1,805
Views
48
CrossRef citations to date
0
Altmetric
Research Article

Synthesis, cytotoxicity and carbonic anhydrase inhibitory activities of new pyrazolines

, , , , , , , & show all
Pages 20-24 | Received 04 Jun 2016, Accepted 20 Jul 2016, Published online: 31 Aug 2016

Abstract

A series of polymethoxylated-pyrazoline benzene sulfonamides were synthesized, investigated for their cytotoxic activities on tumor and non-tumor cell lines and inhibitory effects on carbonic anhydrase isoenzymes (hCA I and hCA II). Although tumor selectivity (TS) of the compounds were less than the reference compounds 5-Fluorouracil and Melphalan, trimethoxy derivatives 4, 5, and 6 were more selective than dimethoxy derivatives 2 and 3 as judged by the cytotoxicity assay with the cells both types originated from the gingival tissue. The compound 6 (4-[3-(4-methoxyphenyl)-5-(3,4,5-trimethoxyphenyl)-4,5-dihydro-1H-pyrazol-1-yl] benzene sulfonamide) showed the highest TS values and can be considered as a lead molecule of the series for further investigations. All compounds synthesized showed superior CA inhibitory activity than the reference compound acetazolamide on hCA I, and II isoenzymes, with inhibition constants in the range of 26.5–55.5 nM against hCA I and of 18.9–28.8 nM against hCA II, respectively.

Introduction

Cancer is one of the most important causes of deaths in the worldCitation1. Various classes of drugs such as alkylating agents, antimetabolites, antibiotics, hormones and plant alkaloids are used in the traditional cancer chemotherapyCitation2. Although selective toxicity is preferable in cancer treatment, the drugs used in clinics have several side effects such as resistance development to the anticancer agents in addition to low selectivitiesCitation3.

Carbonic anhydrase (CA, EC 4.2.1.1) a zinc-dependent metalloenzyme that catalyzes the reversible hydration of CO2. Sixteen different α-CA isoforms have been isolated and characterized so far in mammals: CA I, CA II, CA III, CA VII, CA XIII are cytosolic; CA IV, CA IX, CA XII, CA XIV and CA XV are associated with the cell membrane; CA VA and CA VB are in mitochondria and CA VI is secreted into saliva and milkCitation4.

The CAs contribute many physiological and pathological processes such as pH homeostasis, electrolyte secretion in various tissues and organs, gluconeogenesis, lipogenesis, ureagenesis, bone resorption calcification and tumorigenicityCitation5–7. Thus, the CAs have become remarkable therapeutic targets for the treatment of a wide range of disorders. Several CA inhibitors show anticancerCitation7, diureticsCitation8, antiglaucomaCitation9, antiinfectiveCitation10 and antiobesityCitation11 activities.

Pyrazole/pyrazolines are an important class of heterocyclic compounds containing three carbon atoms and two nitrogen atoms in adjacent positions. These heterocyclic compounds can be seen in nature as alkaloids, vitamins, pigments and constituents of plant and animal cells widelyCitation12. Medicinal chemists have great attention of pyrazolines and substituted pyrazolines because of their various biological activities such as antimicrobial, anticonvulsant, anticancer, anti-inflammatory, antitubercular, antiviral, analgesic and antidepressive activitiesCitation13–17. Sulfonamide is an important functional group in medicinal chemistry having a wide range of bioactivities such as cytotoxic, carbonic anhydrase inhibitory, antibacterial, antimalarial, antihypertensive, antiviral, anti-inflammatory and diuretics activitiesCitation8,13,18–25.

In this study it was aimed to synthesize new compounds bearing pyrazoline and sulfonamide pharmacophores and to test their carbonic anhydrase inhibitory effects on hCA I and II isoenzymes. It was also planned to investigate their cytotoxic activities towards four cancer cell lines (Ca9–22, HSC-2, HSC-3, and HSC-4) and three non-tumor cells (HGF, HPLF and HPC) to see whether any of compounds synthesized is/are tumor-specific cytotoxin/s.

Experimental

Materials and methods

Chemical structure of the compounds were determined by 1H NMR (400 MHz) and 13C NMR (100 MHz) spectroscopies using a Varian Mercury Plus spectrometer (Varian Inc., Palo Alto, CA). Chemical shifts (δ) are reported in ppm and coupling constants (J) are expressed in hertz (Hz). Mass spectra for the compound 1 were taken using a liquid chromatography ion trap-time of flight tandem mass spectrometer (Shimadzu, Kyoto, Japan) equipped with an electrospray ionization (ESI) source, operating in both positive and negative ionization mode. Shimadzu's LCMS Solution software was used for data analysis. Mass spectra for the compounds 26 were undertaken on a HPLC-TOF Waters Micromass LCT Premier XE (Milford, MA) mass spectrometer using an electrospray ion source (ESI). Melting points were determined using an electrothermal 9100/IA9100 instrument (Bibby Scientific Limited, Staffordshire, UK) and are uncorrected. The reactions were monitored using silica gel HF254–366 TLC plates (E. Merck, Darmstadt, Germany). IR spectra of the compounds were taken using a FT-IR spectrometer (Perkin Elmer Spektrum One FT-IR, Bridgeport Avenue Shelton, CT).

Synthesis of chalcone compounds, 1a6a

4-Methoxyacetophenone and suitable aldehyde in 1:1 mol ratio were dissolved in ethanol (10 ml)Citation23. Aqueous NaOH solution (10%, 20 ml) was added into the reaction flask. Reactions were monitored by TLC. When at least one of the starting compounds finished, reactions were stopped. Reaction content was poured on ice-water and neutralized by HCl (10%). The precipitates obtained were washed with cold water and ethanol, filtered, and dried. The purities were checked by TLC and used for the synthesis of pyrazoline derivatives 16 without further purification.

Synthesis of pyrazoline containing benzene sulfonamides, 16

A mixture of a suitable chalcone compound (1a6a) and 4-hydrazinobenzenesulfonamide hydrochloride in 1:1.1 mol ratio in ethanol (25 ml for 13 and 30 ml for 46) in the presence of glacial acetic acid (0.05 ml) was refluxedCitation26. Reactions were monitored by TLC using CHCl3:MeOH (4.8:0.2) as a solvent system. When the reactions were stopped, the precipitate obtained was filtered, dried and crystallized from suitable solvent system. Experimental data of the compounds are presented in . Spectral data of the compounds are presented in .

Table 1. Experimental data of the compounds 1–6.

Table 2. Spectral data of the compounds 1–6.

Biological activity

Carbonic anhydrase inhibition assay

The purification of cytosolic CA isoenzymes (CA I and II) were previously described with a simple one-step method by a sepharose-4B-L tyrosine-sulphanilamide affinity chromatographyCitation27. The protein quantity in the column effluents was determined spectrophotometrically at 280 nm. Sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE) was applied with a Bio-Rad Mini Gel system (Mini-PROTEAN Tetra System, China) after purification of both CA isoenzymesCitation28. Briefly, it was performed in acrylamide for the running (10%) and the stacking gel (3%) contained SDS (0.1%), respectively. Activities of CA I and II isoenzymes were determined according to the method of Verpoorte et alCitation29. The increasing in absorbance of reaction medium was spectrophotometrically recorded at 348 nm (Shimadzu, UV-VIS Spectrophotometer, UVmini-1240, Kyoto-Japan). Also, the quantity of protein was determined at 595 nm according to Bradford methodCitation30. Bovine serum albumin was used as standard protein. The IC50 values were obtained from activity (%) versus compounds plots. For calculation of Ki values, three different concentrations were used. The Lineweaver–Burk curves were drawn and calculations were realisedCitation31.

Cytotoxicity assay

The cytotoxicity of the compounds were assayed towards human oral squamous cell carcinoma cell lines (Ca9–22, HSC-2, HSC-3, HSC-4) and human normal oral cells (HGF, HPLF, HPC) as described with some minor modificationsCitation32–37. All cells were cultured in DMEM supplemented with 10% fetalbovine serum (FBS). All test samples were dissolved in dimethylsulfoxide (DMSO). Near confluent cells were incubated in 96-microwell (Becton Dickinson, Franklin Lakes, NJ) for 48 h with 0.78, 1.56, 3.12, 6.25, 12.5, 25, 50, 100 or 200 μM of each test compound. Any cytotoxicity induced by DMSO (0.002, 0.004, 0.0078, 0.0156, 0.03125, 0.0625, 0.125, 0.25, 0.5 or 1%) were subtraced from the corresponding treated groups. The viable cell numbers were determined by the MTT method. In brief, the treated cells were incubated for another 3 h in fresh culture medium containing 0.2 mg/ml MTT. Cells were then lysed with 0.1 ml of DMSO and the absorbance at 562 nm of the cell lysate was determined using a microplate reader (Sunrise Rainbow RC-R; TECAN, Männedorf, Switzerland). CC50 (the concentrations of the compounds in micromoles which reduce the viable cell number by 50%) was determined from the dose–response curve and the mean value of CC50 for each cell type was calculated from triplicate assays.

Results and discussion

All the compounds studied here are reported for the first time with their detailed synthesis, spectral analysis, and bioactivities except 3 and 6. The synthetic pathway of the compounds are summarized in Scheme 1 and their experimental data (), spectral data (), cytotoxicities of the compounds against human oral squamous cell carcinoma cell lines and human normal oral cells (), the inhibitory potential of the compounds on hCA I and II isoenzymes () are summarized in .

Scheme 1. Synthetic pathway of the pyrazoline bearing benzenesulfonamides 1–6. Reagents and conditions. i: EtOH, aq. NaOH (10%), rt. ii: 4-Hydrazinobenzensulfonamide.HCl, EtOH, glacial CH3COOH, reflux. 1: (R1 = R4 = OCH3, R2 = R3 = H), 2: (R1 = R3 = OCH3, R2 = R4 = H), 3: (R2 = R3 = OCH3, R1 = R4 = H), 4: (R1 = R2 = R3 = OCH3, R4 = H), 5: (R1 = R3 = R4 = OCH3, R2 = H), 6: (R2 = R3 = R4 = OCH3, R1 = H).

Scheme 1. Synthetic pathway of the pyrazoline bearing benzenesulfonamides 1–6. Reagents and conditions. i: EtOH, aq. NaOH (10%), rt. ii: 4-Hydrazinobenzensulfonamide.HCl, EtOH, glacial CH3COOH, reflux. 1: (R1 = R4 = OCH3, R2 = R3 = H), 2: (R1 = R3 = OCH3, R2 = R4 = H), 3: (R2 = R3 = OCH3, R1 = R4 = H), 4: (R1 = R2 = R3 = OCH3, R4 = H), 5: (R1 = R3 = R4 = OCH3, R2 = H), 6: (R2 = R3 = R4 = OCH3, R1 = H).

Table 3. Cytotoxicities of the synthesized compounds towards human oral squamous carcinoma cell lines and human normal oral cells.

Table 4. Carbonic anhydrase inhibitory activities of the compounds on hCA I and II isoenzymes.

Structures of the compounds synthesized were established on the basis of spectral data taken from FT-IR, 1H NMR, 13C NMR and HRMS. The IR spectra of the compounds showed absorption bands in the regions 1599–1571 cm−1 corresponding to C=N stretching bands because of ring closure. Infrared spectra revealed NH2 peaks in the regions 3371–3226 cm−1. FT-IR spectra of the compounds are provided in the Supplementary file. The FT-IR values of the compounds were in accordance with literature valuesCitation23. In the 1H NMR and 13C NMR spectra of pyrazolines, chemical shift values of the three hydrogen atoms attached to the C-4 and C-5 carbon atoms of the heterocyclic ring and C-3, C-4 and C-5 carbon atoms of the pyrazolines were observed at expected ppms (See and Supplementary File).

Cytotoxic activities of the compounds were tested against four cancer cell lines (Ca9–22, HSC-2, HSC-3, HSC-4) and three non-tumor normal cells (HGF, HPLF, HPC). The cytotoxicities of the compounds were compared with reference drugs 5-Fluorouracil (5-FU) and Melphalan (). The compounds 2, 4 (2.50 times), 5 (7.16 times) and 6 (5.51 times) towards Ca9–22 cell line; the compound 6 (2.22 times) towards HSC-2 cell line, the compounds 5 (2.35 times) and 6 (5.19 times) towards HSC-3 cell line, the compounds 5 (1.95 times) and 6 (1.77 times) towards HSC-4 cell line were more cytotoxic than 5-FU. On the other hand, the compounds 2 (1.83 times), 4 (1.83 times), 5 (5.23 times), and 6 (4.03 times) towards Ca9–22 cell line, the compound 6 (1.24 times) towards HSC-2 cell line were more cytotoxic than reference compound Melphalan.

Tumor specificity (TS) of the compounds was considered by TS values. TS values were calculated by two types of calculation. Tumor-specificity (TS) value reflects the selectivity of the compounds against cancer tissues rather than normal ones. First TS calculation was made by dividing the mean CC50 value of each compound against three human oral normal cells (Column D) to the mean CC50 value against four human OSCC cell lines (Column B, ). Second TS calculation was also calculated by dividing the CC50 value of each compound against HGF cells (Column C) to the CC50 value of each compound against Ca9–22 cell line (Column A, ) since these cells are originated from the same tissue (gingiva). When TS values were considered, all the compounds had lower TS values than two reference compounds (5-FU and Melphalan) in both the calculations.

When the first TS calculation of values was considered the compound 6 had the highest TS value (1.5) while the second calculation of TS pointed out compounds 4 (TS = 3.4), 6 (TS = 2.0) and 5 (TS = 1.4) with higher TS values greater than 1. At the second TS calculation, compound 4 was the most tumor selective one. Tri-methoxy derivative compounds 4, 5, and 6 had superior tumor specificity than di-methoxy derivatives 2 and 3. It seems that addition of one methoxy group made a valuable contribution to the cytotoxic activities of the compounds in compounds 4, 5, and 6. This may result from increased possibility to form hydrogen bonding in trimethoxy derivatives.

IC50 (the drug concentration causing 50% inhibition of the desired activity) of the compounds on hCA I isoenzyme were in the range of 30.1–49.2 nM while acetazolamide (AZA) has IC50 value of 293.0 nM. It means that the compounds 16 [1 (30.1), 2 (40.8), 3 (46.2), 4 (38.5), 5 (49.2), 6 (36.5)] were 5.9–9.7 times more potent inhibitor than AZA on hCA I. On the other hand, IC50 values of the compounds on hCA II were in the range of 23.8–30.1 nM while AZA had IC50 value of 146.5 nM. This means that the compounds 16 [1 (24.7), 2 (30.1), 3 (26.7), 4 (23.8), 5 (25.7), 6 (23.9)] were 4.8–6.3 times more potent inhibitor than AZA on hCA II. When Ki values of the compounds were considered they were in the range of 26.5 ± 4.6–55.5 ± 19.4 nM towards hCA I, while they were in the range of 18.9 ± 9.0–28.8 ± 6.5 nM towards hCA II. Ki values of the reference compound AZA were 276.3 ± 98.1 nM and 117.8 ± 14.1 nM towards hCA I and hCA II, respectively.

Introduction of a methoxy group to 4 positions of 2,5-dimethoxy derivative 1 decreased both hCA I and II inhibition in compound 5, while introduction of a methoxy group to 3 positions of 2,4-dimethoxy derivative 2 decreased or had similar inhibitory effect on hCA I and II in compound 4 according to Ki values. On the other hand, introduction to a methoxy group to 5 positions of compounds 2 and 3 increased hCA I inhibitory effect in both cases in compounds 5 and 6, while it decreased the hCA II inhibitory effects in compounds 5 and 6 compared with compounds 2 and 3.

Conclusion

In conclusion; although tumor selectivity of the compounds were less than reference compounds 5-FU and Melphalan, trimethoxy derivatives compounds 4, 5, and 6 were more selective than dimethoxy derivatives 2 and 3 as judged by the cytotoxicity assay with the cells both cell types originated from the gingival tissue. Compound 6 (4-[3–(4-methoxyphenyl)-5–(3,4,5-trimethoxyphenyl)-4,5-dihydro-1H-pyrazol-1-yl] benzenesulfonamide) having the highest TS values in both calculations can be considered as the leading molecule of the series for further investigations in terms of cytotoxicity. Since all the compounds had shown superior inhibitory activity than reference compound AZA on hCA I and II isoenzymes, the compounds synthesized can be candidate compounds for further testing on other CA isoenzymes and new synthetic designs.

Declaration of interest

The authors report no conflict of interest and are responsible for the contents and writing of the paper. This study was supported by the Research Foundation of Ataturk University, Turkey. Grant Number: 2015/078 and 2015/322.

Supplementary material available online

Supplemental material

IENZ_1217852_Supplementary_Material.pdf

Download PDF (438.5 KB)

Acknowledgements

The authors thank Dr. Y. Ozkay (Anadolu University, Eskisehir, Turkey) and Dr. M. K. Sukuroglu (Gazi University, Ankara, Turkey) for HRMS analysis.

References

  • Mahapatra DK, Bharti SK, Asati V. Anti-cancer chalcones: structural and molecular target perspectives. Eur J Med Chem 2015;98:69–114.
  • Qi L, Ding Y. Potential antitumor mechanisms of phenothiazine drugs. Sci China Life Sci 2013;56:1020–7.
  • Bisi A, Meli M, Gobbi S, et al. Multidrug resistance reverting activity and antitumor profile of new phenothiazine derivatives. Bioorg Med Chem 2008;16:6474–82.
  • Supuran CT. How many carbonic anhydrase inhibition mechanisms exist? J Enzyme Inhib Med Chem 2016;31:345–60.
  • Supuran CT, Scozzafava A. Carbonic anhydrases as targets for medicinal chemistry. Bioorg Med Chem 2007;15:4336–50.
  • Kucuk M, Gulcin I. Purification and characterization of the carbonic anhydrase enzyme from Black Sea trout (Salmo trutta Labrax Coruhensis) kidney and inhibition effects of some metal ions on enzyme activity. Environ Toxicol Pharmacol 2016;44:134–9.
  • Abbate F, Casini A, Owa T, et al. Carbonic anhydrase inhibitors: E7070, a sulfonamide anticancer agent, potently inhibits cytosolic isozymes I and II, and transmembrane, tumor-associated isozyme IX. Bioorg Med Chem Lett 2004;14:217–23.
  • Supuran CT. Diuretics: from classical carbonic anhydrase inhibitors to novel applications of the sulfonamides. Curr Pharm Des 2008;14:641–8.
  • Fabrizi F, Mincione F, Somma T, et al. A new approach to antiglaucoma drugs: carbonic anhydrase inhibitors with or without NO donating moieties. Mechanism of action and preliminary pharmacology. J Enzyme Inhib Med Chem 2012;27:138–47.
  • Capasso C, Supuran CT. Anti-infective carbonic anhydrase inhibitors: a patent and literature review. Expert Opin Ther Pat 2013;23:693–704.
  • De Simone G, Supuran CT. Antiobesity carbonic anhydrase inhibitors. Curr Top Med Chem 2007;7:879–84.
  • Kumar V, Kaur K, Gupta GK, Sharma AK. Pyrazole containing natural products: synthetic preview and biological significance. Eur J Med Chem 2013;69:735–53.
  • Mojzych M, Bielawska A, Bielawski K, et al. 3-e][1,2,4]triazine sulfonamides as carbonic anhydrase inhibitors with antitumor activity. Bioorg Med Chem 2014;22:2643–7.
  • Raffa D, Maggio B, Raimondi MV, et al. Recent advanced in bioactive systems containing pyrazole fused with a five membered heterocycle. Eur J Med Chem 2015;97:732–46.
  • Kucukguzel SG, Senkardes S. Recent advances in bioactive pyrazoles. Eur J Med Chem 2015;97:786–815.
  • Li M, Zhao BX. Progress of the synthesis of condensed pyrazole derivatives (from 2010 to mid-2013). Eur J Med Chem 2014;85:311–40.
  • Maccioni E, Alcaro S, Orallo F, et al. Synthesis of new 3-aryl-4,5-dihydropyrazole-1-carbothioamide derivatives. An investigation on their ability to inhibit monoamine oxidase. Eur J Med Chem 2010;45:4490–8.
  • Gul HI, Tugrak M, Sakagami H, et al. Synthesis and bioactivity studies on new 4-(3-(4-substitutedphenyl)-3a,4-dihydro-3H-indeno[1,2-c]pyrazol-2-yl) benzenesulfonamides. J Enzyme Inhib Med Chem 2016. [Epub ahead of print]. doi: 10.3109/14756366.2016.1160077.
  • Gul HI, Kucukoglu K, Yamali C, et al. Synthesis of 4-(2-substituted hydrazinyl)benzenesulfonamides and their carbonic anhydrase inhibitory effects. J Enzyme Inhib Med Chem 2016;31:568–73.
  • Barresi E, Salerno S, Marini AM, et al. Sulfonamides incorporating heteropolycyclic scaffolds show potent inhibitory action against carbonic anhydrase isoforms I, II, IX and XII. Bioorg Med Chem 2016;24:921–7.
  • Pingaew R, Prachayasittikul V, Worachartcheewan A, et al. Novel 1,4-naphthoquinone-based sulfonamides: Synthesis, QSAR, anticancer and antimalarial studies. Eur J Med Chem 2015;103:446–59.
  • Khloya P, Celik G, SitaRam, et al. 4-Functionalized 1,3-diarylpyrazoles bearing benzenesulfonamide moiety as selective potent inhibitors of the tumor associated carbonic anhydrase isoforms IX and XII. Eur J Med Chem 2014;76:284–90.
  • Bashir R, Ovais S, Yaseen S, et al. Synthesis of some new 1,3,5-trisubstituted pyrazolines bearing benzene sulfonamide as anticancer and anti-inflammatory agents. Bioorg Med Chem Lett 2011;21:4301–5.
  • Supuran CT, Innocenti A, Mastrolorenzo A, Scozzafava A. Antiviral sulfonamide derivatives. Mini Rev Med Chem 2004;4:189–200.
  • Mete E, Comez B, Gul HI, et al. Synthesis and carbonic anhydrase inhibitory activities of new thienyl substituted pyrazoline benzensulfonamides. J Enzyme Inhib Med Chem 2016. [Epub ahead of print]. doi: 10.1080/14756366.2016.1181627.
  • Rathish IG, Javed K, Ahmad S, et al. Synthesis and antiinflammatory activity of some new 1,3,5-trisubstituted pyrazolines bearing benzene sulfonamide. Bioorg Med Chem Lett 2009;19:255–8.
  • Akincioglu A, Topal M, Gulcin I, Goksu S. Novel sulphamides and sulfonamides incorporating the tetralin scaffold as carbonic anhydrase and acetylcholine esterase inhibitors. Arch Pharm (Weinheim) 2014;347:68–76.
  • Senturk M, Gulcin I, Dastan A, et al. Carbonic anhydrase inhibitors. Inhibition of human erythrocyte isozymes I and II with a series of antioxidant phenols. Bioorg Med Chem 2009;17:3207–11.
  • Verpoorte JA, Mehta S, Edsall JT. Esterase activities of human carbonic anhydrases B and C. J Biol Chem 1967;242:4221–9.
  • Bradford MM. A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 1976;72:248–54.
  • Lineweaver H, Burk D. The determination of enzyme dissociation constants. J Am Chem Soc 1934;56:658–66.
  • Sakagami H, Shimada C, Kanda Y, et al. Effects of 3-styrylchromones on metabolic profiles and cell death in oral squamous cell carcinoma cells. Toxicol Rep 2015;2:1281–90.
  • Tugrak M, Yamali C, Sakagami H, Gul HI. Synthesis of mono Mannich bases of 2-(4-hydroxybenzylidene)-2,3-dihydroinden-1-one and evaluation of their cytotoxicities. J Enzyme Inhib Med Chem 2015;31:818–23.
  • Yerdelen KO, Gul HI, Sakagami H, Umemura N. Synthesis and biological evaluation of 1,5-bis(4-hydroxy-3-methoxyphenyl)penta-1,4-dien-3-one and its aminomethyl derivatives. J Enzyme Inhib Med Chem 2015;30:383–8.
  • Bilginer S, Gul HI, Mete E, et al. 1-(3-Aminomethyl-4-hydroxyphenyl)-3-pyridinyl-2-propen-1-ones: a novel group of tumour-selective cytotoxins. J Enzyme Inhib Med Chem 2013;28:974–80.
  • Gul HI, Yamali C, Yasa T, et al. Carbonic anhydrase inhibition and cytotoxicity studies of Mannich base derivatives of thymol. J Enzyme Inhib Med Chem 2016. [Epub ahead of print]. doi: 10.3109/14756366.2016.1140755.
  • Gul HI, Tugrak M, Sakagami H. Synthesis of some acrylophenones with N-methylpiperazine and evaluation of their cytotoxicities. J Enzyme Inhib Med Chem 2016;31:147–51.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.