/mLµg/mL µg/mL (( Figure(FigureFigure 2,2, 2, bottombottom bottom tabletable table)).). . valuevaluevaluevalue of of ofof 0.7 0.7 0.70.7 µg/mL µg/mL µg/mLµg/mL ( ( Figure(Figure(FigureFigure 2, 2, 2,2, bottom bottom bottombottom table table tabletable)).).) ..

Cancers 2021, 13, x 6 of 18

2.20. Statistics Data are presented as means ± SE. Statistical significance was analyzed by the 2-tailed Student’s t-test. Values of p < 0.05 were considered significant. Data sets passed D’Ago- stino–Pearson normality (GraphPad Prism 5 utility software, San Diego, CA, USA). All experiments were repeated at least twice with similar results.

3. Results 3.1. Synthesis of Title Compounds The synthesis of triazolo–thiadiazoles was initiated by the conversion of substituted ethyl benzoates (Figure 1A (1a–e)) to their corresponding hydrazides (2a–e). The esters (1a–e) were refluxed with hydrazine hydrate in ethanol, which resulted in the formation of hydrazides (2a–e) as the intermediates. Compounds 2a–e were treated with carbon di- sulphide under basic conditions using potassium hydroxide resulted in the formation of potassium salts (3a–e). The potassium salts were further treated with hydrazine to gener- ate 4-amino-5-substituted phenyl-4H-1,2,4-triazole-3-thiols (4a–e). Compounds 4a–e were reacted with substituted salicylic acid in the presence of phosphorous oxychloride under refluxing conditions to obtain triazolo–thiadiazoles, as shown in Figure 1B. The structures Cancers 2021,of13 ,all 2959 the target compounds presented in Table 1 were characterized by LCMS, 1H NMR, 8 of 17 and 13C NMR spectrometry.

FigureFigure 1. (A) Synthesis1. (A) Synthesis of 4-amino-5-substituted of 4-amino-5-substituted phenyl-4H-1,2,4-triazole-3-thiol. phenyl-4H-1,2,4-triazole (B) Synthesis-3-thiol. of ( substitutedB) Synthesis [1,2,4]triazolo of [1,3,4]thiadiazolesubstituted (where [1,2,4]triazolo[3,4 R1 = 4-NO2,- 3-Br,b] [1,3,4]thiadiazole 3-Cl, 4-Cl, 4-CH3 and(where R2 = R1 3,5-diiodo = 4-NO2, (5a) 3- andBr, 3 5-iodo-Cl, 4- (5b)).Cl, 4-CH3 and R2 = 3,5-diiodo (5a) and 5-iodo (5b)). 3.2. Newly Synthesized Triazolo–Thiadiazoles Inhibit Heparanase Enzymatic Activity Table 1. List of synthesizedThe substituted newly synthesized [1,2,4]triazolo[3,4 compounds-b] [1,3,4]thiadiazoles. presented in Table 1 were examined for their ability to inhibit the enzymatic activity of recombinant heparanase. For the initial screening Triazole Amino Thiol of compounds, we appliedAcid a colorimetric assay basedProducts on the cleavage of the synthetic Derivativesheparin pentasaccharide fondaparinux (Arixtra) [23]. The assay measures the appearance of a disaccharide product of heparanase-catalyzed fondaparinux cleavage, using the tetra- zolium salt WST-1 [23]. All compounds were first tested for inhibition of heparanase at 10 and 50 µg/mL (not shown). Compounds that yielded inhibition at these high concen- trations were further examined at lower concentrations to determine their IC50 (Figure2). Among these compounds, 2-(3-(4-chlorophenyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6- yl)-4-iodophenol (4-CMI), 2,4-diiodo-6-(3-(p-tolyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol- 6-yl)phenol (4-MDI), and 4-iodo-2-(3-(p-tolyl)-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6-yl) 4a 5a phenol (4-MMI) showed good heparanase inhibitory activity with IC50 values of 3, 12.5, 4-NDI and 3.1 µg/mL, respectively. Compound #98, used as a positive control, displayed an IC50 value of 0.7 µg/mL (Figure2, bottom table). To validate the results and better resemble the in vivo situation, we applied metaboli- 35 cally sulfate [Na2 SO4] labeled extracellular matrix (ECM) deposited by cultured endothe- lial cells. This naturally produced substrate closely resembles the subendothelial basement membrane in its composition, biological function, and barrier properties [25]. Years of experience indicate that compounds that effectively inhibit the in this assay are also effective in preclinical animal models [9]. This semi-quantitative assay measures release of radioactive heparan sulfate (HS) degradation fragments from an insoluble extracellular matrix (ECM) that is firmly bound to a culture dish [26]. As demonstrated in Figure3A, compounds 4-CMI, 4-MMI, and 4-MDI inhibited (50–70%) heparanase activity at 25 µg/mL, but there was little or no inhibition at 5 µg/mL. Cancers 2021, 13, 2959 9 of 17 Cancers 2021, 13, x 9 of 18

Figure 2. Figure 2.Screening Screening ofof compoundscompounds for inhibition inhibition of of hepa heparanaseranase enzymatic enzymatic activity activity applying applying the the fondaparinuxfondaparinux (Arixtra) (Arixtra heparanase) heparanase assay. assay. Compound Compound #98 #98 (red) (red =) N-(4-phenyl)-benzamide = N-(4-phenyl)-benzamide = positive = posi- control.tive control. Newly Newly synthesized synthesized compounds compounds were were first first tested tested for for inhibition inhibition of heparanaseof heparanase at at 10 10 and 50andµg/mL 50 μg (not/mL shown).(not shown Compounds). Compounds that that yielded yielded inhibition inhibition at these at these concentrations concentrations were were further fur- testedther tested at lower at lower concentrations concentrations to determine to determine their their IC50 IC. Several50. Several compounds compounds (i.e., (i.e., 4-CMI; 4-CMI; 4-MMI; 4-MMI; 4-MDI;4-MDI; IC IC5050= = 3,3, 3.1, 12.5 μg/mLµg/mL,, respectively respectively)) inhibited inhibited the the enzyme enzyme better better than than 2,4 2,4-Diiodo-6-(3--Diiodo-6-(3- phenyl-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6yl)phenol (DTP; IC50 = 30 g/mL) (bottom table). phenyl-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazol-6yl)phenol (DTP; IC50 = 30 µg/mL) (bottom table).

3.3. CompoundsTo validate 4-CMI the results and 4-MMI and better Inhibit resemble Glioma Cell the Invasionin vivo situation, we applied metabol- icallyThe sulfate causal [Na involvement235SO4] labeled of heparanase extracellular in matrix cancer (ECM metastasis) deposited has long by cultured been demon- endo- stratedthelial incells. various This naturally types of cancer produced [12, 13substrate,24]. Applying closely resembles Matrigel-coated the subendothelial transwell filters, base- wement investigated membrane the in effectits composition, of 4-CMI and biological 4-MMI function, on human and U87 barrier glioma properties cell invasion. [25]. Years As demonstratedof experience inindicate Figure that3B, compounds treatment with that 4-CMIeffectively or 4-MMI inhibit (10the enzymeµg/mL) in significantly this assay are inhibitedalso effective the invasion in preclinical capacity animal of U87 models cells. A [9 similar]. This resultsemi-quantitative was obtained assay with measures compound re- 4-MDIlease of (not radioactive shown). A heparan dose–response sulfate effect(HS) degradation of compound fragments 4-MMI is from presented an insoluble in Figure extra-3C. cellular matrix (ECM) that is firmly bound to a culture dish [26]. As demonstrated in Fig- ure 3A, compounds 4-CMI, 4-MMI, and 4-MDI inhibited (50–70%) heparanase activity at 25 µg/mL, but there was little or no inhibition at 5 µg/mL.

CancersCancers2021 2021,, 1313,, 2959x 10 of 18 10 of 17

Figure 3. Inhibition of ECM degradation and cell invasion. (A) ECM degradation. Active heparanase (200 ng) was incubated for 5 h without (red) or with 5 and 25 µg/mL of 4-CMI, 4-MDI, or 4-MMI on dishes coated with sulfate-labeled ECM. Labeled material released from the ECM was subjected to gel filtration (Sepharose 6B). Heparan sulfate degradation fragments are typically eluted in fractions Cancers 2021, 13, 2959 11 of 17

18–30. (B) Cell invasion. U87 human glioma cells (2 × 105) were plated onto Matrigel-coated 8 µm transwell filters and incubated (6 h, 37 ◦C) in the absence (Con) or presence of 4-MMI or 4-CMI (10 µg/mL). (C) Dose–response showing the effect of compound 4-MMI (2.5, 10, and 40 µg/mL) on U87 cell invasion. Invading cells adhering to the lower side of the membrane were visualized (top panels) and quantified (bottom panel) in 7 random fields as described in ‘Materials and Methods’. The structures of compounds 4-CMI and 4-MMI are presented in Table1.

3.4. Compounds 4-CMI and 4-MMI Mitigate Breast Carcinoma Experimental Metastasis We evaluated the effect of 4-CMI and 4-MMI on 4T1 mouse breast carcinoma exper- imental metastasis. Briefly, luciferase-labeled 4T1 breast carcinoma cells were injected i.v. into BALB/c mice. Compound 4-CMI or 4-MMI was injected (i.p.) 20 min before cell inoculation, and metastases formation was inspected and quantified by IVIS fluores- cence imaging. As demonstrated in Figure4 and Figure S1, compound 4-MMI and, to a lesser extent, compound 4-CMI inhibited the extravasation of 4T1 cells and their sub- sequent colonization in the mouse lungs. The effect of 4-MMI was comparable to that exerted by Roneparstat (=SST), a well-characterized N-acetylated, glycol-split heparin [17] that recently completed phase I clinical trial in myeloma patients [33]. These results in- dicate that triazolo–thiadiazoles attenuate the ability of blood-borne carcinoma cells to Cancers 2021, 13, x 12 of 18 extravasate through the subendothelial basement membrane, attributed to their ability to inhibit heparanase enzymatic activity, ECM degradation, and cell invasion.

FigureFigure 4. 4. InhibitionInhibition of 4T1 of breast 4T1 breastcarcinoma carcinoma experimental experimental metastasis. Luciferase metastasis.-labeled Luciferase-labeled 4T1 breast 4T1 breast carcinomacarcinoma cells cells (1.5×10 (1.55/mouse× 105)/mouse) were injected were i.v. injectedinto BALB/c i.v. mice into (n BALB/c= 6 BALB/cmice mice/group (n = 6). BALB/c mice/group). Vehicle (DMSO alone), positive control (150 µ g/mouse, SST), 4-CMI, or 4-MMI (500 μg/mouse) Vehiclewere injected (DMSO (i.p.; alone),0.1 mL/mouse positive) 20 min control prior to (150 cellµ inoculation.g/mouse, Metastasis SST), 4-CMI, was inspected or 4-MMI by (500 µg/mouse) were injectedIVIS on day (i.p.;s 7 (not 0.1 shown mL/mouse)) and 13 after 20 mincell inoculation. prior to cell Quantification inoculation. of the Metastasis luciferase intensities was inspected by IVIS on daysis shown 7 (not graphically shown) in andthe lower 13 after panel, cell as described inoculation. in Section Quantification 2 (Materials and of Methods the luciferase). * p = intensities is shown 0.02 (SST vs. DMSO alone); * p = 0.02 (4-MMI vs. DMSO alone). graphically in the lower panel, as described in ‘Section2. Materials and Methods’. * p = 0.02 (SST vs. DMSO3.5. Compound alone); 4- *MMIp = 0.02Reduces (4-MMI Tumor vs. Burden DMSO in a alone).Myeloma Model Next, we examined the capacity of compound 4-MMI to inhibit myeloma primary tumor growth. For this purpose, we applied the MPC-11 syngeneic myeloma model [20,32]. Briefly, BALB/c mice were implanted subcutaneously with MPC-11 mouse myeloma cells (0.5 × 106/mouse) and treated with either DMSO (0.1 mL/mouse) or 4-MMI (daily 200 g/mouse, i.p.), starting on day two after tumor cell inoculation. Xenograft Tumor size was measured on days 8 and 12 (Figure 5A), resected, and weighed (Figure 5B) (see also Figure S2). Strikingly, compound 4-MMI effectively and markedly decreased the rate of tumor growth and the tumor burden. Immunostaining of fixed paraffin-embedded tissue sections with anti-VEGF antibodies revealed a marked decrease in the levels of VEGF in tumors derived from mice treated with 4-MMI (Figure S3), likely reflecting an anti-angiogenesis effect of the drug.

Cancers 2021, 13, 2959 12 of 17

3.5. Compound 4-MMI Reduces Tumor Burden in a Myeloma Model Next, we examined the capacity of compound 4-MMI to inhibit myeloma primary tumor growth. For this purpose, we applied the MPC-11 syngeneic myeloma model [20,32]. Briefly, BALB/c mice were implanted subcutaneously with MPC-11 mouse myeloma cells (0.5 × 106/mouse) and treated with either DMSO (0.1 mL/mouse) or 4-MMI (daily 200 µg/mouse, i.p.), starting on day two after tumor cell inoculation. Xenograft Tumor size was measured on days 8 and 12 (Figure5A), resected, and weighed (Figure5B) (see also Figure S2). Strikingly, compound 4-MMI effectively and markedly decreased the rate of tumor growth and the tumor burden. Immunostaining of fixed paraffin-embedded tissue sections with anti-VEGF antibodies revealed a marked decrease in the levels of Cancers 2021, 13, x 13 of 18 VEGF in tumors derived from mice treated with 4-MMI (Figure S3), likely reflecting an anti-angiogenesis effect of the drug.

FigureFigure 5. 5. MPCMPC-11-11 tumor tumor growth. growth. Cells Cells (MPC (MPC-11-11 mouse mouse myeloma myeloma)) from exponential from exponential cultures cultures were washed with PBS and brought to a concentration of 5 × 106 cells/mL. Cell suspension (5 × 105/0.1 were washed with PBS and brought to a concentration of 5 × 106 cells/mL. Cell suspension mL) was inoculated subcutaneously at the right flank of 6-week-old female BALB/c mice (n = 6). (5 × 105/0.1 mL) was inoculated subcutaneously at the right flank of 6-week-old female BALB/c mice Mice were treated with either DMSO (0.1 mL/mouse) or test compound (daily 200 µg/mouse, i.p.), µ starting(n = 6). Miceon day were 2 after treated the withinjection either of DMSOtumor (0.1cells, mL/mouse) for 12 days. or Tumor test compound size was (dailydetermined 200 g/mouse, on daysi.p.), 8 starting and 12 onby dayexternally 2 after measuring the injection the of tumors tumor in cells, two for dimensions 12 days. Tumor using sizea caliper was determined(A, p < 0.001 on). Atdays the 8end and of 12 the by experim externallyent, measuring mice were thesacrificed tumors and in two the dimensionstumors resected, using weighed a caliper ( (BA, ,pp =< 0.003 0.001).), andAt thephotographed. end of the experiment, mice were sacrificed and the tumors resected, weighed (B, p = 0.003), and photographed. 4. Discussion 4. Discussion Heparanase is a multifaceted enzyme that together with heparan sulfate drives signal transduction,Heparanase immune is a multifaceted cell activation, enzyme exosome that formation, together with autoph heparanagy, and sulfate gene drives transcrip- signal transduction, immune cell activation, exosome formation, autophagy, and gene transcrip- tion via enzymatic and non-enzymatic activities [8,34,35]. Heparanase releases a myriad tion via enzymatic and non-enzymatic activities [8,34,35]. Heparanase releases a myriad of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan of HS-bound growth factors, cytokines, and chemokines that are sequestered by heparan sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate–heparanase axis sulfate in the glycocalyx and ECM. Collectively, the heparan sulfate–heparanase axis plays plays pivotal roles in creating a permissive environment for cell proliferation, differentia- pivotal roles in creating a permissive environment for cell proliferation, differentiation, tion, and function, often resulting in the pathogenesis of diseases, such as cancer, chronic and function, often resulting in the pathogenesis of diseases, such as cancer, chronic inflam- inflammation, sepsis, endotheliitis, kidney dysfunction, diabetic nephropathy, diabetes, mation, sepsis, endotheliitis, kidney dysfunction, diabetic nephropathy, diabetes, tissue tissue fibrosis, bone osteolysis, thrombosis, atherosclerosis, and viral infection [36]. Hep- aranase represents a druggable target because (i) there is only a single enzymatically ac- tive heparanase expressed in humans; (ii) the enzyme is present in low levels in normal tissues but is markedly elevated in cancer, inflammation, and other pathologies; and (iii) heparanase deficient mice appear normal [37]. Thus, properly designed heparanase inhib- itors will likely have few, if any, negative side effects. The development of heparanase inhibitors has focused predominantly on carbohydrate-based compounds with heparin- like properties [38–40]. These compounds bind to the heparin-binding domains that flank the of heparanase thereby inhibiting cleavage of heparan sulfate. Four different heparin mimics are currently in clinical trials in human cancer patients. However, all of these mimics have the disadvantage that they are not specific for heparanase and likely interact with different heparin-binding proteins with unknown consequences and off-tar- get effects [38,39]. Therefore, even if they prove efficacious in patients, it will be impossible

Cancers 2021, 13, 2959 13 of 17

fibrosis, bone osteolysis, thrombosis, atherosclerosis, and viral infection [36]. Heparanase represents a druggable target because (i) there is only a single enzymatically active hep- aranase expressed in humans; (ii) the enzyme is present in low levels in normal tissues but is markedly elevated in cancer, inflammation, and other pathologies; and (iii) heparanase deficient mice appear normal [37]. Thus, properly designed heparanase inhibitors will likely have few, if any, negative side effects. The development of heparanase inhibitors has focused predominantly on carbohydrate-based compounds with heparin-like proper- ties [38–40]. These compounds bind to the heparin-binding domains that flank the active site of heparanase thereby inhibiting cleavage of heparan sulfate. Four different heparin mimics are currently in clinical trials in human cancer patients. However, all of these mimics have the disadvantage that they are not specific for heparanase and likely inter- act with different heparin-binding proteins with unknown consequences and off-target effects [38,39]. Therefore, even if they prove efficacious in patients, it will be impossible to attribute their effect solely to heparanase inhibition. Moreover, three of the four mimics are heterogeneous in structure, adding to their complexity and uncertainty as viable drugs for use in humans. Small-molecule inhibitors of heparanase have the potential to overcome some of the limitations of polysaccharides, offering an opportunity for specificity, favorable pharmacokinetics, and oral availability [36,41]. Numerous heparanase-inhibiting small molecules were reported [39,42,43], but none entered clinical testing. Surprisingly, only one compound was found to inhibit experimental metastasis (B16 melanoma model) [44], and there are no reports of their preclinical testing in xenograft tumor models. We have previously reported the results of an in vitro screening of a library of small molecules characterized by a variety of scaffolds. More than 150 compounds were tested for their ability to inhibit the enzymatic activity of human heparanase, identify- ing [1,2,4]triazolo[3,4-b][1,3,4]thiadiazole derivatives as active substances. The most potent anti-heparanase derivative, DTP, was found to inhibit tumor cell proliferation, migration, and invasion in vitro with IC50 values in the micromolar range [22]. The present study focuses on the development of related triazolo–thiadiazole compounds that inhibit hep- aranase enzymatic activity, ECM degradation, cell invasion, experimental metastasis, and tumor growth in mouse models. We used DTP as a template structure for the synthesis of substituted triazolo–thiadiazoles endowed with better heparanase inhibitory activity. The structure of DTP was modified by removing iodine at the second position of the phenyl ring and the addition of a methyl group on the terminal benzene ring, which resulted in the formation of compound 4-MMI. MMI showed relatively potent heparanase inhibitory and antitumor activity compared to its parent compound. Other structural analogs showed var- ious degrees of heparanase inhibition. The newly designed compounds were examined for their capacity to inhibit heparanase enzymatic activity, using as substrates heparin-derived pentasaccharide [23] and HS naturally embedded in the ECM produced by endothelial cells [25]. To translate the in vitro inhibition of heparanase enzymatic activity and cell invasion to preclinical models, the most promising triazolo–thiadiazole compounds were examined in the 4T1 mouse mammary carcinoma model of experimental metastasis and the MPC-11 mouse model of myeloma tumor growth [20,32]. Breast cancer metastasis is the leading cause of female mortality worldwide and accounts for 25% of the total number of cancer cases and 15% of all cancer-associated female mortality [45]. Importantly, compound 4-MMI yielded a nearly fourfold inhibition of 4T1 breast carcinoma metastasis, comparable to the effect exerted by Roneparstat (=SST), a well-characterized heparin-like heparanase inhibitor (Figure4)[15,17,33,36,39,41]. Multiple myeloma is a devastating cancer of plasma cells that is highly dependent on the bone marrow microenvironment for growth and survival [46]. Heparanase has been shown to critically accelerate myeloma tumor growth and bone colonization [47], and the heparanase inhibitor Roneparstat has been examined in myeloma patients, yield- ing promising results [33]. Strikingly, treatment with compound 4-MMI resulted in a profound inhibition of tumor growth and burden in the aggressive MPC-11 myeloma model (Figure5). We also noted a profound reduction in VEGF levels in tumor sections Cancers 2021, 13, 2959 14 of 17

derived from 4-MMI treated vs. untreated mice, likely due to inhibition of heparanase- mediated VEGF and/or release [6,7,10]. Due to limited amounts of the 4-MMI compound, we could not perform dose–response and pharmacokinetic studies. Yet, there were no signs of distress and toxicity in response to the daily administration of 200 ug/mouse/day, a dose that is well below the amounts of anti-cancer small molecules given to mice and the equivalent dose in humans. In subsequent studies we will examine the effect of 4-MMI in additional tumor models, with emphasis on orthotopic models of breast and pancreatic carcinomas. We will also apply medicinal chemistry and computational principles to further optimize the compound for improved potency; specificity; selectiv- ity; and pharmacokinetic properties, including stability, permeability, solubility, and oral bioavailability.

5. Conclusions To the best of our knowledge, this is the first report showing a marked decrease in primary tumor growth in mice treated with a small molecule that inhibits heparanase enzymatic activity. Given these encouraging results, studies are underway to better eluci- date the mode of action and clinical significance of compound 4-MMI and related triazolo– thiadiazoles. Among other aspects, the compound is being examined for an effect on human tumor xenografts, primarily myeloma and breast carcinoma, pharmacokinetic characteris- tics, efficacy, and mode of administration. Docking and SAR studies are being performed, applying the recently resolved crystal structure of the heparanase protein [48–50]. Selected molecules exerting little or no side effects will then be examined for oral availability and anti-cancer effects in combination with currently available treatments. We will also study the effect of our lead compounds on other pathological indications involving heparanase, such as chronic inflammation (i.e., colitis, pancreatitis) [51,52], tissue fibrosis [53], kidney dysfunction (i.e., AKI, diabetic nephropathy) [54–57], and diabetes [58]. Of particular relevance are recent studies on the presumed involvement of heparanase and heparan sulfate in the pathogenesis of COVID-19 [59–62]. Studies on the effect of 4-MMI and related compounds on SARS-CoV-2 infectivity are ongoing.

Supplementary Materials: The following are available online at https://www.mdpi.com/article/10 .3390/cancers13122959/s1. Figure S1: Inhibition of 4T1 breast carcinoma experimental metastasis, Figure S2: MPC-11 tumor growth, Figure S3: Immunostaining The spectra and other information are freely available with this paper. Author Contributions: Conceptualization and design, K.S.R., C.D.M., B.B. and I.V.; experimental work, U.B., I.B., D.V. and S.R.; writing—original draft, C.D.M., S.R., D.V., B.B., I.B. and U.B.; writing— review and editing, C.D.M., B.B., K.S.R. and I.V.; supervision, K.S.R., C.D.M., B.B. and I.V. All authors have read and agreed to the published version of the manuscript. Funding: This work was supported by the Israel Science Foundation (ISF) and the University Grants Commission (UGC), India, awarded to I.V. and K.S.R. within the ISF-UGC joint research program framework (grant No. 2277/15). It was also supported by grants from the Israel Science Foundation (1021/19) and the Israel Cancer Research Fund (ICRF). I.V. is a Research Professor of the Israel Cancer Research Fund (ICRF). B. thanks the Department of Biotechnology (BT/PR24978/NER/95/938/2017) and Vision Group on Science and Technology (CESEM), Government of Karnataka for funding. This project was supported by DST PhD Fellowship from KSTePS to D.V. Institutional Review Board Statement: The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board (018-08-18). Informed Consent Statement: Not applicable. Data Availability Statement: All the data associated with this study are freely available. Acknowledgments: K.S.R. thanks the Institution of Excellence, DST-PURSE, University of Mysore, for providing infrastructure and other research facilities. K.S.R. also thanks the Council of Scientific and Industrial Research for providing an emeritus scientist fellowship. Conflicts of Interest: The authors declare no conflict of interest. Cancers 2021, 13, 2959 15 of 17

References 1. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [CrossRef] 2. Pozzi, A.; Yurchenco, P.D.; Iozzo, R.V. The nature and biology of basement membranes. Matrix Biol. 2017, 57–58, 1–11. [CrossRef] 3. Karamanos, N.K.; Piperigkou, Z.; Theocharis, A.D.; Watanabe, H.; Franchi, M.; Baud, S.; Brézillon, S.; Götte, M.; Passi, A.; Vigetti, D.; et al. Proteoglycan Chemical Diversity Drives Multifunctional Cell Regulation and Therapeutics. Chem. Rev. 2018, 118, 9152–9232. [CrossRef] 4. Kjellén, L.; Lindahl, U. Specificity of glycosaminoglycan-protein interactions. Curr. Opin. Struct. Biol. 2018, 50, 101–108. [CrossRef] 5. Basappa; Kumar, C.A.; Swamy, S.N.; Sugahara, K.; Rangappa, K.S. Anti-tumor and anti-angiogenic activity of novel hydantoin derivatives: Inhibition of VEGF secretion in liver metastatic osteosarcoma cells. Bioorganic Med. Chem. 2009, 17, 4928–4934. [CrossRef] 6. Basappa; Murugan, S.; Kavitha, C.V.; Purushothaman, A.; Nevin, K.G.; Sugahara, K.; Rangappa, K.S. A small oxazine compound as an anti-tumor agent: A novel pyranoside mimetic that binds to VEGF, HB-EGF, and TNF-α. Cancer Lett. 2010, 297, 231–243. [CrossRef][PubMed] 7. Hari, S.; Swaroop, T.R.; Preetham, H.D.; Mohan, C.D.; Muddegowda, U.; Basappa, S.; Vlodavsky, I.; Sethi, G.; Rangappa, K.S. Synthesis, Cytotoxic and Heparanase Inhibition Studies of 5-oxo-1-arylpyrrolidine-3- carboxamides of Hydrazides and 4-amino-5-aryl-4H-1,2,4-triazole-3-thiol. Curr. Org. Synth. 2020, 17, 243–250. [CrossRef][PubMed] 8. Ilan, N.; Bhattacharya, U.; Barash, U.; Boyango, I.; Yanku, Y.; Gross-Cohen, M.; Vlodavsky, I. Heparanase—The Message Comes in Different Flavors. Adv. Exp. Med. Biol. 2020, 1221, 253–283. [CrossRef][PubMed] 9. Vlodavsky, I.; Ilan, N.; Sanderson, R.D. Forty Years of Basic and Translational Heparanase Research. Adv. Exp. Med. Biol. 2020, 1221, 3–59. [CrossRef] 10. Hao, N.-B.; Tang, B.; Wang, G.-Z.; Xie, R.; Hu, C.-J.; Wang, S.-M.; Wu, Y.-Y.; Liu, E.; Xie, X.; Yang, S.-M. Hepatocyte growth factor (HGF) upregulates heparanase expression via the PI3K/Akt/NF-κB signaling pathway for gastric cancer metastasis. Cancer Lett. 2015, 361, 57–66. [CrossRef] 11. Kundu, S.; Xiong, A.; Spyrou, A.; Wicher, G.; Marinescu, V.D.; Edqvist, P.-H.D.; Zhang, L.; Essand, M.; Dimberg, A.; Smits, A.; et al. Heparanase Promotes Glioma Progression and Is Inversely Correlated with Patient Survival. Mol. Cancer Res. 2016, 14, 1243–1253. [CrossRef] 12. Ilan, N.; Elkin, M.; Vlodavsky, I. Regulation, function and clinical significance of heparanase in cancer metastasis and angiogenesis. Int. J. Biochem. Cell Biol. 2006, 38, 2018–2039. [CrossRef] 13. Vlodavsky, I.; Beckhove, P.; Lerner, I.; Pisano, C.; Meirovitz, A.; Ilan, N.; Elkin, M. Significance of Heparanase in Cancer and Inflammation. Cancer Microenviron. 2011, 5, 115–132. [CrossRef] 14. Mohan, C.D.; Hari, S.; Preetham, H.D.; Rangappa, S.; Barash, U.; Ilan, N.; Nayak, S.C.; Gupta, V.K.; Basappa; Vlodavsky, I.; et al. Targeting Heparanase in Cancer: Inhibition by Synthetic, Chemically Modified, and Natural Compounds. iScience 2019, 15, 360–390. [CrossRef][PubMed] 15. Cassinelli, G.; Torri, G.; Naggi, A. Non-Anticoagulant as Heparanase Inhibitors. Adv. Exp. Med. Biol. 2020, 1221, 493–522. [CrossRef] 16. Chhabra, M.; Ferro, V. PI-88 and Related Heparan Sulfate Mimetics. Adv. Exp. Med. Biol. 2020, 1221, 473–491. [CrossRef] [PubMed] 17. Noseda, A.; Barbieri, P. Roneparstat: Development, Preclinical and Clinical Studies. Adv. Exp. Med. Biol. 2020, 1221, 523–538. [CrossRef] 18. Hammond, E.; Dredge, K. Heparanase Inhibition by Pixatimod (PG545): Basic Aspects and Future Perspectives. Adv. Exp. Med. Biol. 2020, 1221, 539–565. [CrossRef][PubMed] 19. Priya, B.; Kumar, C.A.; Swamy, S.N.; Basappa; Naveen, S.; Prasad, J.S.; Rangappa, K. 2-(2-(2-Ethoxybenzoylamino)-4- chlorophenoxy)-N-(2-ethoxybenzoyl)benzamine inhibits EAT cell induced angiogenesis by down regulation of VEGF secretion. Bioorganic Med. Chem. Lett. 2007, 17, 2775–2780. [CrossRef][PubMed] 20. Sanderson, R.D.; Sneed, T.B.; Young, L.A.; Sullivan, G.L.; Lander, A.D. Adhesion of B lymphoid (MPC-11) cells to type I collagen is mediated by integral membrane proteoglycan, syndecan. J. Immunol. 1992, 148, 3902–3911. [PubMed] 21. Alishekevitz, D.; Bril, R.; Loven, D.; Miller, V.; Voloshin, T.; Gingis-Velistki, S.; Fremder, E.; Scherer, S.J.; Shaked, Y. Differential Therapeutic Effects of Anti-VEGF-A Antibody in Different Tumor Models: Implications for Choosing Appropriate Tumor Models for Drug Testing. Mol. Cancer Ther. 2013, 13, 202–213. [CrossRef] 22. Baburajeev, C.P.; Mohan, C.D.; Rangappa, S.; Mason, D.J.; Fuchs, J.E.; Bender, A.; Barash, U.; Vlodavsky, I.; Basappa; Rangappa, K.S. S. Identification of Novel Class of Triazolo-Thiadiazoles as Potent Inhibitors of Human Heparanase and their Anticancer Activity. BMC Cancer 2017, 17, 235. [CrossRef] 23. Hammond, E.; Li, C.P.; Ferro, V. Development of a colorimetric assay for heparanase activity suitable for kinetic analysis and inhibitor screening. Anal. Biochem. 2010, 396, 112–116. [CrossRef] 24. Vlodavsky, I.; Friedmann, Y.; Elkin, M.; Aingorn, H.; Atzmon, R.; Ishai-Michaeli, R.; Bitan, M.; Pappo, O.; Peretz, T.; Michal, I.; et al. Mammalian heparanase: Gene cloning, expression and function in tumor progression and metastasis. Nat. Med. 1999, 5, 793–802. [CrossRef][PubMed] Cancers 2021, 13, 2959 16 of 17

25. Vlodavsky, I. Preparation of Extracellular Matrices Produced by Cultured Corneal Endothelial and PF-HR9 Endodermal Cells. Curr. Protoc. Cell Biol. 1999, 1.[CrossRef][PubMed] 26. Vlodavsky, I.; Fuks, Z.; Bar-Ner, M.; Ariav, Y.; Schirrmacher, V. Lymphoma cell-mediated degradation of sulfated proteoglycans in the subendothelial extracellular matrix: Relationship to tumor cell metastasis. Cancer Res. 1983, 43, 2704–2711. [PubMed] 27. Zetser, A.; Bashenko, Y.; Miao, H.-Q.; Vlodavsky, I.; Ilan, N. Heparanase affects adhesive and tumorigenic potential of human glioma cells. Cancer Res. 2003, 63, 7733–7741. 28. Barash, U.; Spyrou, A.; Liu, P.; Vlodavsky, E.; Zhu, C.; Luo, J.; Su, D.; Ilan, N.; Forsberg-Nilsson, K.; Vlodavsky, I.; et al. Heparanase promotes glioma progression via enhancing CD24 expression. Int. J. Cancer 2019, 145, 1596–1608. [CrossRef][PubMed] 29. Loka, R.S.; Sletten, E.T.; Barash, U.; Vlodavsky, I.; Nguyen, H.M. Specific Inhibition of Heparanase by a Glycopolymer with Well-Defined Sulfation Pattern Prevents Breast Cancer Metastasis in Mice. ACS Appl. Mater. Interfaces 2019, 11, 244–254. [CrossRef] 30. Barash, U.; Lapidot, M.; Zohar, Y.; Loomis, C.; Moreira, A.; Feld, S.; Goparaju, C.; Yang, H.; Hammond, E.; Zhang, G.; et al. Involvement of Heparanase in the Pathogenesis of Mesothelioma: Basic Aspects and Clinical Applications. J. Natl. Cancer Inst. 2018, 110, 1102–1114. [CrossRef] 31. Weissmann, M.; Arvatz, G.; Horowitz, N.; Feld, S.; Naroditsky, I.; Zhang, Y.; Ng, M.; Hammond, E.; Nevo, E.; Vlodavsky, I.; et al. Heparanase-neutralizing antibodies attenuate lymphoma tumor growth and metastasis. Proc. Natl. Acad. Sci. USA 2016, 113, 704–709. [CrossRef] 32. Barash, U.; Zohar, Y.; Wildbaum, G.; Beider, K.; Nagler, A.; Karin, N.; Ilan, N.; Vlodavsky, I. Heparanase enhances myeloma progression via CXCL10 downregulation. Leukemia 2014, 28, 2178–2187. [CrossRef][PubMed] 33. Galli, M.; Chatterjee, M.; Grasso, M.; Specchia, G.; Magen, H.; Einsele, H.; Celeghini, I.; Barbieri, P.; Paoletti, D.; Pace, S.; et al. Phase I study of the heparanase inhibitor roneparstat: An innovative approach for ultiple myeloma therapy. Haematologica 2018, 103, e469–e472. [CrossRef][PubMed] 34. Vlodavsky, I.; Singh, P.; Boyango, I.; Gutter-Kapon, L.; Elkin, M.; Sanderson, R.D.; Ilan, N. Heparanase: From basic research to therapeutic applications in cancer and inflammation. Drug Resist. Updates 2016, 29, 54–75. [CrossRef] 35. Vlodavsky, I.; Gross-Cohen, M.; Weissmann, M.; Ilan, N.; Sanderson, R.D. Opposing Functions of Heparanase-1 and Heparanase-2 in Cancer Progression. Trends Biochem. Sci. 2018, 43, 18–31. [CrossRef][PubMed] 36. Giannini, G.; Battistuzzi, G.; Rivara, S. The Control of Heparanase through the Use of Small Molecules. Adv. Exp. Med. Biol. 2020, 1221, 567–603. [CrossRef][PubMed] 37. Zcharia, E.; Jia, J.; Zhang, X.; Baraz, L.; Lindahl, U.; Peretz, T.; Vlodavsky, I.; Li, J.-P. Newly Generated Heparanase Knock-Out Mice Unravel Co-Regulation of Heparanase and Matrix Metalloproteinases. PLoS ONE 2009, 4, e5181. [CrossRef] 38. Pisano, C.; Vlodavsky, I.; Ilan, N.; Zunino, F. The potential of heparanase as a therapeutic target in cancer. Biochem. Pharmacol. 2014, 89, 12–19. [CrossRef] 39. Rivara, S.; Milazzo, F.M.; Giannini, G. Heparanase: A rainbow pharmacological target associated to multiple pathologies including rare diseases. Future Med. Chem. 2016, 8, 647–680. [CrossRef] 40. Vlodavsky, I.; Ilan, N.; Naggi, A.; Casu, B. Heparanase: Structure, Biological Functions, and Inhibition by Heparin-Derived Mimetics of Heparan Sulfate. Curr. Pharm. Des. 2007, 13, 2057–2073. [CrossRef] 41. Messore, A.; Madia, V.N.; Pescatori, L.; Saccoliti, F.; Tudino, V.; De Leo, A.; Bortolami, M.; De Vita, D.; Scipione, L.; Pepi, F.; et al. Novel Symmetrical Benzazolyl Derivatives Endowed with Potent Anti-Heparanase Activity. J. Med. Chem. 2018, 61, 10834–10859. [CrossRef] 42. Simizu, S.; Ishida, K.; Osada, H. Heparanase as a molecular target of cancer chemotherapy. Cancer Sci. 2004, 95, 553–558. [CrossRef][PubMed] 43. Xu, Y.-J.; Miao, H.-Q.; Pan, W.; Navarro, E.C.; Tonra, J.R.; Mitelman, S.; Camara, M.M.; Deevi, D.S.; Kiselyov, A.S.; Kussie, P.; et al. N-(4-{[4-(1H-Benzoimidazol-2-yl)-arylamino]-methyl}-phenyl)-benzamide derivatives as small molecule heparanase inhibitors. Bioorganic Med. Chem. Lett. 2006, 16, 404–408. [CrossRef] 44. Pan, W.; Miao, H.-Q.; Xu, Y.-J.; Navarro, E.C.; Tonra, J.R.; Corcoran, E.; Lahiji, A.; Kussie, P.; Kiselyov, A.S.; Wong, W.C.; et al. 1-[4-(1H-Benzoimidazol-2-yl)-phenyl]-3-[4-(1H-benzoimidazol-2-yl)-phenyl]-urea derivatives as small molecule heparanase inhibitors. Bioorganic Med. Chem. Lett. 2006, 16, 409–412. [CrossRef] 45. Torre, L.A.; Bray, F.; Siegel, R.L.; Ferlay, J.; Lortet-Tieulent, J.; Jemal, A. Global cancer statistics, 2012: Global Cancer Statistics, 2012. CA Cancer J. Clin. 2015, 65, 87–108. [CrossRef][PubMed] 46. Marino, S.; Roodman, G.D. Multiple Myeloma and Bone: The Fatal Interaction. Cold Spring Harb. Perspect. Med. 2018, 8, a031286. [CrossRef] 47. Purushothaman, A.; Sanderson, R.D. Heparanase: A Dynamic Promoter of Myeloma Progression. Adv. Exp. Med. Biol. 2020, 1221, 331–349. [CrossRef] 48. Wu, L.; Davies, G.J. An Overview of the Structure, Mechanism and Specificity of Human Heparanase. Adv. Exp. Med. Biol. 2020, 1221, 139–167. [CrossRef] 49. Wu, L.; Viola, C.M.; Brzozowski, A.M.; Davies, G.J. Structural characterization of human heparanase reveals insights into substrate recognition. Nat. Struct. Mol. Biol. 2015, 22, 1016–1022. [CrossRef][PubMed] 50. Wu, L.; Jiang, J.; Jin, Y.; Kallemeijn, W.W.; Kuo, C.-L.; Artola, M.; Dai, W.; Van Elk, C.; van Eijk, M.; Van Der Marel, G.A.; et al. Activity-based probes for functional interrogation of retaining β-glucuronidases. Nat. Chem. Biol. 2017, 13, 867–873. [CrossRef] 51. Khamaysi, I.; Hamo-Giladi, D.B.; Abassi, Z. Heparanase in Acute Pancreatitis. Adv. Exp. Med. Biol. 2020, 1221, 703–719. [CrossRef] Cancers 2021, 13, 2959 17 of 17

52. Lerner, I.; Hermano, E.; Zcharia, E.; Rodkin, D.; Bulvik, R.; Doviner, V.; Rubinstein, A.M.; Ishai-Michaeli, R.; Atzmon, R.; Sherman, Y.; et al. Heparanase powers a chronic inflammatory circuit that promotes colitis-associated tumorigenesis in mice. J. Clin. Investig. 2011, 121, 1709–1721. [CrossRef] 53. Masola, V.; Gambaro, G.; Onisto, M. Impact of Heparanse on Organ Fibrosis. Adv. Exp. Med. Biol. 2020, 1221, 669–684. [CrossRef] 54. Abassi, Z.; Goligorsky, M.S. Heparanase in Acute Kidney Injury. Adv. Exp. Med. Biol. 2020, 1221, 685–702. [CrossRef] 55. Van Der Vlag, J.; Buijsers, B. Heparanase in Kidney Disease. Adv. Exp. Med. Biol. 2020, 1221, 647–667. [CrossRef] 56. Rabelink, T.; Berg, B.M.V.D.; Garsen, M.; Wang, G.; Elkin, M.; Van Der Vlag, J. Heparanase: Roles in cell survival, extracellular matrix remodelling and the development of kidney disease. Nat. Rev. Nephrol. 2017, 13, 201–212. [CrossRef] 57. Gil, N.; Goldberg, R.; Neuman, T.; Garsen, M.; Zcharia, E.; Rubinstein, A.M.; Van Kuppevelt, T.; Meirovitz, A.; Pisano, C.; Li, J.-P.; et al. Heparanase Is Essential for the Development of Diabetic Nephropathy in Mice. Diabetes 2011, 61, 208–216. [CrossRef] [PubMed] 58. Simeonovic, C.J.; Popp, S.K.; Brown, D.J.; Li, F.-J.; Lafferty, A.R.A.; Freeman, C.; Parish, C.R. Heparanase and Type 1 Diabetes. Adv. Exp. Med. Biol. 2020, 1221, 607–630. [CrossRef][PubMed] 59. Shi, C.; Wang, C.; Wang, H.; Yang, C.; Cai, F.; Zeng, F.; Cheng, F.; Liu, Y.; Zhou, T.; Deng, B.; et al. The Potential of Low Molecular Weight Heparin to Mitigate Cytokine Storm in Severe COVID-19 Patients: A Retrospective Cohort Study. Clin. Transl. Sci. 2020, 13, 1087–1095. [CrossRef] 60. Buijsers, B.; Yanginlar, C.; Maciej-Hulme, M.L.; de Mast, Q.; van der Vlag, J. Beneficial non-anticoagulant mechanisms underlying heparin treatment of COVID-19 patients. EBioMedicine 2020, 59, 102969. [CrossRef][PubMed] 61. Martino, C.; Kellman, B.P.; Sandoval, D.R.; Clausen, T.M.; Marotz, C.A.; Song, S.J.; Wandro, S.; Zaramela, L.S.; Salido Benítez, R.A.; Zhu, Q.; et al. Bacterial modification of the host glycosaminoglycan heparan sulfate modulates SARS-CoV-2 infectivity. bioRxiv 2020.[CrossRef] 62. Agelidis, A.; Shukla, D. Heparanase, Heparan Sulfate and Viral Infection. Adv. Exp. Med. Biol. 2020, 1221, 759–770. [CrossRef] [PubMed]