Open access peer-reviewed chapter

Synthetic Strategies and Biological Activities of 1,5-Disubstituted Pyrazoles and 2,5-Disubstituted Thiazoles

Written By

Erika Lozano, Melissa M. Lewis-Bakker and Lakshmi P. Kotra

Submitted: 30 October 2022 Reviewed: 07 November 2022 Published: 27 November 2022

DOI: 10.5772/intechopen.108923

From the Edited Volume

Strategies for the Synthesis of Heterocycles and Their Applications

Edited by Premlata Kumari and Amit B. Patel

Chapter metrics overview

143 Chapter Downloads

View Full Metrics

Abstract

Pyrazoles and thiazoles belong to 5-membered aromatic heterocycles called azoles. In addition to a nitrogen, pyrazoles contain an additional nitrogen in a 1,2 linkage and thiazoles contain a sulfur atom in a 1,3 linkage. These compounds are useful pharmacophores that offer a broad range of therapeutic applications. Pyrazoles can be synthesized by (i) the condensation of 1,3 dipolar compounds and alkenes/alkynes, (b) cyclocondensation of hydrazines and dicarbonyl reagents, and (c) multi-component reactions. Access to thiazoles is typically via (a) the condensation of α-haloketones with nucleophilic thioamides containing the N-C-S fragment, (b) a reaction between α-aminonitriles and various reactants containing an X-C-S fragment, and (c) a reaction of acylaminocarbonyls and phosphorus pentasulfide. This chapter will focus on the strategies and our perspectives on the synthesis of pyrazoles and thiazoles including derivatives at the 1,5 positions and 2, 4, 5 positions respectively, reported during 2015–2022. Additionally, their therapeutic and biological evaluations will be discussed.

Keywords

  • Pyrazoles
  • 1
  • 5-subsititutions
  • thiazoles
  • 2
  • 4
  • 5-subsititutions
  • therapeutic
  • biological evaluations

1. Introduction

Heterocycles are cyclic compounds containing atoms other than carbons and hydrogens and may be synthesized or occur naturally in plants, animals and human beings. In particular, nitrogen-containing heterocycles are considered very important building blocks in the chemical and pharmaceutical industries (among others) since they can be used as catalysts, protecting groups, chiral auxiliaries and chemical intermediates.

Azoles are 5-membered nitrogen-containing aromatic heterocycles, to which thiazoles and pyrazoles belong. Pyrazoles contain two nitrogens in a 1,2 linkage while thiazoles contain nitrogen and sulfur in a 1,3 linkage. Pyrazoles and thiazoles are useful pharmacophores with a potential for various therapeutic applications.

This chapter will focus on the following as it relates to pyrazoles and thiazoles reported during 2015–2022: (i) strategies and perspectives on the synthesis of 1,5-disubstituted pyrazoles and 2,4,5-trisubstituted thiazoles derivatives and (ii) their therapeutic and biological evaluations.

Advertisement

2. Pyrazoles

2.1 Structure

The pyrazole structure exists as two tautomers due to the movement of the C-N double bond (Figure 1). When both nitrogens are unsubstituted, each has a similar reactivity. However, when any carbon or nitrogen atom is substituted, the substitution(s) influences the reactivity of the other positions of the ring. This in turn may lead to several regioisomers of the substitituted pyrazole.

Figure 1.

Tautomeric structures of an unsubstituted pyrazole.

2.2 Biological applications of Pyrazoles

Pyrazoles form core structures of many pharmaceutical products They have therapeutic properties such as antibacterial, anticancer, anti-inflammatory, anti-oxidant, anti-tuberculosis and anti-depressive agents [1, 2]. Examples of drugs containing the pyrazole moiety are Fomepizole for alcohol poisoning, Sildenafil or Viagra® for erectile dysfunction and Rimonabant for obesity treatment (Figure 2) [1].

Figure 2 .

Examples of drugs containing the pyrazole moiety.

2.3 Conventional synthesis of 1, 5-Pyrazoles

Pyrazoles are conventionally formed by (i) cycloadditions between 1,3 dipolar compounds and alkenes/alkynes and (ii) cyclocondensations of hydrazines and dicarbonyls (first reported in 1883 using dicarbonyls) as shown in Figure 3 [2]. Additionally, multi-component or one-pot reactions utilizing a combination of consisting of at least three of the afore-mentioned compounds can also generate various substituted pyrazoles. Disadvantages of these methods include the use of unstable reagents such as the diazo compounds in cycloadditions and product obtained as a mixture of regioisomers when substituted starting materials are used.

Figure 3.

Conventional reactions to form pyrazoles. A) Cycloadditions B) Cyclocondensations.

2.4 Recent synthetic routes towards 1, 5-Pyrazoles

2.4.1 Ruthenium-catalyzed Cyclocondensations of Propargyl alcohols and Hydrazines

Metal catalysts have proved to be quite efficient in organic chemistry as means of carrying out various and sometimes difficult transformations. In the synthesis of pyrazoles, the bifunctional ruthenium cyclopentadienone complex (Ru Catalyst) catalyzes the reaction between secondary propargyl alcohols and nucleophiles such as hydrazine giving 1,5-disubstituted pyrazoles along with byproducts [3]. This cascade conversion likely proceeds via cycloisomerization, Michael addition, cyclocondensation and dehydrogenation/oxidation) steps [3]. The reaction proceeds via microwave irradiation in toluene and when the nucleophile is phenyl hydrazine (2), both the 1,5- and 1,3-disubstituted regioisomers (a and b) are formed along with the 3-phenylbutene byproduct c (Figure 4) [3]. The trends in the yields indicates that when the R group of the propargyl alcohol 1 is bulky or has unsaturated side chains, the reaction favors the 1,5-disubstituted pyrazoles leading to higher yield of this regioisomer.

Figure 4.

Regioisomeric pyrazoles produced by the ruthenium catalyzed cascade conversions between secondary propargyl alcohols and phenyl hydrazine [3].

From a mechanistic perspective as shown in Figure 5, Kaufmann et al [3] reported that the propargyl alcohol is activated by the Ru catalyst to form the by the chelated π-complex I, which is then converted to the alkynyl species II. A 1,2-hydrogen shift forms the alkenyl complex III which then undergoes a regioselective nucleophilic attack by phenyl hydrazine to give complex IV. Cyclocondensation of complex IV gives the alkyl complex V which subsequently undergoes a β-hydride elimination to give the 1,5-pyrazole. The active catalyst is regenerated by the elimination of hydrogen from the hydride complex VI. The 1,3- regioisomer is formed from the hydrazone of intermediate III while the byproduct is formed when the internal carbon of the alkynyl complex II undergoes a nucleophilic attack, when non-bulky propargyl alcohols are used.

Figure 5.

Proposed mechanisms for the ruthenium catalyzed cascade conversions [3].

2.4.2 Ruthenium-catalyzed Cyclocondensation of Diazonium salts and Cyclopropanols

Another Ruthenium-catalyzed reaction to synthesize pyrazoles was reported by Cardinale et al [4]. The photocatalysis of arenediazoniums (10 and 12) and arylcyclopropanols (12 and 13) using [Ru(bpy)3]2+ under blue light irradiation in acetonitrile yielded 1, 5 -disubstituted pyrazoles 1421 (Figure 6). This photocatalysis provides several advantages such mild reaction conditions (20 mins at room temperature), compatibility with functional groups such as I, SF5, SO2NH2, N3, CN and high regioselectivity and yields.

Figure 6.

Ruthenium-catalyzed synthesis of pyrazoles under blue light. Synthetic scope of (a) diazonium salts and (B) cyclopropanols [4].

The proposed mechanism of the photocatalysis is seen in Figure 7 [4]. The reaction is proposed to be initiated when the [Ru2+]* is the oxidatively quenched by the arenediazonium salt 10 (or 12) to give [Ru3+] which oxidizes the cyclopropanol 11 (or 13) to give VII•+. The radical cation VIII•+ is formed when VII•+ undergoes a fast ring-opening and trapping with the arenediazonium salt 10 (or 12). The newly formed VIII•+ can either (i) quench the excited catalyst thereby closing the photocatalytic cycle or (ii) become involved in a radical chain process by oxidizing another molecule of 11 (or 13). As a result of the dominance of the radical chain process, the arenediazonium 10 (or 12) is used in slight excess to activate the catalyst and produce the oxidant [Ru(bpy)33+] salt. The intermediate IX then undergoes cyclization followed by loss of water to produce the pyrazole.

Figure 7.

Proposed mechanism for the ruthenium photocatalysis of pyrazoles [4].

2.4.3 Copper-catalyzed reaction of α, β-Cyanoesters and Hydrazines

In another recent report, Cu(I) catalyst, Cu(PPh3)2NO3 was used to synthesize 1,5-disubstituted pyrazoles from hydrazines 22 and α,β-unsaturated cyanoester 23 under ultrasound irradiation (Figure 8) [5]. This efficient route uses short reaction times, moderate temperatures, facile work-ups, high regioselectivity and yields [5].

Figure 8.

Synthesis of the 1,5 pyrazoles using ultrasonication and a Cu(I) catalyst [5].

From a mechanistic perspective, the authors proposed that copper coordinates easily to π-bonds and heteroatoms. This would lead to the formation of the C-N bond via an oxidative addition followed by reductive elimination, similar to that observed in Pd(0) cross coupling reactions [5].

2.4.4 Reaction of Ferrocenes and Hydrazines

Ferrocene is an organometallic compound that has excellent redox properties and may be incorporated as part of a drug’s structural moiety, leading to additional therapeutic applications. Ferrocenyl compounds have shown potential as antimalarial, anticancer, cytotoxic and DNA-cleaving agents [6]. Ferrocenyl pyrazoles have also been shown to have potential therapeutic applications as well.

Recently, ferrocenyl pyrazoles 30 and 32 were prepared from ferrocenes 29 and 31, respectively, and hydrazines in a simple two-step procedure to regioselectively give the desired 1,5-pyrazoles in good yields (Figure 9A and 9B) [7].

Figure 9.

Synthesis of ferrocenyl pyrazoles [6].

The ferrocenyl pyrazoles 30 and 32 were then evaluated in four antimicrobial assays involving two strains each of fungi (Aspergillus niger, Trichophyton rubrum) and bacteria (Staphylococcus aureus, Klebsiella pneumoniae). Standard antimicrobial agents fluconazole and neomycin were used in the fungal and bacterial studies, respectively. In these assays, the zone of inhibition (Tables 1 and 2) was measured and the corresponding minimum inhibitory concentrations (MICs) were calculated. The MIC values ranged from 85 to 95 μg/mL indicating potential as antimicrobial agents.

Bacterial strains
S. aureusKlebsiella pneumoniae
Conc. (ppm)100150200250100150200250
Zone of Inhibition (mm)3012 ± 0.516 ± 0.717 ± 0.618 ± 0.611 ± 0.513 ± 0.518 ± 0.520 ± 0.7
3212 ± 0.515 ± 0.717 ± 0.621 ± 0.610 ± 0.514 ± 0.516 ± 0.519 ± 0.7
Standards21 mm (Fluconazole)23 mm (Fluconazole)

Table 1.

Antibacterial activities of 30 and 32 [6].

Fungal strains
A. nigerTrichophyton rubrum
Conc. (ppm)100150200250100150200250
Zone of Inhibition (mm)3010 ± 0.512 ± 0.515 ± 0.518 ± 0.59 ± 0.712 ± 0.414 ± 0.518 ± 0.6
3210 ± 0.513 ± 0.516 ± 0.518 ± 0.59 ± 0.712 ± 0.415 ± 0.517 ± 0.6
Standards24 mm (Fluconazole)9 mm (Fluconazole)

Table 2.

Antifungal activities of 30 and 32 [6].

DNA photo-cleavage activity of the ferrocenyl pyrazoles was also evaluated using super coiled plasmid DNA by gel electrophoresis [6]. When the faster super-coiled DNA (form I) was treated with the pyrazoles, it converted to the slower-moving nicked DNA (form II), in comparison to the untreated normal DNA. These results confirmed the DNA cleavage activity of both ferrocenyl pyrazoles.

2.4.5 C5-electrophilic fluorination of Pyrazoles

The 5-fluoropyrazole core is an important structural moiety in the agrochemical and pharmaceutical industries. Synthesis of 5-fluoropyrazoles typically occurs by reaction of (i) 1,3-dicarbonyl containing CF3/CF2 (fluorinated synthon) with hydrazines followed by HF-elimination [8]; (ii) copper-catalyzed click reaction of fluorosydnones with alkynes [9]; (iii) Selectfluor and pyrazoles containing carboxylic acids followed by decarboxylation [10] and (iv) KF with pyrazoles [11, 12]. The electrophilic fluorination of pyrazoles to give 5-fluoropyrazoles was recently described using N-fluorobenzenesulfonimide (NFSI) [13]. As shown in Figure 10 1-substituted pyrazoles were subjected to C5-deprotonation by lithium base, which was then fluorinated using NFSI to yield 1-substituted-5-fluoropyrazoles [13].

Figure 10.

Synthesis of 1-substituted-5-fluoropyrazoles [13].

Advertisement

3. Thiazoles

3.1 Structure

Thiazoles are five-membered aromatic heterocycles belonging to the azole group. While azoles are generally characterized by the presence of a nitrogen atom, the thiazole ring features the N in a 1,3-linkage with sulfur. The sulfur atom bears a lone pair of electrons which are delocalized throughout the ring, while C2 bears an acidic proton allowing for a range of reactions to occur in this position (Figure 11) [14].

Figure 11.

Resonance of unsubstituted 1,3-thiazole.

3.2 Biological applications of Thiazoles

Thiazoles are noted for their utility in medicinal chemistry as an active and often potent pharmacophore [15] and are investigated for their therapeutical potential. Thiazoles are found in biologically relevant compounds such as the natural product Thiamin or Vitamin B1 and the first effective antibiotic drug series, Penicillin (Figure 12).

Figure 12.

Biologically relevant thiazole derivatives.

Additionally, there has been a growing interest in the synthesis of thiazole-containing compounds for applications as photosensitizers, sensors, catalysts, pigments, and more [16].

Certain biological applications of thiazoles were first identified in 1887 by Hantzsch and Weber. Since this discovery, thiazoles are a notable moiety present in several modern compounds of interest for their antitubercular [17], antidiabetic [18], antimalarial [19], antibacterial [19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31], antiviral [26, 32, 33, 34], antifungal [22, 24, 25, 26, 27, 29, 30, 33, 34], antioxidant [35], anti-inflammatory [30, 35], anti-cancer [36, 37], and anti-proliferative [27, 38, 39, 40], activities.

Figure 13 represents a few examples of approved drugs that contain a thiazole ring. Epothilones are a class of anti-neoplastic agents first identified in 1995, which act by stabilizing microtubules and inducing apoptosis [41, 42]. Ritonavir was discovered in 1998 as a potent and effective HIV protease inhibitor [43] and today, is used in combination with other antiretroviral agents in the treatment of HIV infection. More recently, it has been used in combination therapy with lopinavir to treat severe Covid-19 [44, 45]. The prodrug Cefpodoxime Proxetil was identified in 1993 as a safe and effective broad-spectrum antibiotic [46, 47, 48]. The thiazole moiety is unquestionably a vital functionality in the structure of many drugs.

Figure 13.

Approved drugs bearing thiazole moieties.

3.3 Conventional synthesis of 2, 4, 5-Thiazoles

The synthesis of thiazoles has been broadly described in literature over the years. The main methods used to prepare thiazoles are the Hantzsch, Cook-Heilbron and Gabriel’s syntheses as shown in Figure 14 [16].

Figure 14.

General synthetic scheme of 2,4,5-subsituted thiazoles by a) Hantzsch’s synthesis B) Cook-Heilbron’s synthesis and C) Gabriel’s synthesis [16].

Hantzsch’s synthesis of thiazole rings was first published in 1887 by Hantzsch and Weber [49]. Hantzsch’s synthesis involves the condensation and aromatization of α-haloketones with nucleophilic thioamides containing the N-C-S fragment, where cyclization yields the thiazole moiety [16].

The Cook-Heilbron reaction involves the interaction of aminonitrile derivatives with esters or various reactants containing a X-C-S fragment (such as dithioacids, carbon disulfide) under mild conditions to yield aminothiazoles [16, 50, 51, 52, 53]. This synthesis generally introduces different moieties to the 2-position of 4- or 5-aminothiazoles.

Gabriel’s synthesis was introduced in 1910 and describes the reaction between acylaminocarbonyls and phosphoruspentasulfide (such as Lawesson’s reagent) to yield a thiazole ring with phenyl and alkyl substitutions in the 2- and 5-positions [16, 54].

3.4 Recent synthetic routes towards 2,4,5-substituted thiazoles

3.4.1 Hantzsch’s synthesis from Thioamide andα-Halocarbonyls

Figure 15 describes the synthesis of MSI-1 (3-(5-isopropyl-4-(4-methylpyridin-3-yl) thiazol-2-yl) benzamide), a natural product monomer which acts as a SREBP-1 inhibitor in the treatment for lung squamous cell carcinoma (LUSC). It also enhances the sensitivity of these cancer cells to antitumor agents. Compound 40 was brominated to obtain 41 and in a parallel reaction, the thioamide derivative 43 was generated from 42. Subsequently, MSI-1 was obtained from the cyclocondensation reaction between α -halocarbonyl 41 and thioamide derivative 43 [55].

Figure 15.

Hantszch’s reaction between α-halocarbonyl 41 and thioamide 43 [55].

The reaction mechanism may proceed as shown in Figure 16 as described by Hantzsch A and Weber [49], where bromine of 45 acts as a leaving group of allowing the coupling of sulfur of 46 to the α-position of the carbonyl. Following a series of several proton transfers, cyclization occurs via the nucleophilic attack of nitrogen to the electron-deficient carbonyl carbon and the desired product 47 is obtained after elimination of water [55].

Figure 16.

Plausible mechanism of Hantzsch’s reaction [49].

Molecular docking studies show that MSI-1 enters the hydrophobic pocket of SREBP-1, binding through π-π conjugation. The complex is stabilized by further π-π interactions with three amino acid residues PHE271, TYR335, and PHE349. MSI-1 was shown to impedes the activation of SREBP-1 by inhibiting downstream genes of SREBP-1 associated to lipid metabolism in a dose dependent manner. Additionally, MSI-1 inhibits the Warburg Effect of cancerous and malignant cells and the Epithelial-Mesenchymal Transition process (indicative of chemo-resistance) in LUSC cell line NCI-H226. This effect was demonstrated by a decrease in glucose uptake and lactate production, as well as a reduction in ATP production and LDH activation [55].

3.4.2 Hantzsch’s synthesis from Thiourea andα-Halocarbonyls

Wang et al recently reported the synthesis of thiazole-naphthalene hybrids and their antiproliferative activities as tubulin polymerization inhibitors from thiourea and α-halocarbonyls (Figure 17) [56]. The condensation of 1-methoxynaphtalene 48 with phenylacetic acid 49 in presence of trifluoroacetic anhydride (TFAA) in trifluoroacetic acid (TFA) yielded deoxybenzoin 50. Compound 50 was then treated with pyridinium tribromide (PyBr3) in CH2Cl2 to brominate the α-position of the carbonyl. Finally, under reflux in ethanol, the cyclocondensation reaction of 51 and thiourea 52 produced desired compound 53. Three thiazole-naphthalene derivatives were prepared with this protocol. Moreover, fourteen compounds were generated by introducing substitutions to the amine in the presence of acid anhydride [56].

Figure 17.

Hantszch reaction between α-halocarbonyl 51 and thiourea 52 [56].

Compound 53 showed potent antiproliferative activity on the human breast cancer (MCF-7) and human lung adenocarcinoma (A549) cell lines when compared to standard treatments (cisplatin, 5-fluorouracil, tamoxifen, and CA-4). They determined that the 4-ethoxyphenyl substitution was more favorable than 4-methoxyphenyl. Additionally, replacement with 2-bromo-3,4,5-trimethoxyphenyl reduced the antiproliferative activity [56]. Compound 53 exhibited low toxicity in human normal cell line (IC50 = 16.37 ± 4.61 μM). Its in vitro tubulin polymerization inhibitory activity of was investigated and revealed that the compound acts as a tubulin destabilizing agent with an IC50 of 3.3 μM (compared to reference colchicine IC50 of 9.1 μM). Furthermore, it was demonstrated that compound 53 leads to cell cycle arrest at the metaphase in a dose-dependent manner, and an Annexin V-FITC/PI assay showed that it can effectively induce apoptosis in MCF-7 cells. Molecular docking studies revealed that 53 can bind with tubulin by adopting an “L-shaped” conformation. The naphthalene moiety can accommodate in the hydrophobic pocket of the protein, and it binds to the colchicine site of tubulin [56].

3.4.3 Holzapfel: Meyers: Nicolaou modification of Hantzsch’s synthesis

The Holzapfel-Meyers-Nicolaou modification is based on the Hantzsch reaction between thioamide and an α-halocarbonyl, however, it involves the generation of a hydroxythiazoline intermediate under basic conditions. This intermediate is then dehydrated in the presence of trifluoroacetic anhydride (TFAA) and pyridine, followed by the addition of triethylamine (TEA) to yield the desired thiazole [57].

Figure 18 shows a recent example of the Holzapfel-Meyers-Nicolau modification reported in the synthesis of 5-acylamino-1,3-thiazoles from α-chloroglycinates and thioamide derivatives [40]. ESI-MS monitoring revealed the presence of the hydroxy intermediate X. Dehydration of X results in the desired 5-acylaminothiazole derivative 56 in a high yield (87%). Phenylic substitutions were introduced successfully to the 2 and 4-positions using this approach [58].

Figure 18.

Holzapfel-Meyers-Nicolau reaction between α-chloroglycinate 54 and thioamide 55 [58].

3.4.4 Cook-Heilbron’s synthesis

Avadhani et al developed an efficient one-pot reaction for the preparation of 4-amino 2-aryl-5-substituted thiazoles. Figure 19 presents the reaction of cyanamide 57 with dithioester 58a or 58b with excess NaH, followed by the addition of halo-carbonyl derivative 59 to yield potent antiproliferative agents 4-amino-2-phenyl and 2-(thiophne-3-yl)- 5-(2,3,4-trimethoxybenzoyl)-thiazoles 60a and 60b in high yields (77% and 81% respectively). The Thorpe-Ziegler-type reaction proceeds via base-mediated intermolecular cyclization of N-cyanothioimidate intermediate XI [59].

Figure 19.

Cook-Heilbron’s reaction between cyanamide 57 and dithioate derivative 58 [59].

The 2-methylthio and arylamino substitutions can be introduced at the 2-position using this method, while the 5-position supports ester, nitrile, and carbonyl substituents. The latter can be utilized to introduce reactive functionalities, expanding the possible range of substitutions achieved by this protocol. The novel methodology was further validated by the synthesis of 26 4-amino-2-(het)aryl/alkyl 5-substituted thiazole derivatives [59]. It should be noted that this example introduces the amino moiety to the 4-position. The 5-aminothiazoles can be synthesized from the reaction with 2-aminoacetonitrile instead of cyanamide.

3.4.5 Lactic acid-mediated one-pot reaction

Figure 20 shows the one-pot Hantzsch synthesis of the 2-aminothiazole 63 using lactic acid as a green solvent and catalyst [60]. The ketone 61 was brominated in-situ followed by heterocyclization with thiourea 52 to produce the substituted aminothiazole 63. Lactic acid was selected as a solvent and catalyst due to its ability to solubilize all the reactants, its increased product yield and shorter reaction times in comparison to acetic acid. Maximum yields of 2-aminothiazoles (up to 96%) were obtained when the temperature was increased from room temperature (45% in 1.2 h) to 90–100°C [60]. Lower yields were reported yields when strong deactivating groups were introduced (such as -NO2), likely due to the reduced in-situ formation of the α-brominated ketone. Overall, this work developed an effective, rapid, and sustainable one-pot synthesis of 2-aminothiazoles in excellent yields [60].

Figure 20.

Lactic acid-mediated one-pot synthesis of 2-aminothiazoles [60].

3.4.6 Aluminum oxide/PVA thin film-catalyzed reaction

The catalytic activity of hybrid PVA/Al2O3 nanofilms in the synthesis of thiazole derivatives was recently reported by Riyadh et al [61]. The reaction between the thiosemicarbazone 2-benzylidenehydrazine 64 and the α-haloester ethyl-2-chloro-3-oxobutanoate 65 in the presence of the PVA/Al2O3 nanocomposite as a basic catalyst is demonstrated in Figure 21. Under thermal conditions, the optimal loading catalyst was 10 wt% and the desired compounds were obtained after 180 min [61].

Figure 21.

Polyvinyl alcohol/aluminum oxide (PVA/Al2O3) thin film nanocomposite as a catalyst in the synthesis of 2,4,5-thiazoles [61].

Figure 22 describes a plausible reaction mechanism where the Al2O3 particles act as a base in the deprotonation of the thiol group from carbothioamide tautomeric intermediate XVIII. The thiolate anion XIX then attacks α-halocarbonyl 65, displacing -Cl and generating the intermediate XX. The cyclocondensation of XX yielded the desired thiazole derivatives. The catalyst was recycled three times and recovered in excellent yields (90%) [61].

Figure 22.

Proposed mechanism of PVA/Al2O3-catalyzed reaction [61].

Advertisement

4. Conclusion

This chapter reviewed novel synthetic methods involving the presence of catalysts, solvent-free conditions, and green chemistry approaches reported during 2015–2022, for the preparation of pyrazoles and thiazoles. Additionally, their therapeutic and biological evaluations were discussed. These structural moieties will continue to play an important role in therapeutic and other applications, therefore further development, versatility and diversification of their syntheses will continue to be of great interest to chemists worldwide.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Ansari A, Ali A, Shamsuzzaman AM. Review: Biologically active pyrazole derivatives. New England Journal of Chemistry. 2017;41:16-41. DOI: 10.1039/C6NJ03181A
  2. 2. Karrouchi K, Radi S, Ramli Y, Taoufik J, Mabkhot YN, Al-aizari FA, et al. Synthesis and pharmacological activities of Pyrazole derivatives: A review. Molecules. 2018;23:134. DOI: 10.3390/molecules23010134
  3. 3. Kaufmann J, Jäckel E, Haak E. Ruthenium-catalyzed formation of pyrazoles or 3-hydroxynitriles from propargyl alcohols and hydrazines. ARKIVOC. 2019;iv:91-101. DOI: 10.24820/ark.5550190.p010.893
  4. 4. Cardinale L, Neumeier M, Majek M, Jacobi von Wangelin A. Aryl Pyrazoles from photocatalytic cycloadditions of Arenediazonium. Organic Letters. 2020;22:7219-7224. DOI: 10.1021/acs.orglett.0c02514
  5. 5. Pise AS, Bururngale AS, Devkate SS. Ultrasound assisted synthesis of 1,5-Disubstituted Pyrazole using Cu(I) catalyst. Asian Journal of Chemistry. 2020;32:575-579. DOI: 10.14233/ajchem.2020.22432
  6. 6. Patra M, Gasser G. The medicinal chemistry of ferrocene and its derivatives. Nature Reviews Chemistry. 2017;1:0066. DOI: 10.1038/s41570-017-0066
  7. 7. Kumar M, Sharma S, Singh Tuli H, Khare R, Parkash V. A novel synthesis, characterization and biological studies of Ferrocenyl substituted Pyrazoles. Asian Journal of Chemistry. 2019;31:2729-2732. DOI: 10.14233/ajchem.2019.22161
  8. 8. Ohtsuka Y, Uraguchi D, Yamamoto K, Tokuhisa K, Yamakawa T. Syntheses of 2-(trifluoromethyl)-1,3-dicarbonyl compounds through direct trifluoromethylation with CF3I and their application to fluorinated pyrazoles syntheses. Tetrahedron. 2012;68:2636-2649. DOI: 10.1016/j.tet.2012.01.075
  9. 9. Liu H, Audisio D, Plougastel L, Decuypere E, Buisson DA, Koniev O, et al. Ultrafast click chemistry with Fluorosydnones. Angewandte Chemie, International Edition. 2016;55:12073-12077. DOI: 10.1002/anie.201606495
  10. 10. Yuan X, Yao JF, Tang ZY. Decarboxylative fluorination of electron-rich Heteroaromatic carboxylic acids with Selectfluor. Organic Letters. 2017;19:1410-1413. DOI: 10.1021/acs.orglett.7b00335
  11. 11. Neeff A, Pazenok S. Method for the production of 5-fluoro-1,3-dialkyl-1h-pyrazol-4-carbonyl fluorides. 2009
  12. 12. Pazenok S, Lui N, Gary S. Process for the preparation of 5-Fluoro-1-alkyl-3-fluoroalkyl-1H-pyrazole-4-carbonyl chlorides and fluorides. 2011
  13. 13. Levchenko V, Dmytriv YV, Tymtsunik AV, Liubchak K, Rudnichenko A, Melnyk AV, et al. Preparation of 5-Fluoropyrazoles from Pyrazoles and N-Fluorobenzenesulfonimide (NFSI). The Journal of Organic Chemistry. 2018;83:3265-3274. DOI: 10.1021/acs.joc.8b00199
  14. 14. Arshad MF, Alam A, Alshammari AA, Alhazza MB, Alzimam IM, Alam MA, et al. Thiazole: A versatile standalone moiety contributing to the development of various drugs and biologically active agents. Molecules. 2022;27:3994. DOI: 10.3390/molecules27133994
  15. 15. Ali SH, Sayed AR. Review of the synthesis and biological activity of thiazoles. Synthetic Communications. 2021;51:670-700. DOI: 10.1080/00397911.2020.1854787
  16. 16. Ibrahim SA, Rizk HF. Synthesis and biological evaluation of thiazole derivatives. In: Kuznetsov A, editor. Azoles. Rijeka, London, UK: IntechOpen; 2020. p. Ch. 2. DOI: 10.5772/intechopen.93037
  17. 17. Omar MA, Masaret GS, Abbas EMH, Abdel-Aziz MM, Harras MF, Farghaly TA. Novel anti-tubercular and antibacterial based benzosuberone-thiazole moieties: Synthesis, molecular docking analysis, DNA gyrase supercoiling and ATPase activity. Bioorganic Chemistry. 2020;104:104316. DOI: 10.1016/j.bioorg.2020.104316
  18. 18. Li Z, Xu X, Hou J, Wang S, Jiang H, Zhang L. Structure-based optimization of free fatty acid receptor 1 agonists bearing thiazole scaffold. Bioorganic Chemistry. 2018;77:429-435. DOI: 10.1016/j.bioorg.2018.01.039
  19. 19. Vekariya RH, Patel KD, Vekariya MK, Prajapati NP, Rajani DP, Rajani SD, et al. Microwave-assisted green synthesis of new imidazo [2,1-b] thiazole derivatives and their antimicrobial, antimalarial, and antitubercular activities. Research on Chemical Intermediates. 2017;43:6207-6231. DOI: 10.1007/s11164-017-2985-5
  20. 20. Eryılmaz S, Türk Çelikoğlu E, İdil Ö, İnkaya E, Kozak Z, Mısır E, et al. Derivatives of pyridine and thiazole hybrid: Synthesis, DFT, biological evaluation via antimicrobial and DNA cleavage activity. Bioorganic Chemistry. 2020;95:103476. DOI: 10.1016/j.bioorg.2019.103476
  21. 21. Lu Q , Yu Q , Zhu Y, Weng J, Yuan J, Hu D, et al. Novel stilbene analogues containing thiazole moiety: Synthesis, biological evaluation and docking study. Journal of Molecular Structure. 2019;1180:780-786. DOI: 10.1016/j.molstruc.2018.12.068
  22. 22. Eissa SI, Farrag AM, Abbas SY, El Shehry MF, Ragab A, Fayed EA, et al. Novel structural hybrids of quinoline and thiazole moieties: Synthesis and evaluation of antibacterial and antifungal activities with molecular modeling studies. Bioorganic Chemistry. 2021;110:104803. DOI: 10.1016/j.bioorg.2021.104803
  23. 23. Reddy GM, Garcia JR, Reddy VH, de Andrade AM, Camilo A, Pontes Ribeiro RA, et al. Synthesis, antimicrobial activity and advances in structure-activity relationships (SARs) of novel tri-substituted thiazole derivatives. European Journal of Medicinal Chemistry. 2016;123:508-513. DOI: 10.1016/j.ejmech.2016.07.062
  24. 24. Nastasă C, Tiperciuc B, Duma M, Benedec D, Oniga O. New hydrazones bearing thiazole scaffold: Synthesis, characterization, antimicrobial, and antioxidant investigation. Molecules. 2015;20:17338. DOI: 10.3390/molecules200917325
  25. 25. Rajagopal K, Dhandayutham S, Nandhagopal M, Narayanasamy M, Elzagheid I, Rhyman ML, et al. Thiazole derivatives: Synthesis, characterization, biological and DFT studies. Journal of Molecular Structure. 2022;1255:132374. DOI: 10.1016/j.molstruc.2022.132374
  26. 26. Farghaly TA, Althagafi I, Ibrahim MH, Al-Qurashi NT, Farooq U. Synthesis under microwaves irradiation, structure elucidation, docking study for inhibiting COVID-19 and DFT calculations of novel azoles incorporated indole moiety. Journal of Molecular Structure. 2021;1244:131263. DOI: 10.1016/j.molstruc.2021.131263
  27. 27. Bera P, Aher A, Brandao P, Manna SK, Mondal G, Jana A, et al. Induced apoptosis against U937 cancer cells by fe(II), co(III) and ni(II) complexes with a pyrazine-thiazole ligand: Synthesis, structure and biological evaluation. Polyhedron. 2020;182:114503. DOI: 10.1016/j.poly.2020.114503
  28. 28. Althagafi I, El-Metwaly N, Farghaly TA. New series of thiazole derivatives: Synthesis, structural elucidation, antimicrobial activity, molecular modeling and MOE docking. Molecules. 2019;24:1741. DOI: 10.3390/molecules24091741
  29. 29. Blaja SP, Lungu LV, Kuchkova KI, Ciocarlan AG, Barba AN, Vornicu N, et al. Norlabdane compounds containing thiosemicarbazone or 1,3-thiazole fragments: Synthesis and antimicrobial activity. Chemistry of Natural Compounds. 2021;57:101-110. DOI: 10.1007/s10600-021-03292-3
  30. 30. Nadaf AA, Barretto DA, Najare MS, Mantur S, Garbhagudi M, Gaonkar S, et al. Design, synthesis and biological evaluation of novel N,4-diphenylthiazol-2-amine derivatives. Medicinal Chemistry Research. 2020;29:442-458. DOI: 10.1007/s00044-019-02495-2
  31. 31. Makam P, Kannan T. 2-aminothiazole derivatives as antimycobacterial agents: Synthesis, characterization, in vitro and in silico studies. European Journal of Medicinal Chemistry. 2014;87:643-656. DOI: 10.1016/j.ejmech.2014.09.086
  32. 32. Sokolova AS, Yarovaya OI, Bormotov NI, Shishkina LN, Salakhutdinov NF. Synthesis and antiviral activity of camphor-based 1,3-thiazolidin-4-one and thiazole derivatives as orthopoxvirus-reproduction inhibitors. Medicinal Chemistry Communications. 2018;9:1746-1753. DOI: 10.1039/C8MD00347E
  33. 33. Farghaly TA, Alsaedi AMR, Alenazi NA, Harras MF. Anti-viral activity of thiazole derivatives: An updated patent review. Expert Opinion on Therapeutic Patents. 2022;32:791-815. DOI: 10.1080/13543776.2022.2067477
  34. 34. Stankova I, Chuchkov K, Chayrov R, Mukova L, Galabov A, Marinkova D, et al. Adamantane derivatives containing thiazole moiety: Synthesis, antiviral and antibacterial activity. International Journal of Peptide Research and Therapeutics. 2020;26:1781-1787. DOI: 10.1007/s10989-019-09983-4
  35. 35. Ramagiri RK, Vedula RR, Thupurani MK. A facile one-step multi-component approach toward the synthesis of 3-(2-amino-4-thiazolyl) coumarins by using trimethylsilyl isothiocyanate and their antioxidant and anti-inflammatory activity. Phosphorus, Sulfur, and Silicon and the Related Elements. 2015;190:1393-1397. DOI: 10.1080/10426507.2014.990016
  36. 36. Ramalingam A, Sarvanan J. Synthesis, docking and anti-cancerous activity of some novel thiazole derivatives of biological interest. IJPI. 2020;10:594-603. DOI: 10.5530/ijpi.2020.4.104
  37. 37. Coman F, Mbaveng AT, Leonte D, Bencze LC, Vlase L, Imre S, et al. Heterocycles 44. Synthesis, characterization and anticancer activity of new thiazole ortho-hydroxychalcones. Medicinal Chemistry Research. 2018;27:1396-1407. DOI: 10.1007/s00044-018-2156-2
  38. 38. Łączkowski KZ, Anusiak J, Świtalska M, Dzitko K, Cytarska J, Baranowska-Łączkowska A, et al. Synthesis, molecular docking, ctDNA interaction, DFT calculation and evaluation of antiproliferative and anti-toxoplasma gondii activities of 2,4-diaminotriazine-thiazole derivatives. Medicinal Chemistry Research. 2018;27:1131-1148. DOI: 10.1007/s00044-018-2136-6
  39. 39. Guerrero-Pepinosa NY, Cardona-Trujillo MC, Garzón-Castaño SC, Veloza LA, Sepúlveda-Arias JC. Antiproliferative activity of thiazole and oxazole derivatives: A systematic review of in vitro and in vivo studies. Biomedicine & Pharmacotherapy. 2021;138:111495. DOI: 10.1016/j.biopha.2021.111495
  40. 40. Spanò V, Attanzio A, Cascioferro S, Carbone A, Montalbano A, Barraja P, et al. Synthesis and antitumor activity of new thiazole nortopsentin analogs. Marine Drugs. 2016;14:226. DOI: 10.3390/md14120226
  41. 41. Forli S. Epothilones: From discovery to clinical trials. Current Topics in Medicinal Chemistry. 2014;14:2312-2321. DOI: 10.2174/1568026614666141130095855
  42. 42. Bollag DM. Epothilones: Novel microtubule-stabilising agents. Expert Opinion on Investigational Drugs. 1997;6:867-873. DOI: 10.1517/13543784.6.7.867
  43. 43. Kempf DJ, Sham HL, Marsh KC, Flentge CA, Betebenner D, Green BE, et al. Discovery of ritonavir, a potent inhibitor of HIV protease with high oral bioavailability and clinical efficacy. Journal of Medicinal Chemistry. 1998;41:602-617. DOI: 10.1021/jm970636+
  44. 44. Hung Y, Lee J, Chiu C, Lee C, Tsai P, Hsu I, et al. Oral nirmatrelvir/ritonavir therapy for COVID-19: The dawn in the dark? Antibiotics. 2022;11:220. DOI: 10.3390/antibiotics11020220
  45. 45. Cao B, Wang Y, Wen D, Liu W, Wang J, Fan G, et al. A trial of Lopinavir–ritonavir in adults hospitalized with severe covid-19. New England Journal of Medicine. 2020;382:1787-1799. DOI: 10.1056/NEJMoa2001282
  46. 46. Chugh K, Agrawal S. Cefpodoxime: Pharmacokinetics and therapeutic uses. The Indian Journal of Pediatrics. 2003;70:227-231. DOI: 10.1007/BF02725589
  47. 47. Chocas EC, Paap CM, Godley PJ. Cefpodoxime proxetil: A new, broad-spectrum, oral cephalosporin. The Annals of Pharmacotherapy. 1993;27:1369-1377. DOI: 10.1177/106002809302701111
  48. 48. Borelli C, Schaller M, Niewerth M, Nocker K, Baasner B, Berg D, et al. Modes of action of the new arylguanidine abafungin beyond interference with ergosterol biosynthesis and in vitro activity against medically important fungi. Chemotherapy. 2008;54:245-259. DOI: 10.1159/000142334
  49. 49. Hantzsch A, Weber JH. Ueber verbindungen des thiazols (pyridins der thiophenreihe). Ber. Dtsch. Chem.Ges. 1887;20:3118-3132. DOI: 10.1002/cber.188702002200
  50. 50. Cook AH, Heilbron I, Levy AL. Studies in the azole series. Part II. The interaction of α-amino-nitriles and carbon disulphide. Journal of the Chemical Society. 1947;2:1598-1609. DOI: 10.1039/JR9470001598
  51. 51. Cook AH, Heilbron I, Levy AL. Studies in the azole series. Part I. a novel route to 5-aminothiazoles. Journal of the Chemical Society. 1947;1:1594-1598. DOI: 10.1039/JR9470001594
  52. 52. Cook AH, Heilbron I, Stern ES. 412. Studies in the azole series. Part X. some 5-amino-2-mercapto-4-alkylthiazoles and 2: 4-dithio-5-alkylhydantions. Journal of the Chemical Society. 1948;1948:2031-2033. DOI: 10.1039/JR9480002031
  53. 53. Capp CW, Cook AH, Downer JD, Heilbron I. 268. Studies in the azole series. Part VIII. The interaction of α-amino-nitriles and carbethoxyisothiocyanate. Journal of the Chemical Society. 1948:1340-1345. DOI: 10.1039/JR9480001340
  54. 54. Vernin G. General synthetic methods for thiazole and thiazolium salts. In: Metzger JV, editor. Chemistry of Heterocyclic Compounds. New Jersey, USA: Wiley; 1979. pp. 165-335. DOI: 10.1002/9780470187081.ch3
  55. 55. Ma D, Liu X, Jia H, Zhang Y, Jiang Q , Sun H, et al. A novel small-molecule inhibitor of SREBP-1 based on natural product monomers upregulates the sensitivity of lung squamous cell carcinoma cells to antitumor drugs. Frontiers in Pharmacology. 2022;13:895744. DOI: 10.3389/fphar.2022.895744
  56. 56. Wang G, Liu W, Fan M, He M, Li Y, Peng Z. Design, synthesis and biological evaluation of novel thiazole-naphthalene derivatives as potential anticancer agents and tubulin polymerisation inhibitors. Journal of Enzyme Inhibition and Medicinal Chemistry. 2021;36:1693-1701. DOI: 10.1080/14756366.2021.1958213
  57. 57. Merritt EA, Bagley MC. Holzapfel-meyers-nicolaou modification of the hantzsch thiazole synthesis. Synthesis. 2007;2007:3535-3541. DOI: 10.1055/s-2007-990851
  58. 58. Tomassetti M, Lupidi G, Piermattei P, Rossi FV, Lillini S, Bianchini G, et al. Catalyst-free synthesis of polysubstituted 5-acylamino-1,3-thiazoles via hantzsch cyclization of α-chloroglycinates. Molecules. 2019;24:3846. DOI: 10.3390/molecules24213846
  59. 59. Avadhani A, Iniyavan P, Kumar Y, Ila H. Single-pot preparation of 4-amino-2-(het)aryl-5-substituted thiazoles employing functionalized dithioesters as thiocarbonyl precursors. The Journal of Organic Chemistry. 2021;86:8508-8515. DOI: 10.1021/acs.joc.1c00616
  60. 60. Bodireddy MR, Khaja Mohinuddin PM, Gundala TR, Gangi Reddy NC. Lactic acid-mediated tandem one-pot synthesis of 2-aminothiazole derivatives: A rapid, scalable, and sustainable process. Cogent Chemistry. 2016;2:1154237. DOI: 10.1080/23312009.2016.1154237
  61. 61. Riyadh SM, Khalil KD, Bashal AH. Structural properties and catalytic activity of binary poly (vinyl alcohol)/Al2O3 nanocomposite film for synthesis of thiazoles. Catalysts. 2020;10:100. DOI: 10.3390/catal10010100

Written By

Erika Lozano, Melissa M. Lewis-Bakker and Lakshmi P. Kotra

Submitted: 30 October 2022 Reviewed: 07 November 2022 Published: 27 November 2022