Subscribe to RSS

DOI: 10.1055/a-2728-8329
Oxazole Analogues as Potential Therapeutic Agents: A Comprehensive Review
Authors
Funding None.

Abstract
Oxazole, a five-membered heterocyclic motif composed of three carbon atoms, one oxygen atom, and one nitrogen atom with one carbon atom between the oxygen and nitrogen atoms, has attained remarkable notice in medicinal chemistry because of its diverse and potent biological effects. The unique structural features of oxazole make it a valuable scaffold for the generation of novel therapeutic agents. Over the years, oxazole derivatives have demonstrated a wide range of therapeutic actions, including anti-inflammatory, antitumor, antidepressant, analgesic, antidiabetic, diuretic, antibacterial, and anticonvulsant effects. These biological activities make them promising candidates for the treatment of various diseases. As a result, there has been an increasing interest in synthesizing new oxazole-based compounds with enhanced efficacy and selectivity for therapeutic applications. The present review aimed to provide a comprehensive overview of the chemical characteristics of oxazole and its congeners, highlighting their biological roles and potential for drug development. In addition, structure–activity relationship analysis explored the key substituents that favor the pharmacological potential against a range of clinical disorders. By consolidating current knowledge on the therapeutic potential of oxazole, this review seeks to stimulate further research and innovation in the pharmaceutical industry, material science, and academia, with the goal of addressing unmet medical needs.
Introduction
The history of medicine is intertwined with the history of disease, as humans have long sought to understand and combat various illnesses.[1] From the earliest times, medicines were derived from natural sources, such as plants, animals, and minerals. However, the limited effectiveness and potential toxicity of these natural remedies underscored the need for new, more effective drugs.[1] [2]
Among these, heterocyclic compounds, particularly those with five-membered rings like oxazole, thiazole, oxadiazole, and triazine, have gained prominence due to their diverse pharmacological and therapeutic activities.[3] [4] Oxazole, a heterocyclic aromatic compound as depicted in [Fig. 1], is characterized by a five-membered ring structure composed of three carbon atoms, one oxygen atom, and one nitrogen atom, with one carbon atom between the oxygen and nitrogen atoms.[5] First synthesized in 1962, oxazole gained particular attention during World War II, when it was speculated that penicillin contained an oxazole ring system. The compound was eventually characterized through Diels–Alder reactions as well as 1,3-dipolar cycloaddition methodologies, leading to the discovery of its mesoionic heterocyclic structure.[6] Due to its unique chemical structure, oxazole and its analogues presented a wide range of therapeutic effects such as antibacterial, anti-inflammatory, antifungal, anti-inflammatory, anticancer, antidiabetic, and antiepileptic effects. These attributes make oxazole an important scaffold to design and generate novel therapeutic candidates, contributing significantly to the treatment of various diseases.[7] [8]


Properties of Oxazole
Oxazole is a five-membered heterocyclic compound with a chemical formula of C3H3NO and a molecular mass of 69.06 g/mol. Oxazole typically exists as a colorless to pale yellow liquid at room temperature (boiling point 69–70°C) and has a density of 1.050 g/mL. The synonym name of oxazole is 3-azafuran. It has a relatively low acidity, with a pKa of 0.8 at 33°C for its conjugate acid, and a basicity with a pKb of 13.20. Oxazole undergoes phase transitions between solid, liquid, and gas phases.[9] The compound exhibits notable nuclear magnetic resonance (NMR) and the 1H NMR chemical shifts appear at downfield (δ 7–8) due to the strong deshielding effect of the aromatic ring. Its vapor pressure is 132.01 mm Hg, indicating moderate volatility. The compound's octanol/water partition coefficient (log P) is 0.12, indicating low hydrophobicity. Oxazole is water-soluble and has a melting point of −82°C. It shows distinct spectral features in ultraviolet, infrared (IR), NMR, and mass spectrometry (MS) analyses. The heat of vaporization is 29.8 kJ/mol, and its refractive index at 17.5°C is 1.4285. These show the aromatic features, but less than thiazole. It is a weak base, having pKa −0.8 as compared with imidazole, having 7.[9] [10] There are numerous synthetic routes leading to oxazole and its analogs, and some important methods of them have been known as name reactions of oxazole synthesis.[11] These include Robinson–Gabriel synthesis,[12] Fischer oxazole reaction,[7] [13] Bredereck synthesis,[14] Van Leusen synthesis,[15] and cycloisomerization of certain propargyl amides[16] as illustrated in [Scheme 1].


Marketed Drugs with Oxazole Ring
Oxazole-containing approved drugs cover a wide range of therapeutic areas, including inflammation, infectious diseases, oncology, central nervous system disorders, and so on.[17] The marketed drugs with oxazole ring are listed in [Fig. 1]. In this context, valdecoxib is a selective cyclooxygenase (COX)-2 inhibitor belonging to the class of nonsteroidal anti-inflammatory drugs and used to treat osteoarthritis, rheumatoid arthritis, and primary dysmenorrhea.[18] Further, oxaprozin is a propionic acid-derived nonsteroidal anti-inflammatory drug (NSAID) used to relieve symptoms of arthritis such as inflammation, swelling, stiffness, and joint pain, and hence, employed in osteoarthritis and rheumatoid arthritis.[19] In the antidiabetic field, Aleglitazar is an oxazole containing PPAR-α/γ dual agonist, which is designed to treat type 2 diabetes mellitus (DM) by improving glucose and lipid control.[20] Furthermore, merimepodib (VX-497) is a potent inosine monophosphate dehydrogenase inhibitor with broad-spectrum antiviral properties.[21] In contrast, ditazole is a platelet aggregation inhibitor from the class of non-NSAID antithrombotics, i.e., used in the past to prevent thromboembolic events.[22] Moreover, mubritinib (TAK-165) is an HER2 tyrosine kinase inhibitor that is currently being investigated for HER2-positive breast cancer.[23] To exhibit antibacterial properties, oxacillin is a β-lactam antibiotic under the penicillin class, which is specifically used against penicillinase-producing staphylococci, whereas sulfisoxazole is a sulfonamide antibiotic used in urinary tract infections, otitis media, and chancroid.[24] [25] In case of inhibitory potential against viruses, pleconaril remains an effective option, which targets picornaviruses like enteroviruses and rhinoviruses by inhibiting viral uncoating.[26] Beside this, leflunomide is a disease-modifying antirheumatic drug, i.e., used in rheumatoid and psoriatic arthritis via inhibition of dihydroorotate dehydrogenase and pyrimidine biosynthesis.[27] Whereas, isocarboxazid is a nonselective and irreversible monoamine oxidase inhibitor used to treat atypical and treatment-resistant depression.[28] Also, broxaterol acts as a β2-adrenergic receptor agonist and is used as a bronchodilator in chronic obstructive pulmonary disease and asthma. Its oxazole-containing structure facilitates the selectivity toward bronchial smooth muscle β2-receptors with reduced cardiovascular side effects.[29]
Preclinical or Clinical Trial Candidates Bearing Oxazole Scaffold
Numerous oxazole-based molecules are currently under preclinical and clinical investigation due to their promising biological activities and favorable pharmacokinetic profiles ([Fig. 2]). These investigational agents target a wide range of diseases, with the oxazole ring playing a critical role in receptor binding, enzymatic inhibition, or protein degradation pathways.[30] In this sequence, PRT3789 is an SMARCA2-targeting degrader having an oxazole core incorporated within a proteolysis-targeting chimera (PROTAC). Currently, it is being evaluated in a Phase I dose-escalation trial (NCT05639751) for adult patients with advanced or metastatic solid tumors harboring SMARCA4 mutations. This trial assesses safety, tolerability, pharmacokinetics, pharmacodynamics, and suggests the recommended Phase II dose for both monotherapy and in combination with docetaxel. The study is actively recruiting patients under the sponsorship of Prelude Therapeutics.[31] A planned Phase II follow-on study combining PRT3789 with pembrolizumab (Keytruda) for SMARCA4-mutated cancers is listed in collaboration with Merck & Co.[32] Further, MK-4409 is a 1,3-oxazole-based FAAH inhibitor, i.e., developed by Merck for potential treatment of neuropathic and inflammatory pain. According to the Food and Drug Administration and pipeline records, MK-4409 reached Phase I trials for neuropathic pain around 2009 and is currently in early clinical development.[33] Next, luminespib (NVP-AUY922) is a 1,2-oxazole-based Hsp90 inhibitor originally discovered by The Institute of Cancer Research/Vernalis, while later licensed to Novartis. It was advanced into Phase I/II clinical trials from 2011 to 2014 across multiple cancer indications, such as relapsed/refractory multiple myeloma, nonsmall cell lung cancer (NSCLC), breast cancer, and other solid tumors. The largest of these trials evaluated the dose-escalation, combination therapy with bortezomib, as well as safety or tolerability endpoints (e.g., MTD, adverse events).[34] [35] Furthermore, rhizoxin is a macrocyclic lactone antibiotic containing an oxazole ring, which was originally isolated from Rhizopus species. Mechanistically, it disrupts microtubule polymerization, which makes it an antimitotic agent. It proceeded through both Phase I as well as multiple Phase II oncology trials, including studies in advanced solid tumors, squamous cell head and neck cancer, breast cancer, melanoma, and NSCLC. Phase I investigations used a 72-hour continuous intravenous infusion, whereas Phase II trials administered 1.5 to 2.0 mg/m2 every 3 weeks. Hereby, the main toxicities were leukopenia, stomatitis, neutropenia, and occasional phlebitis.[36] [37] Moreover, GNF6702 is a 2,4-dimethyl-1,3-oxazole-5-carboxamide derivative, which was discovered by the Genomics Institute of the Novartis Research Foundation. It acts as an allosteric proteasome inhibitor and selectively targets kinetoplastid parasites (e.g., Leishmania, Trypanosoma). In preclinical mouse models for visceral leishmaniasis, Chagas disease, and sleeping sickness, GNF6702 demonstrated complete parasite clearance, favorable safety, and high selectivity, as compared with mammalian cells. However, it remains in preclinical development with no current human clinical trials registered.[38] [39]


Advancements in the Biological Activities of Oxazole Derivatives
Oxazole-containing derivatives have a large number of pharmacological implications, which have drawn their attention toward both medicinal and industrial applications. In terms of biological activity, oxazole derivatives display a range of therapeutic effects.[40] They are known to inhibit key enzymes such as COX and tyrosinase, contributing to their anti-inflammatory and anticancer properties.[41] They also exhibit antibacterial,[42] antifungal,[43] and antioxidant activities,[44] as well as antitubercular[45] [46] effects. Other notable biological activities include anticonvulsant[47] and antihyperglycemic[48] properties.
Beyond their pharmaceutical uses, oxazole derivatives serve as functional materials with different applications. They are utilized in the production of homopolymers, peptides, and condensation reagents, and as active ingredients in herbicides, pesticides, and fungicides.[49] [50] Additionally, oxazole compounds are employed as profitable luminophores in plastic materials, laser dyes, and polymer systems.[51] [52] They are also used as chiral ligands in asymmetric synthesis[53] and employed as intermediates in the synthesis of agrochemicals.[54] Furthermore, oxazole derivatives play a role in polymerization reactions, further expanding their industrial and commercial utility.[55] This broad range of applications highlights the versatility and importance of oxazole derivatives in both biological and industrial contexts.
Advancements in the Antibacterial Activity of Oxazole Analogues
A growing global concern is the increasing resistance of pathogenic bacteria to existing antibiotics and antifungal agents, which has become a significant challenge in health care.[56] To address this issue, it is crucial to discover and develop novel classes of drugs that can effectively combat the rising threat of resistant microbes.[56] [57] This has emerged as one of the most important areas of antibacterial research. Among the promising candidates, heterocyclic compounds such as oxazole derivatives have gained attention due to their broad spectrum of biological effects, especially antimicrobial properties.[58]
In 2022, Singagari and Sundararajan designed and synthesized a series of 4,5-dihydro-1H-pyrazol-3-yl substituted 4-benzylidene-2-methyloxazol-5(4H)-one derivatives (1a-o, [Fig. 3]) and evaluated their antimicrobial activities.[59] Among the synthesized series, 1h exhibited the highest antibacterial activity, having a minimum inhibitory concentration (MIC) value of 1.56 µg/mL against Bacillus subtilis, Staphylococcus albus, and Proteus vulgaris, i.e., equivalent to the standard drug ciprofloxacin (MIC = 1.56 µg/mL). Results of antitubercular assays indicated that compounds 1j and 1l exhibited the strongest inhibitory profile with a MIC value of 3.13 µg/mL against Mycobacterium tuberculosis H37Rv, as compared with isoniazid (MIC = 0.05 µg/mL). Further, molecular docking studies demonstrated that 1l showed the best binding score of −9.94 kcal/mol against MtKasA (PDB: 2WGE), whereas 1j was found to be most potent with a binding score of −8.91 kcal/mol against MtpKnB (PDB: 2FUM) by surpassing even the reference agent mitoxantrone. These results suggested that oxazole derivatives bearing unsubstitution, meta-OH and meta-CH3 groups at the R position on the phenyl linked to the pyrazoline ring exhibited superior antibacterial potency.


Keivanloo et al reported a novel series of 1,3-oxazole-quinoxaline amine hybrids (2a-l, [Fig. 3]) and evaluated their antibacterial activity.[60] Among them, compounds 2b, 2c, 2g, and 2j displayed the most potent inhibitory effect against Micrococcus luteus and Pseudomonas aeruginosa with MIC values of 62.5 and 31.25 µg/mL, respectively, which were almost equipotent to the standard drug tetracycline. Next, the molecular docking analysis highlighted that compound 2j is the most potent with the best docking score of −8.49 kcal/mol toward Yersinia pestis DHPS (PDB ID: 5JQ9).[60] Structure–activity relationship (SAR) data suggested that phenyl and 4-Cl as R group on oxazole nucleus, while aliphatic or aryl substituted aliphatic 2° amines as the R1 group were the favorable substituents for encouraging antibacterial activity.
Seelam et al developed a series of thiadiazole-tethered oxazole hybrids (3a–j, [Supplementary Table S1], available in the online version). The introduction of electron-withdrawing groups (-4-Cl, 4-Br, 4-NO2, 2-Cl, 3-NO2) at phenyl on the isoxazole ring is essential for suitable antibacterial activity. Among these compounds, 3a, 3b, 3c, 3e, and 3j demonstrated excellent antibacterial potential compared with streptomycin against B. subtilis and Bacillus thuringiensis, possessing MIC values of approximately 3.125 µg/mL, as compared with streptomycin (MIC values 6.25 and 12.5 µg/mL, respectively). Due to the variability inherent in the disc diffusion assay, the activity of these derivatives at lower concentrations was further investigated using the broth dilution method. The bacterial strains B. thuringiensis, P. aeruginosa, B. subtilis, and Escherichia coli were tested, and the outcomes of their antimicrobial potential.[61]
Mohanty et al synthesized a series of oxazole 2,4-diamine compounds (4, [Fig. 3]) and evaluated their antibacterial potential against both Gram-positive and Gram-negative bacterial strains. Electron-withdrawing groups (EWGs), like as fluoro- or choro-substitution on the phenyl ring, are essential to deliver potent antibacterial activity. All the synthesized derivatives demonstrated significant antibacterial activity. Ciprofloxacin (50 µg/mL) was used as the standard drug. Among the newly synthesized compounds, 4b and 4c showed the highest antibacterial activity.[62]
Chilumula et al have prepared a series of novel benzoxazole-5-carboxylate derivatives (5a-i, [Fig. 3]) and evaluated their antibacterial activity using the cup plate assay. The compounds demonstrated excellent antibacterial potency toward Staphylococcus aureus and B. subtilis (Gram-positive bacteria), as well as E. coli and Salmonella typhi (Gram-negative strains). Electron-donating groups (EDGs) favor antibacterial potency. Disubstitution of the Ar group at the meta and para positions (2-OH-4-OCH3-C6H3) synergizes the antibacterial efficacy. Among all the molecules tested, compound 5g exhibited superior antimicrobial results against both tested strains, with zone of inhibition values ranging between 20 and 24 mm, which was more potent than standard antibiotic ampicillin (zone of inhibition = 17–22 mm).[63]
In the same year, Dabholkar et al synthesized novel compounds containing the oxazole ring (6a–e, [Fig. 3]) and tested their antifungal and antibacterial activities by utilizing the disc diffusion assay technique. Ampicillin trihydrate was taken as the reference antibacterial agent. Results of in vitro antibacterial studies indicated that compound 6c was the most effective molecule against S. aureus (zone of inhibition = 11 mm), 6b, and 6e showed the topmost antibacterial efficacy toward Corynebacterium diphtheriae, with zone of inhibition values of 11 mm. Also, 6b experienced the most promising antibacterial potency for P. aeruginosa (zone of inhibition = 12 mm), whereas compounds 6a, 6c displayed remarkable antibacterial activity against E. coli with zone of inhibition values of 12 mm, as compared to standard antibiotic ampicillin trihydrate (zone of inhibition = 21–28 mm).[64] SAR findings suggested that attachment of EDGs like methyl and methoxy groups at R and R1 positions, respectively, whereas the unsubstituted or OH-substituted-R4 position showed the most promising antibacterial results.
Reddy et al synthesized new oxazole derivatives (7a-l, [Supplementary Table S3], available in the online version) and tested their antimicrobial activities against different bacterial strains.[65] These compounds exhibited significant inhibitory effects at concentrations ranging from 258 to 564 µg/mL in 20% water in dimethyl sulfoxide. The favorable substituents were para-halo-substituted (4-Cl or 4-Br) phenyl at the R position. Among the synthesized compounds, 7b, 7f, 7e, and 7g showed pronounced antibacterial activities ([Fig. 3]), with the following order of increasing potency: 7g > 7f > 7e > 7b. Specifically, compounds 7c, 7d, 7h, and 7k presented significant antibacterial potency toward E. coli, while 7i and 7j were more effective against B. subtilis, and 7m and 7l showed better activity against Klebsiella pneumoniae. Ampicillin was selected as the reference drug for comparison of antimicrobial efficacy.
Kamble et al synthesized piperidinyl-substituted oxazole-containing derivatives (8a–b, [Fig. 3]) with unsubstituted phenyl or p-nitrophenyl at Ar. They evaluated the antibacterial potency of the compounds toward S. aureus and E. coli bacterial strains.[66] The results are presented in [Supplementary Table S4] (available in the online version).
Advancements in Antifungal Activity of Oxazole Analogues
The development of antifungal agents has been taken after antibacterial agents. Bacteria are prokaryotic, and hence, they have a large number and metabolic sites, which are distinct from the possession of a hominid host.[67] Fungus is eukaryotic in nature and the utmost poison to the host cell. By virtue of this fungus commonly spreads slowly and generally in a multicellular system. There are numerous new antifungal agents that particularly discharge fungal pathogens from a hominid host, along with lesser toxicity, and are hence called antimycotic drugs.[68] [69] [70]
Bąchor and colleagues reported a new series of isoxazole-based α-acyloxyamide derivatives (9, [Fig. 4]) and evaluated their antifungal activity against Candida albicans. Sulfur-rich heterocycles (thiazole or thiophene) at Ar, whereas a benzyl group at Ar' enhanced the antifungal activity. Among all, compounds 9a and 9b demonstrated the most prominent anti-fungal activity against C. albicans biofilm (MIC ≥ 10 μg/mL; % minimum biofilm eradication concentration (MBEC) reduction = 54.9% and 24.6%, respectively) without adverse interference to beneficial Lactobacillus spp., which was a major advantage over conventional antifungals like clotrimazole or octenidine dihydrochloride. Further, compounds 9a and 9b also exhibited lower cytotoxicity against Henrietta Lacks (HeLa) cells.[71]


In the same year, Tlapale-Lara et al assessed pyrazole and oxazoline (10a–j and 11a–j, [Fig. 4]) derivatives as potent antifungal agents. The in silico evaluation indicated that all molecules adhere to Lipinski's rules with low toxicity and suggesting good drug-like behavior. Further, Homology modeling of CYP51 was conducted based on available Candida species to generate validated 3D models for in silico interactions analysis. The models confirmed that these compounds fit well within the enzyme's active site. In addition, Molecular docking revealed that these compounds exhibit better binding energies, and compound 11d exhibited the highest binding energy (–14.23 Kcal/mol) against C. albicans, as compared with fluconazole (−7.29 Kcal/mol). In vitro antifungal testing across several Candida species demonstrated that compounds 10a–j and 11a–j showed significant MIC90 values ranging between 0.11–14.4 and 0.05–42.3 μg/mL, respectively, as compared with fluconazole (MIC90 = 1.4– > 57.6 μg/mL).[72] Pharmacophore hybridization in pyrazolyl oxazoles (11a–j) exerted synergistic advantages in antifungal action, and a Bulkier halogen at R (e.g., Br) showed promising activity.
In 2009, Ryu et al prepared novel benzoxazole derivatives (12a–b, [Fig. 4]) and investigated their antimycotic potential against various fungal strains. The control drug, 5-fluorocytosine, was used for comparison of antifungal activity. Results of this study presented the antifungal effect of synthesized molecules either superior to or comparable with that of the reference drug, as presented in [Supplementary Table S5] (available in the online version).[73] para-Bromophenyl substitution on the core scaffold was found to be more beneficial for the antifungal profile in contrast to the unsubstituted counterpart.
Rawat and Shukla synthesized a new series of oxazole derivatives (13, [Fig. 4]) with antifungal potency. The antimycotic potential was evaluated using the cup plate assay method, where ketoconazole (100 µg/mL) was selected as the reference antifungal agent. The result suggested that the -p-NO2 group at R displayed the most favorable antifungal profile, as compared with other EDGs (OH, OCH3) or EWGs (halogens) substitutions. The oxazole imine compound 13e exhibited the maximum inhibition against both C. albicans and Aspergillus niger, having zone of inhibition values of 17 and 16 mm, respectively, as compared with the standard antifungal drug ketoconazole (zone of inhibition = 20 mm; C. albicans and 18 mm; A. niger). Also, its analogues 13a, 13b, 13c, 13d, and 13f showed moderate activity against both fungal strains, with zone of inhibition values ranging from 9 to 14 mm against the tested fungal species.[74]
Kakkar et al synthesized new compounds containing the oxazole ring (14, [Fig. 4]) and evaluated their antimycotic potential. The introduction of the Cl group contributed to C. albicans inhibition, and the phenoxy group to A. niger inhibition. Compound 14a exhibited the most potent activity against A. niger, whereas derivatives 14b and 14c showed moderately antifungal activity against A. niger. In contrast, compounds 14b and 14c were found to be highly active toward C. albicans, as compared with Fluconazole ([Supplementary Table S6], available in the online version).[22]
Advancements in Antitubercular Activity of Oxazole Analogues
Tuberculosis (TB) is a virulent disorder that primarily affects the lungs. In contrast to another disease, which is induced by a particular virulent agent, TB is the second massive common killer all over the world. The World Health Organization estimates that near about 9 million persons in a year diagnosed with TB, along with 3 million of them forgotten by the health system.[75] A series of bicyclic nitroimidazole oxazole has been mainly searched as radiosensitizers in cancer chemotherapy, but it is also found useful against the culture replication of airborne pathogen M. tuberculosis.[76] [77]
Thakare et al focused on the design, synthesis, and evaluation of novel benzoxazole derivatives (15a–i, [Supplementary Fig. S5], available in the online version) with potential antibacterial and antitubercular activities.[78] In this work, these derivatives were characterized by IR, NMR, and MS to confirm their structures and purity. Furthermore, the antibacterial activity was assessed via serial dilution methods against Gram-positive bacteria (S. aureus) and Gram-negative bacteria (E. coli). Here, methoxy and halogen (Br > I > I) groups at R and X positions, respectively, were found to be necessary for anti-TB activity. Whereas propyl ester exerted the most promising inhibitory effect against M. tuberculosis. Among the compounds, 15a, 15c, 15f, and 15i showed significant inhibition at concentrations of 50 and 100 μg/mL. Similarly, the anti-tubercular efficacy was evaluated using the Alamar blue assay against M. tuberculosis H-37RV strain and compounds 15c and 15i, demonstrating noteworthy anti-TB activity at similar concentrations.[78]
Shinde et al reported the synthesis of novel oxazole-based hybrid molecules (16 and 17, [Fig. 5]) to develop potent anti-tubercular agents. Here, these molecules were biologically evaluated against M. tuberculosis H37Rv, including drug-resistant strains like MDR and XDR, using a colorimetric microplate assay. SAR data revealed that 4-nitrophenyl at the second position of the oxazole nucleus enhanced the biological activity. Among all these compounds, only 16a exhibited significant anti-TB activity with a MIC value of 6.25 μg/mL, in comparison to Isoniazid (MIC = 3.125 μg/mL). Further, molecular docking was performed for compound 16a against bacterial enzyme deoxyribonucleic acid gyrase (PDB ID: 4B6C). The docking results revealed van der Waals interactions with VAL 77 and various pi-cation or pi-alkyl interactions with amino acids ARG 82, ILE 84, and PRO 85 that likely contribute to its efficacy.[79]


Moraski and their group synthesized various derivatives (18a–i, [Supplementary Table S7], available in the online version) using the Suzuki coupling reaction and evaluated their antitubercular activity. The MICs were determined against M. tuberculosis H37Rv. Rifampicin and clinical candidate PA-824 were chosen as reference anti-TB agents. The results revealed that, except for compound 18h (>128 μmol/L), the derivatives were generally less effective against verda reno (means green kidney) cells. The 4-benzyloxy phenyl ring at R showed the most promising antitubercular activity as compared with single aromatic ring substitutions. Among all the tested molecules, compound 18h was found to be most effective against M. tuberculosis H37Rv with a MIC value of 0.60 μmol/L, as compared with rifampicin (MIC = 0.06 μmol/L). The results are presented in [Supplementary Table S7] (available in the online version).[80]
Prior to Moraski's work, Zwawiak et al synthesized and investigated novel 2,3-dihydroimidazo [2,1-b]oxazoles (19, [Fig. 5]) as potential antitubercular agents. The in vitro antitubercular assay of synthesized molecules was performed by taking isoniazid (INH) as the standard antitubercular drug. Tiny alkyl groups are advantageous for the anti-TB profile (ethyl > methyl). Compounds 19a and 19b were tested against M. tuberculosis, Mycobacterium avium, and Mycobactrium bacillus Calmette–Guérin, whereas two wild species were collected from TB patients. Among these strains, 1676 showed resistance to INH, and the 456 strain exhibited resistance to both reference agents. Further, the antitubercular potential of these derivatives was assessed, and MIC values of the synthesized compounds are presented in [Supplementary Table S8] (available in the online version).[81]
Advancements in Anticancer Activity of Oxazole Analogues
Malignancy is one of the serious diseases, where anomalous cells arise and might develop in existing cells at each one phase of mortal life.[82] Here, nearly a hundred types of tumors have been involved, such as blood tumor, skin tumor, brain tumor, breast tumor, prostate tumor, lung tumor, colon tumor, etc.[83] There are different therapies for tumors, inclusive of chemotherapy, radiation, and surgery. After the existence of these therapies, tumors have into the biggest challenge with the health complications globally. However, a large number of anticancer drugs were tested a few years ago due to the existence of distinct cell lines and the use of various techniques.[84] [85] [86]
Khowdiary et al discussed the synthesis and evaluation of a new series of oxazole derivatives tethered with an oxadiazole moiety (20, [Fig. 6]) as potent antileukemic agents. In their work, the synthesized compounds were tested for their in vitro inhibitory effect against HL-60 and PLB-985 enzymes, in comparison to Etoposide. It was noticed that the 4-trifluoromethyl group was the most favorable substitution to elicit anticancer effect (4-CF3 > 2,5-di-F > 3,5-di-OH). Compound 20b exhibited the most potent dual inhibitory profile with IC50 values of 8.50 and 12.56 µg/mL, respectively, as compared with etoposide (IC50 = 10.50 and 15.20 µg/mL, respectively). Further, molecular docking revealed strong binding affinities within the active site, and 20a, 20b, and 20c have the highest binding energy. Likewise, ADMET analysis showed that potent molecules do not violate the Lipinski rule of 5 and have a good therapeutic profile.[87]


Quite recently, Komirishetti and Mittapelli reported the synthesis and biological evaluation of a novel series of imidazole clubbed pyrimidinyl oxazole hybrids (21a–j, [Supplementary Fig. S6], available in the online version) as potent anticancer agents. Further, all the compounds were investigated for anticancer assessment against four human cancer cell lines, viz. MCF-7, Colo-205, A549, and A2780 cells were tested with the help of the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, whereas etoposide was used as a standard treatment. Trimethoxy substitution (3,4,5-trimethoxy) at R showed an advantageous anticancer influence than mono-/di-substituted methoxy groups. Among all the compounds tested, compound 21a emerged as the most potent cytotoxic agent, having an IC50 value of 0.06 ± 0.0072 µmol/L against MCF-7, which was more active than Etoposide (IC50 = 2.19 ± 1.87 µmol/L).[88]
Premakumari et al synthesized a novel amido-sulfomido methane-associated bis-oxazole compound (22, [Fig. 6]).[89] SAR outcomes concluded that the EWGs substituted (4-Cl-phenyl) was more beneficial than the unsubstituted or 4-CH3 substituted counterparts. The antitumor potential of 22c against various cancer cell lines is described in [Supplementary Table S9] (available in the online version).
Mathew et al synthesized 23a–c ([Fig. 6]), which were found to be less potent than sulindac sulfide amide. Among these, 23b, which was substituted with dimethyl derivatives of sulindac, showed modest activity against all the described cell lines in vitro, as presented in [Supplementary Table S10] (available in the online version). Further, 24b, shows notable activity as a carboxylate and demonstrates moderate potency compared with sulindac, as detailed in [Supplementary Table S10] (available in the online version).[90] The results suggested that favorable substituents for compounds 23, 24, 25 were a 3,4,5-trimethoxyphenyl ring attached to the indole ring via an alkene linkage. The results suggested that favorable substituents for compounds 23, 24, and 25 were a 3,4,5-trimethoxyphenyl ring attached to the indole ring via alkene linkage.
Kachaeva et al synthesized several derivatives of oxazole (26a–f, [Fig. 6]) and investigated their antitumor potential. Derivatives 26a, 26b, 26c, and 26e, which contain amino, sulfanyl, or sulfonyl groups at position 5 of the 1,3-oxazole ring, exhibited modest antitumor activity. Results revealed that all tested oxazole analogues possessed significant anticancer potential against selected cancerous cells, having GI50 values in the range of 0.2 to 0.6 μmol/L. Compound 26e exhibited the topmost anticancer activity against the tested NCI-60 cancer cell lines with GI50 values ranging between 0.15 and 6.4 μmol/L.[91]
In the work by Romagnoli et al, a library of antitubulin agents (27a–c, [Fig. 6]) was synthesized and investigated for their antitumor potential against various cancer cell lines, while Combretastatin A-4 (CA-4) was taken as the reference drug. Further, compounds 27b and 27c showed the best anticancer profile among the derivatives, outperforming CA-4 in several cases. Compound 27b showed comparable potency to CA-4 toward SEM as well as Jurkat cells, while it was found to be many-fold more potent against other tumor cell lines ([Supplementary Table S11], available in the online version).[92]
Ranjith and their team synthesized a series of benzoxazole derivatives (28, [Fig. 6]). All the compounds were tested for antitumor potential using the MTT assay against the MCF-7 cancer cells. The results showed that a single EWG (bromo/nitro) substituted phenyl ring showed a potent anticancer effect, with 28a and 28b exhibiting the highest potency, with % growth inhibition values of 45.568 and 42.236, respectively.[93]
Advancements in Antioxidant Activity of Oxazole Analogues
Free radicals like superoxide anion radicals (O2 •−), hydroxyl radicals (OH•), peroxy radicals (RO2 •), nitric oxide (NO), and (O) atoms are important for many physicochemical systems. The drugs that are most correlated to antioxidant activity can metabolize the free radicals and their intermediary products diffusively into nontoxic compounds.[94] Increment in cancers, cardiovascular system diseases, and aging occurred due to oxidative cellular damage. Antioxidants are substances that mitigate oxidative stress in cells.[95]
Rao et al designed and synthesized a series of oxazole clubbed 1,2,4-triazolothiadiazole hybrids (29, [Fig. 7]) as potent antimicrobial agents. Herein, antimicrobial efficacy was tested against six Gram-positive and Gram-negative strains by using ofloxacin as the standard drug. The substituents at the meta position of phenyl ring linked to the oxazole ring were preferred to exhibit biological activity (isopropyl > 4-methoxyphenyl > allyl).


Among these, compounds 29a and 29b exhibited the most potent antibacterial activity with zone of inhibition values ranging from 18–36 and 16–32 mm against all tested bacterial strains. Additionally, the antioxidant activity revealed that compounds 29c and 29b emerged as the most potent in 2, 2-diphenyl-1-picrylhydrazyl (DPPH) and H2O2 radical scavenging assay with half-maximal inhibitory concentration (IC50) values of 24.39 and 18.76 µg/mL, respectively, as compared with ascorbic acid (IC50 = 20.20 and 21.76 µg/mL). Molecular docking study against E. coli with PDB ID:4CKL revealed that 29b elicited the highest binding energy −5.67 kcal/mol.[96]
In this context, Sagud et al synthesized naphthoxazole derivatives with a methoxy group at the naphthoxazole ring system (30a–b, [Fig. 7]) and evaluated their antioxidant potential with the help of DPPH radical scavenging and Ferric reducing ability of plasma assay methods.[97] The half-maximal DPPH radical scavenging concentration (IC50) of derivative 30a was determined to be 0.4 µmol/L. The results were presented in [Supplementary Table S12] (available in the online version).
Canan et al synthesized a series of derivatives of oxazole-5(4H)-one (31a–j, [Fig. 7]) and tested their antioxidant activity using male albino Wistar rats.[98] NADPH-dependent lipid peroxidation was determined spectrophotometrically using the formation of thiobarbituric acid-reactive substances technique. The result showed that mono-substitution of tiny EDG, like CH3 at R, whereas di-substitution of EWG (fluoro) at the X position, is preferred. Compound 31c exhibited the highest reactivity in terms of microsomal ethoxyresorufin O-deethylase (EROD) activity of 4.47 ± 0.04 pmol/mg/min, as compared with standard caffeine (EROD = 6.41 ± 0.99 pmol/mg/min). Substitution with a biphenyl group decreased the EROD activity but had no effect on lipid peroxidation levels, demonstrating moderate antioxidant activity.
Advancements in Antiviral Activity of Oxazole Analogues
In 2024, Severin et al synthesized a novel series of 1,3-oxazole-4-carbonitriles and 4-sulfonylamide-5-phenyl-1,3-thiazoles (32, [Fig. 8]) as a potent marker of human papillomavirus.[99] A QSAR model employing artificial neural networks was developed to predict their anti-human papillomavirus (HPV) activity. Further, these compounds were evaluated for in vitro activity against HPV types 11, 16, and 18 using C33-A cell lines to determine antiviral efficacy measured through EC90, half-maximal cytotoxic concentration (CC50), and selectivity indices SI90. It was revealed that 32a exhibited the highest antiviral activity against HPV11 with EC50, CC90, and SI90 values of 20.44, 88.10, and 4 µmol/L, respectively, whereas standard drug 9-[2-phosphono-methoxy)ethyl]guanine had EC50, CC90, and SI90 values of 82.94, ≥150, and ≥2 µmol/L, respectively. The result showed that the phenyl ring at R, whereas the sulfonyl phenyl linked to sulfonyl piperazine at R1 presented promising antiviral action.


Kachaeva et al synthesized a range of 1,3-oxazole analogs (33a–b, [Fig. 8]) and evaluated the prevalence of HPV infection. 33a and 33b showed EC50 values of ≥50 and 2.43 µmol/L against HPV-18 and HEK-293 cells, respectively, in comparison to the cidofovir showing EC50 values of 148 µmol/L against both cells, suggesting that sulfonyl piperidine is more efficacious than the N-unsubstituted sulfonamide group counterpart. In this work, regression analysis was used to evaluate the antiviral activity of the compounds. The results of EC50 and CC50 values of these analyses were presented in [Supplementary Table S13] (available in the online version).[100]
Makarov et al developed a library of isooxazole analogs (34a–c, [Fig. 8]) and evaluated their antiviral activity. HeLa cells were used for plaque reduction assays to isolate CVB3 97-927 and for Carrageenan paw edema test (cytopathic effect) inhibition assays with CVB3 Nancy, HRV-14, and HRV-2. The activities of the R substituents are as follows: - 4-F > 3-F > 3-NO2. The range of CC50 values for the derivatives was found to be 4.6 to >50 µg/mL, as shown in [Supplementary Table S14] (available in the online version).[101]
Advancements in Anticonvulsant Activity of Oxazole Analogs
Epilepsy is a life-threatening and serious disease for humans. Epilepsy refers to a neurodegenerative disorder in which disruption of nerve cell activity occurs in the brain. More than 1 million cases per year were considered in India. To eradicate epilepsy, the discovery of potent antiepileptic drugs is too important.[102] [103]
Srilakshmi et al designed a series of thiazole-oxazolone derivatives (35a–m, [Fig. 9]) and evaluated their anticonvulsant activity. In their research, in silico molecular docking revealed significant binding affinities of these derivatives toward targets like carbonic anhydrase (PDB ID: 1HOW) and GABAAT (PDB ID: 3F86). Molecular docking study against carbonic anhydrase enzyme revealed that 35b showed the highest docking score of −9.97 kcal/mol in comparison to Phenytoin (binding energy = − 6.65 kcal/mol). In addition, 35f showed the topmost binding energy of −7.37 kcal/mol against the GABAAT receptor in comparison to Phenytoin (binding energy = − 3.78 kcal/mol). Furthermore, the biological evaluation through the maximal electroshock test (MES) and scPTZ seizure models demonstrated that compounds 35a, 35c, and 35e exhibited notable anticonvulsant activity at various doses, with 35c emerging as the most potent.[104] The results showed that halo-substitution on the meta position (m-Cl, m-Br) of the phenyl ring tethered via Schiff base linkage increased the activity of the compounds.


Song et al developed a series of triazole-containing benzo[d]oxazole derivatives (36a–d, [Fig. 9]) and tested them for anticonvulsant activity using the scPTZ (subcutaneous pentylenetetrazol) and MES models.[105] The newly developed compounds were administered to mice at different time intervals (0.5 and 4 hours). Compounds 36a–d showed the highest activity at a drug concentration of 30 mg/kg after 0.5 hours in the MES screening. Compound 36d demonstrated significant activity at 30 mg/kg after 4 hours, whereas compounds 36a–c required a higher dose of 100 mg/kg to show comparable activity at the same time interval. 36d showed an ED50 value of 12.7 and 29.5 mg/kg via the intraperitoneal route in MES and scPTZ models, respectively, compared with carbamazepine, demonstrating ED50 values of 9.8 and >100 mg/kg, respectively. The results showed that 4-F-benzyl substitution at the benzoxazole core elicited more promising anti-convulsant action than 2-F- or 3-F-benzyl counterparts. The data are presented in [Supplementary Table S15] (available in the online version).[106]
Singh et al prepared novel derivatives of triazole thione semicarbazones (37a–e, [Fig. 9]) and investigated their antiepileptic potential as GABA agonists. In this study, Albino mice of either sex, weighing 20 to 25 g, were used for testing the anticonvulsant activity with diazepam as the reference drug for comparison. The anticonvulsant potential of the synthesized molecules is depicted in [Supplementary Table S16] (available in the online version).[107] 37d, 37e, and 37c showed maximum activity, whereas compounds 37a and 37b exhibited moderate activity. So, these are the lead molecules for another therapeutic potential.
Advancements in Anti-Inflammatory and Analgesic Activity of Oxazole Analogs
Nonsteroidal anti-inflammatory agents/analgesics are a class of analgesics (pain-relieving) that reduce pain, fever, and inflammation without loss of consciousness. Besides therapeutic uses, side effects like gastric bleeding, ulceration also occurred by administering these drugs. So, it becomes the biggest challenge to overcome these side effects and provide much better and safer drugs in the market. With this, the discovery of new drugs became more important.[105] [108] [109]
Garg et al discovered a series of oxazole derivatives (38, [Fig. 10]).[110] After synthesis, spectroscopic methods such as FT-IR and 1H NMR were used to validate the chemical structures of the synthesized analogs. Among the compound, derivative 38b, with a p-amino benzaldehyde linked to the amino group of phenyl oxazole amine was the most effective anti-inflammatory candidate with 28.67% inflammatory inhibition (Paw edema volume = 1.28 ± 0.03) in the third hour in the carrageenan-induced rat hind paw edema method, as compared with indomethacin (% inhibition = 45.86%; Paw edema volume = 1.44 ± 0.02).


Meenakshi et al investigated the dual functionality of 2,4,5-trisubstituted oxazole derivatives (39, [Fig. 10]) as aquaporin-4 (AQP4) inhibitors as well as anti-inflammatory agents in human lung cells. In this research work, molecular docking was conducted against the AQP4 receptor (PDB ID: 3GD8), which revealed that compound 39a, with methyl, a tiny EDG at R while an unsubstituted phenyl ring at the Ar position, respectively, exhibited the strongest binding affinity of −7.3 kcal/mol via interacting primarily with GLY144, GLY146, and VAL147 amino acid residues. Moreover, the ADME study showed that these compounds followed Lipinski's rule of five. In vitro studies on NCI-H460 lung cells demonstrated that compound 39a effectively inhibited LPS-induced upregulation of the AQP4 gene and suppressed the transcription of proinflammatory cytokines. In addition, hemolysis assays confirmed its excellent biocompatibility with less than 1.6% hemolytic potential even at drug concentrations up to 600 µmol/L.[111]
Shakya et al discovered oxazole ring-containing derivatives (40a–b, [Fig. 10]) and investigated their anti-inflammatory potential.[112] Albino rats were used for preclinical trials. The newly developed compounds were given at a dose of 20 mg/kg body mass via the oral route, and paw volume was measured plethysmographically. An equal amount of normal saline was used as the control group, and ibuprofen (20 mg/kg body weight) was used as the reference drug. Compounds 40a and 40b exhibited significant anti-inflammatory potential, surpassing the reference drug, as shown in [Supplementary Table S17] (available in the online version).[112]
Sarkate and Shinde developed oxazole-based compounds (41, [Fig. 10]) and assessed their anti-inflammatory, analgesic, and nitric oxide-releasing properties.[113] Among all synthesized compounds, compounds 41a and 41b were found to be the most potent anti-inflammatory analogues with 69.26 and 68.78% inflammatory inhibition, respectively, at 3 hours. Results of 41a were equipotent to celecoxib (standard COX-2 inhibitor) at 3 hours. The biological evaluation results are presented in [Supplementary Table S18] (available in the online version).
Advancements in Antidiabetic Activity of Oxazole Analogs
DM, a metabolic disease that induces an elevated blood glucose level due to impairment of the insulin hormone. Untreated high blood sugar leads to hyperglycemia and long-term complications, including angiopathy, neuropathy, and retinopathy. So, it is very necessary to treat this disease to prevent its complications.[114] [115] [116]
Husain et al targeted the PPARγ receptor and designed a series of oxazole derivatives as potential antidiabetic agents. In this work, a series of distinctive oxazole compounds (42-43, [Fig. 11]) was structurally modeled using ChemDraw 3D and optimized for physicochemical properties. The favorable substituents were the Schiff base of benzamide and cyclopentadiene clubbed with o-hydroxy benzaldehyde at the second position of the oxazole core. Among the compounds, 43a and 43b have the topmost drug scores (0.91 and 0.86) than Rosiglitazone (0.80). Next, all compounds were docked using PyRx software into the PPARγ receptor (PDB ID: 1PRG), which revealed that 42a and 42b displayed better binding energies, i.e., −11.1 and −10.1 kcal/mol, respectively, than rosiglitazone (binding energy = − 9.1 kcal/mol). The binding interactions included hydrogen bonds, pi-cation, and pi–pi interactions, which contributed to their stability and specificity.[117]


Mariappan et al established a series of oxazolone derivatives and investigated their oral hypoglycemic potential (44a–c, [Fig. 11]) using the streptozotocin-induced diabetes model in rats. Rosiglitazone was used as the reference drug, demonstrating 69% blood glucose-decreasing potential at a dose of 100 mg/kg orally. It was revealed that compound 44b showed the maximum potential amongst all compounds compared with the control drug, as shown in [Supplementary Table S19] (available in the online version).[48]
Conclusion and Future Outlook
The data presented in this review underscore the broad spectrum of therapeutic activities exhibited by oxazole and its derivatives, including antimicrobial, anticancer, antitubercular, antidiabetic, anti-inflammatory, analgesic, anticonvulsant, and antiviral properties. A thorough literature survey reveals that compounds containing an oxazole nucleus have gained significant attention from researchers and biochemists due to their promising biological activities. This has driven efforts to explore and develop novel strategies for synthesizing oxazole-based drugs. This review emphasizes that oxazole derivatives hold considerable potential to generate potent bioactive compounds, making them a valuable focus for the discovery of new therapeutic agents. Future directions should emphasize artificial intelligence tools to identify target sites for therapeutic heterocycles, followed by evaluation of available binding residues that may help to design new potent molecules. Further, hybridization of therapeutically beneficial heterocyclic scaffolds can become a synergistic therapeutic strategy against a wide range of disorders. Also, advancements in multitarget inhibitors, along with associated novel drug delivery systems, can afford medicinal advantages. In addition, adaptive SAR modeling on the basis of resistance data and their pharmacokinetic assessment is essential for clinical translation. In contrast, interdisciplinary efforts will be decisive to accelerate the development of next-generation heterocyclic therapeutics.
Conflict of Interest
None declared.
Supporting Information
The chemical structures of 1a–o, 2a–i, 5a–i, 6a–e, 15a–i, 21a–j ([Supplementary Figs. S1]–[S6]), available in the online version; 3a–j ([Supplementary Table S1], available in the online version), 7a–l ([Supplementary Table S3], available in the online version), 18a–i ([Supplementary Table S7], available in the online version), and the in vitro activity data of compounds mentioned in the text ([Supplementary Tables S1]–[S19], available in the online version) can be found in the “Supporting Information” section of this article's webpage.
# These authors contributed equally to this work.
-
References
- 1 Benton ML, Abraham A, LaBella AL, Abbot P, Rokas A, Capra JA. The influence of evolutionary history on human health and disease. Nat Rev Genet 2021; 22 (05) 269-283
- 2 Filatova EV, Shadrina MI, Slominsky PA. Major depression: one brain, one disease, one set of intertwined processes. Cells 2021; 10 (06) 1283
- 3 Dhameja M, Gupta P. Synthetic heterocyclic candidates as promising α-glucosidase inhibitors: an overview. Eur J Med Chem 2019; 176: 343-377
- 4 Guan Q, Xing S, Wang L. et al. Triazoles in medicinal chemistry: physicochemical properties, bioisosterism, and application. J Med Chem 2024; 67 (10) 7788-7824
- 5 Zhang HZ, Zhao ZL, Zhou CH. Recent advance in oxazole-based medicinal chemistry. Eur J Med Chem 2018; 144: 444-492
- 6 Neha K, Ali F, Haider K, Khasimbi S, Wakode S. Synthetic approaches for oxazole derivatives: a review. Synth Commun 2021; 51: 3501-3519
- 7 Zheng X, Liu W, Zhang D. Recent advances in the synthesis of oxazole-based molecules via van Leusen oxazole synthesis. Molecules 2020; 25 (07) 1594
- 8 Sisco E, Barnes KL. Design, synthesis, and biological evaluation of novel 1,3-oxazole sulfonamides as tubulin polymerization inhibitors. ACS Med Chem Lett 2021; 12 (06) 1030-1037
- 9 Kakkar S, Narasimhan B. A comprehensive review on biological activities of oxazole derivatives. BMC Chem 2019; 13 (01) 16
- 10 Kulkarni S, Kaur K, Jaitak V. Recent developments in oxazole derivatives as anticancer agents: Review on synthetic strategies, mechanism of action and SAR studies. Anticancer Agents Med Chem 2022; 22 (10) 1859-1882
- 11 Joshi S, Bisht AS, Juyal D. Systematic scientific study of 1, 3-oxazole derivatives as a useful lead for pharmaceuticals: a review. Pharma Innov 2017; 6: 109
- 12 Wasserman HH, Vinick FJ. Mechanism of the Robinson-Gabriel synthesis of oxazoles. J Org Chem 1973; 38: 2407-2408
- 13 Cornforth JW, Cornforth RH. 218. Mechanism and extension of the Fischer oxazole synthesis. J Chem Soc 1949; 1028-1030
- 14 Weitman M, Lerman L, Cohen S. et al. Facile structural elucidation of imidazoles and oxazoles based on NMR spectroscopy and quantum mechanical calculations. Tetrahedron 2010; 66: 1465-1471
- 15 Van Leusen AM, Hoogenboom BE, Siderius H. A novel and efficient synthesis of oxazoles from tosylmethylisocyanide and carbonyl compounds. Tetrahedron Lett 1972; 13: 2369-2372
- 16 Venkatesha NJ, Bhat YS, Prakash BJ. Manifestation of zeolitic pore characteristics of modified montmorillonite in oxazole synthesis by propargylation and cycloisomerization reactions. Appl Catal A Gen 2015; 496: 51-57
- 17 Alghamdi SS, Suliman RS, Alshehri RA, Almahmoud RS, Alhujirey RI. N-Heterocycle derivatives: an update on the biological activity in correlation with computational predictions. J Appl Pharm Sci 2022; 12: 59-77
- 18 Harale AB, Chaudhari SY, Baviskar SN. Novel nitric oxide-donating pyrazoline-coumarin as a cyclooxygenase-2 inhibitors for anti-inflammatory activity. Syst Rev Pharm 2022; 13 (04) 251-255
- 19 Hernández-Cortez E. Non-steroidal anti-inflammatory analgesics in children. Anest Mex 2006; 18: 162
- 20 Han CL, Qu CZ. Cardiovascular risk and safety evaluation of a dual peroxisome proliferator–activated receptor-alpha/gamma agonist, aleglitazar, in patients with type 2 diabetes: a Meta-analysis. J Cardiovasc Pharmacol 2020; 75 (04) 351-357
- 21 Sepúlveda CS, García CC, Damonte EB. Inhibitors of nucleotide biosynthesis as candidates for a wide spectrum of antiviral chemotherapy. Microorganisms 2022; 10 (08) 1631
- 22 Kakkar S, Kumar S, Lim SM. et al. Design, synthesis and biological evaluation of 3-(2-aminooxazol-5-yl)-2H-chromen-2-one derivatives. Chem Cent J 2018; 12 (01) 130
- 23 Bi Y, Gong L, Liu P, Xiong X, Zhao Y. Nuclear ErbB2 represses DEPTOR transcription to inhibit autophagy in breast cancer cells. Cell Death Dis 2021; 12 (04) 397
- 24 Jo SJ, Jiang R, Jung SI, Rhee JH, Kim YR. Restoration of oxacillin susceptibility in MRSA strains by NPPB. Sci Rep 2025; 15 (01) 23739
- 25 Mahwish N, Bairy LK, Srinivasamurthy S. Antivitamins: A silver lining in the era of antimicrobial resistance. J Pharmacol Pharmacother 2022; 13: 5-13
- 26 Roux H, Touret F, Rathelot P, Vanelle P, Roche M. From the “One-Molecule, One-Target, One-Disease” concept towards looking for multi-target therapeutics for treating non-polio enterovirus (NPEV) infections. Pharmaceuticals (Basel) 2024; 17 (09) 1218
- 27 Zheng K, Chen Y, Liu S. et al. Leflunomide: traditional immunosuppressant with concurrent antiviral effects. Int J Rheum Dis 2023; 26 (02) 195-209
- 28 Meyer JH, Matveychuk D, Holt A, Santhirakumar A, Baker GB. Monoamine oxidase inhibitors in depressive disorders. NeuroPsychopharmacotherapy 2021; 26: 1-33
- 29 Tanwar S, Dhingra G, Goyal S, Chaturvedi V, Tanwar K. Beta-2-agonists in the management of asthma and chronic obstructive pulmonary disease. J Pharmaceut Educ Res 2022; 11: 4
- 30 Shahin AI, Zaraei SO, Alzuraiqi S. et al. Evaluation of 2,3-dihydroimidazo[2,1-b]oxazole and imidazo[2,1-b]oxazole derivatives as chemotherapeutic agents. Future Med Chem 2023; 15 (20) 1885-1901
- 31 Guo R, Dowlati A, Dagogo-Jack I. et al. 603O first clinical results from a phase I trial of PRT3789: a first-in-class intravenous SMARCA2 degrader, in patients with advanced solid tumors with a SMARCA4 mutation. Ann Oncol 2024; 35: S483-S484
- 32 Yap TA, Gutierrez M, Iams WT. et al. A phase 2 safety and efficacy study of PRT3789 in combination with pembrolizumab in patients with advanced or metastatic solid tumors and a SMARCA4 mutation. J Clin Oncol 2025; 43: TPS8667-TPS8667
- 33 Kaur R, Sidhu P, Singh S. What failed BIA 10-2474 Phase I clinical trial? Global speculations and recommendations for future phase I trials. J Pharmacol Pharmacother 2016; 7 (03) 120-126
- 34 Epp-Ducharme B, Dunne M, Fan L. et al. Heat-activated nanomedicine formulation improves the anticancer potential of the HSP90 inhibitor luminespib in vitro . Sci Rep 2021; 11 (01) 11103
- 35 Seggewiss-Bernhardt R, Bargou RC, Goh YT. et al. Phase 1/1B trial of the heat shock protein 90 inhibitor NVP-AUY922 as monotherapy or in combination with bortezomib in patients with relapsed or refractory multiple myeloma. Cancer 2015; 121 (13) 2185-2192
- 36 Jin Z. Muscarine, imidazole, oxazole, and thiazole alkaloids. Nat Prod Rep 2011; 28 (06) 1143-1191
- 37 Schwartsmann G, Da Rocha AB, Mattei J, Lopes R. Marine-derived anticancer agents in clinical trials. Expert Opin Investig Drugs 2003; 12 (08) 1367-1383
- 38 Nagle A, Biggart A, Be C. et al. Discovery and characterization of clinical candidate LXE408 as a kinetoplastid-selective proteasome inhibitor for the treatment of leishmaniases. J Med Chem 2020; 63 (19) 10773-10781
- 39 Khare S, Nagle AS, Biggart A. et al. Proteasome inhibition for treatment of leishmaniasis, Chagas disease and sleeping sickness. Nature 2016; 537 (7619) 229-233
- 40 Gujjarappa R, Sravani S, Kabi AK. et al. An overview on biological activities of oxazole, isoxazoles and 1,2,4-oxadiazoles derivatives. In: Swain BP. eds. Nanostructured Biomaterials. Materials Horizons: From Nature to Nanomaterials. Springer, Singapore; 2022: 379-400
- 41 Chiacchio MA, Lanza G, Chiacchio U. et al. Oxazole-based compounds as anticancer agents. Curr Med Chem 2019; 26 (41) 7337-7371
- 42 Padmavathi V, Prema Kumari C, Venkatesh BC, Padmaja A. Synthesis and antimicrobial activity of amido linked pyrrolyl and pyrazolyl-oxazoles, thiazoles and imidazoles. Eur J Med Chem 2011; 46 (11) 5317-5326
- 43 Yan Z, Liu A, Huang M. et al. Design, synthesis, DFT study and antifungal activity of the derivatives of pyrazolecarboxamide containing thiazole or oxazole ring. Eur J Med Chem 2018; 149: 170-181
- 44 Reddy PR, Seenaiah D, Padmaja A, Padmavathi V, Krishna NS. Synthesis, antioxidant, and cytotoxic activities of bis (oxazolyl/thiazolyl/imidazolyl) amidomethanesulfonyl Acetamides. Med Chem Res 2015; 24: 86-98
- 45 Abhale YK, Sasane AV, Chavan AP. et al. Synthesis and antimycobacterial screening of new thiazolyl-oxazole derivatives. Eur J Med Chem 2017; 132: 333-340
- 46 Chavan AP, Deshpande RR, Borade NA. et al. Synthesis of new 1, 3, 4-oxadiazole and benzothiazolylthioether derivatives of 4-arylmethylidene-3-substituted-isoxazol-5 (4H)-one as potential antimycobacterial agents. Med Chem Res 2019; 28: 1873-1884
- 47 Tan Y, He X, Rao B, Cheng B, Song M, Deng X. Synthesis and evaluation of the anticonvulsant activities of triazole-containing benzo [d] oxazoles. Youji Huaxue 2016; 36: 2449
- 48 Mariappan G, Saha BP, Datta S, Kumar D, Haldar PK. Design, synthesis and antidiabetic evaluation of oxazolone derivatives. J Chem Sci 2011; 123: 335-341
- 49 Ferrer Flegeau E, Popkin ME, Greaney MF. Suzuki coupling of oxazoles. Org Lett 2006; 8 (12) 2495-2498
- 50 Anagnostopoulou K, Nannou C, Evgenidou E, Lambropoulou D. Overarching issues on relevant pesticide transformation products in the aquatic environment: a review. Sci Total Environ 2022; 815: 152863
- 51 Carayon C, Fery-Forgues S. 2-Phenylbenzoxazole derivatives: a family of robust emitters of solid-state fluorescence. Photochem Photobiol Sci 2017; 16 (07) 1020-1035
- 52 Ponomarenko SA, Surin NM, Skorotetcky MS. et al. Ultrafast intramolecular energy transfer in a nanostructured organosilicon luminophore based on p-terphenyl and 1,4-bis(5-phenyloxazol-2-yl)benzene. J Mater Chem C Mater Opt Electron Devices 2019; 7: 14612-14624
- 53 Singh BS, Lobo HR, Pinjari DV, Jarag KJ, Pandit AB, Shankarling GS. Ultrasound and deep eutectic solvent (DES): a novel blend of techniques for rapid and energy efficient synthesis of oxazoles. Ultrason Sonochem 2013; 20 (01) 287-293
- 54 Lamberth C. Oxazole and isoxazole chemistry in crop protection. J Heterocycl Chem 2018; 55: 2035-2045
- 55 Kant K, Patel CK, Banerjee S. et al. Recent advancements in strategies for the synthesis of imidazoles, thiazoles, oxazoles, and benzimidazoles. ChemistrySelect 2023; 8: e202303988
- 56 Salam MA, Al-Amin MY, Salam MT. et al. Antimicrobial resistance: a growing serious threat for global public health. Healthcare (Basel) 2023; 11 (13) 1946
- 57 Puri B, Vaishya R, Vaish A. Antimicrobial resistance: current challenges and future directions. Med J Armed Forces India 2025; 81 (03) 247-258
- 58 Grinev VS, Demeshko IA, Evstigneeva SS, Yegorova AY. Synthesis, optical properties, and antibacterial activity of oxazol-5 (4H)-one arylhydrazones. Russ Chem Bull 2023; 72: 1654-1660
- 59 Singagari S, Sundararajan R. Novel pyrazole substituted oxazole derivatives: design, in silico studies, synthesis & biological activities. J Res Pharm 2022; 26: 625-640
- 60 Keivanloo A, Abbaspour S, Sepehri S, Bakherad M. Synthesis, antibacterial activity and molecular docking study of a series of 1,3-oxazole-quinoxaline amine hybrids. Polycycl Aromat Compd 2022; 42: 2378-2391
- 61 Seelam N, Shrivastava SP, Prasanthi S. Synthesis and antimicrobial activity of some novel fused heterocyclic moieties. Org Commun 2013; 6: 78
- 62 Mohanty SK, Khuntia A, Harika MS, Sarangi SP, Susmitha DJ. Synthesis, characterization and antimicrobial activity of some oxazole and thiazole derivatives. Int J Pharm Pharm Sci 2015; 2: 60-66
- 63 Chilumula NR, Gudipati R, Ampati S, Manda S, Gadhe D. Synthesis of some novel methyl-2-(2-(arylideneamino)oxazol-4-ylamino)benzoxazole-5-carboxylate derivatives as antimicrobial agents. Int J Chemtech Res 2010; 1 (02) 1-6
- 64 Dabholkar VV, Syed SA. Synthesis of novel oxazoles and their hydrazones. Rasayan J Chem 2010; 3: 761-765
- 65 Reddy AB, Hymavathi RV, Swamy GN. A new class of multi-substituted oxazole derivatives: synthesis and antimicrobial activity. J Chem Sci 2013; 125: 495-509
- 66 Kamble VS, Habade BM, Patil GK, Agasimundin Y. Synthesis and evaluation of 4-(1-benzofuran-2-yl)-1,3-oxazole-2-amine and its derivatives. Int J Res Pharm Chem 2012; 2: 32-36
- 67 Richardson MD, Warnock DW. Fungal Infection: Diagnosis and Management. John Wiley & Sons; 2012
- 68 Mallikarjuna Rao R, Sreeramulu J, Ravindranath LK. et al. Synthesis and biological screening of some pyridine and pyrrole derivatives of pyrazolo [3,4-c] pyrazoles. J Chem Pharm Res 2012; 4: 17
- 69 Rabaan AA, Sulaiman T, Al-Ahmed SH. et al. Potential strategies to control the risk of antifungal resistance in humans: a comprehensive review. Antibiotics (Basel) 2023; 12 (03) 608
- 70 Kumar S, Jain T, Banerjee D. Fungal diseases and their treatment: a holistic approach. In: Hameed S, Fatima Z. eds. Pathogenicity and Drug Resistance of Human Pathogens: Mechanisms and Novel Approaches. Springer Singapore; 2019: 111-134
- 71 Bąchor U, Brożyna M, Junka A, Chmielarz MR, Gorczyca D, Mączyński M. Novel isoxazole-based antifungal drug candidates. Int J Mol Sci 2024; 25 (24) 13618
- 72 Tlapale-Lara N, López J, Gómez E. et al. Synthesis, in silico study, and in vitro antifungal activity of new 5-(1,3-diphenyl-1H-pyrazol-4-yl)-4-tosyl-4,5-dihydrooxazoles. Int J Mol Sci 2024; 25 (10) 5091
- 73 Ryu CK, Lee RY, Kim NY, Kim YH, Song AL. Synthesis and antifungal activity of benzo[d]oxazole-4,7-diones. Bioorg Med Chem Lett 2009; 19 (20) 5924-5926
- 74 Rawat BS, Shukla SK. Synthesis and evaluation of some new thiazole/oxazole derivatives for their biological activities. World J Pharm Pharm Sci 2016; 5: 1473-1482
- 75 Al-Asady IN, Ali JF. Virulence factors of Mycobacterium tuberculosis . J Res Appl Sci Biotechnol 2023; 2: 221-237
- 76 Yadav K, Prakash S. Tuberculosis: an airborne disease. Int J Microbiol Res 2017; 5: 225-243
- 77 Gupta R, Sharma S, Singh R, Vishwakarma RA, Mignani S, Singh PP. Functionalized nitroimidazole scaffold construction and their pharmaceutical applications: a 1950–2021 comprehensive overview. Pharmaceuticals (Basel) 2022; 15 (05) 561
- 78 Thakare VM, Kadam AB, Shaha A. et al. Design, synthesis, evaluation of benzoxazole derivatives for anti-tubercular and anti-bacterial activity. Cuest Fisioter 2025; 54: 2666-2676
- 79 Shinde SR, Inamdar SN, Obakachi VA. et al. Discovery of oxazole-dehydrozingerone-based hybrid molecules as potential anti-tubercular agents and their docking for MTB DNA gyrase. Results Chem 2022; 4: 100374
- 80 Moraski GC, Franzblau SG, Miller MJ. Utiliztion of the Suzuki coupling to enhance the antituberculosis activity of aryl oxazoles. Heterocycles 2010; 80 (02) 977-988
- 81 Zwawiak J, Olender D, Zwolska Z, Augustynowicz-Kopeć E, Zaprutko L. Synthesis of 2,3-dihydro-7-nitroimidazo[5,1-b]oxazoles as potential tuberculostatic agents. Acta Pol Pharm 2008; 65 (02) 229-233
- 82 Liu J. The “life code”: a theory that unifies the human life cycle and the origin of human tumors. Semin Cancer Biol 2020; 60: 380-397
- 83 Albuquerque TAF, Drummond do Val L, Doherty A, de Magalhães JP. From humans to hydra: patterns of cancer across the tree of life. Biol Rev Camb Philos Soc 2018; 93 (03) 1715-1734
- 84 García JJS, Flores-Álamo M, Martínez-Klimova E, Apan TR, Klimova EI. Diferrocenyl (areno) oxazoles, spiro (arenooxazole) cyclopropenes, quinolines and areno [1,4-] oxazines: synthesis, characterization and study of their antitumor activity. J Organomet Chem 2018; 867: 312-322
- 85 Gillet JP, Calcagno AM, Varma S. et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Natl Acad Sci U S A 2011; 108 (46) 18708-18713
- 86 Rixe O, Ortuzar W, Alvarez M. et al. Oxaliplatin, tetraplatin, cisplatin, and carboplatin: spectrum of activity in drug-resistant cell lines and in the cell lines of the National Cancer Institute's Anticancer Drug Screen panel. Biochem Pharmacol 1996; 52 (12) 1855-1865
- 87 Khowdiary MM, Khan S, Iqbal T. et al. Anti-leukemic profiling of oxazole-linked oxadiazole derivatives: a computational and kinetic approach. Pharmaceuticals (Basel) 2025; 18 (05) 625
- 88 Komirishetti D, Mittapelli V. Synthesis and biological evaluation of 4-(1H-imidazol-2-yl)-2-(pyrimidin-5-yl) oxazoles as potent anticancer agents. Results Chem 2025; 13: 101938
- 89 Premakumari C, Muralikrishna A, Padmaja A. et al. Synthesis, antimicrobial and anticancer activities of amido sulfonamido methane linked bis heterocycles. Arab J Chem 2014; 7: 385-395
- 90 Mathew B, Hobrath JV, Connelly MC, Guy RK, Reynolds RC. Oxazole and thiazole analogs of sulindac for cancer prevention. Future Med Chem 2018; 10 (07) 743-753
- 91 Kachaeva MV, Hodyna DM, Obernikhina NV. et al. Dependence of the anticancer activity of 1,3-oxazole derivatives on the donor/acceptor nature of his substitutes. J Heterocycl Chem 2019; 56: 3122-3134
- 92 Romagnoli R, Baraldi PG, Prencipe F. et al. Synthesis and biological evaluation of 2-methyl-4, 5-disubstituted oxazoles as a novel class of highly potent antitubulin agents. Sci Rep 2017; 7: 46356
- 93 Ranjith R. Synthesis, characterization, and anticancer activity of benzoxazole derivatives. J Chem Pharm Res 2015; 7: 380-387
- 94 Denova LD, Ivanov DD. Influence of oxidative, carbonyl, and nitrosative stresses on the course of chronic kidney disease (analytical review). Kidneys 2022; 11: 53-61
- 95 Saxena P, Selvaraj K, Khare SK, Chaudhary N. Superoxide dismutase as multipotent therapeutic antioxidant enzyme: role in human diseases. Biotechnol Lett 2022; 44 (01) 1-22
- 96 Rao AT, Naveen K, Mutyalanaidu G, Venkanna B, Shree AJ. Design, synthesis, and characterization of novel substituted dihydrooxazoles: biological evaluation and molecular docking. Russ J Org Chem 2022; 202 (58) 125-135
- 97 Sagud I, Skoric I, Burcul F. Naphthoxazoles and heterobenzoxazoles: cholinesterase inhibition and antioxidant activity. Turk J Chem 2019; 43: 118-124
- 98 Kuş C, Uğurlu E, Özdamar ED, Can-Eke B. Synthesis and antioxidant pr operties of new oxazole-5 (4H)-one derivatives. Turk J Pharm Sci 2017; 14 (02) 174-178
- 99 Severin OO, Kachaeva MV, Pilyo SG. et al. Synthesis, characterization, and study of anti-HPV activity and cell cytotoxicity of novel 1,3-oxazole-4- carbonitrile and 4-sulfonylamide-5-phenyl-1, 3- thiazole derivatives in vitro . Lett Appl NanoBioScience 2024; 13: 1-28
- 100 Kachaeva M, Pilyo S, Kornienko A. et al. In vitro activity of novel 1, 3-oxazole derivatives against human papillomavirus. Ibnosina J Med Biomed Sci 2017; 9: 111-118
- 101 Makarov VA, Riabova OB, Granik VG, Wutzler P, Schmidtke M. Novel [(biphenyloxy)propyl]isoxazole derivatives for inhibition of human rhinovirus 2 and coxsackievirus B3 replication. J Antimicrob Chemother 2005; 55 (04) 483-488
- 102 Barry JJ, Huynh N, Lembke A. Depression in individuals with epilepsy. Curr Treat Options Neurol 2000; 2 (06) 571-585
- 103 Löscher W, Klitgaard H, Twyman RE, Schmidt D. New avenues for anti-epileptic drug discovery and development. Nat Rev Drug Discov 2013; 12 (10) 757-776
- 104 Srilakshmi S, Sundararajan R. Design, in silico studies, synthesis, characterization, and anticonvulsant activities of novel thiazole-substituted oxazole derivatives. Rasayan J Chem 2022; 15: 711-725
- 105 Abdellatif KR, Noha H, Asma A, Mohammed A. Synthesis of some benzoxazole derivatives and their anti-inflammatory evaluation. J Chem Pharm Res 2016; 8: 1253-1261
- 106 Song MX, Rao BQ, Cheng BB. et al. Design, synthesis, and evaluation of the antidepressant and anticonvulsant activities of triazole-containing benzo [d] oxazoles. CNS Neurol Disord Drug Targets 2017; 16 (02) 187-198
- 107 Singh BK, Singh UK, Goswami D. Synthesis and anticonvulsant activity (Chemo-Shock) of some potent benzoxazole semicarbazone derivatives as GABA agonists. Saudi J Med Pharm Sci 2018; 4: 806-814
- 108 Marabathuni VJ, Mariyamma K, Sravani K, Ramaiah S, Ravindra P, Srihari R. Synthesis, characterization, anti-microbial, analgesic activities of 3-(3-chlorophenyl)-5-phenyl-4,5-dihydro-1,2- oxazole derivatives. Asian J Pharm Res 2017; 7: 171-174
- 109 Rao P, Knaus EE. Evolution of nonsteroidal anti-inflammatory drugs (NSAIDs): cyclooxygenase (COX) inhibition and beyond. J Pharm Pharm Sci 2008; 11 (02) 81s-110s
- 110 Garg AK, Singh RK, Saxena V, Sinha SK, Rao S. Synthesis, characterization, and anti-inflammatory activity of some novel oxazole derivatives. Drug Delivery Therapeut 2023; 13: 26-28
- 111 Meenakshi M, Kannan A, Jothimani M. et al. Evaluation of dual potentiality of 2,4,5-trisubstituted oxazole derivatives as aquaporin-4 inhibitors and anti-inflammatory agents in lung cells. RSC Adv 2023; 13 (37) 26111-26120
- 112 Shakya AK, Kaur A, Al-Najjar BO, Naik RR. Molecular modeling, synthesis, characterization and pharmacological evaluation of benzo[d]oxazole derivatives as non-steroidal anti-inflammatory agents. Saudi Pharm J 2016; 24 (05) 616-624
- 113 Sarkate AP, Shinde DB. Synthesis and docking studies of 2-(nitrooxy)-ethyl-2-(substituted-2, 5-diphenyl-oxazole)-acetate as anti-inflammatory, analgesic and nitric oxide releasing agents. Int J Pharm Pharm Sci 2015; 7: 128-135
- 114 Lotfy M, Adeghate J, Kalasz H, Singh J, Adeghate E. Chronic complications of diabetes mellitus: a mini review. Curr Diabetes Rev 2017; 13 (01) 3-10
- 115 Jenkins AJ, O'Neal DN, Nolan CJ, Januszewski AS. The pathobiology of diabetes mellitus. Pancreatic Islet Biology 2016; 1-48
- 116 Kumar A, Ahmad P, Maurya RA, Singh AB, Srivastava AK. Novel 2-aryl-naphtho[1,2-d]oxazole derivatives as potential PTP-1B inhibitors showing antihyperglycemic activities. Eur J Med Chem 2009; 44 (01) 109-116
- 117 Husain A, Joshi S, Choudhary AN. et al. Computational design and toxicity prediction of oxazole derivatives targeting PPARγ as potential therapeutics for diabetes mellitus in comparison to rosiglitazone and pioglitazone. J Chil Chem Soc 2024; 69: 6056-6064
Address for correspondence
Publication History
Received: 16 May 2025
Accepted: 22 October 2025
Article published online:
21 November 2025
© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
-
References
- 1 Benton ML, Abraham A, LaBella AL, Abbot P, Rokas A, Capra JA. The influence of evolutionary history on human health and disease. Nat Rev Genet 2021; 22 (05) 269-283
- 2 Filatova EV, Shadrina MI, Slominsky PA. Major depression: one brain, one disease, one set of intertwined processes. Cells 2021; 10 (06) 1283
- 3 Dhameja M, Gupta P. Synthetic heterocyclic candidates as promising α-glucosidase inhibitors: an overview. Eur J Med Chem 2019; 176: 343-377
- 4 Guan Q, Xing S, Wang L. et al. Triazoles in medicinal chemistry: physicochemical properties, bioisosterism, and application. J Med Chem 2024; 67 (10) 7788-7824
- 5 Zhang HZ, Zhao ZL, Zhou CH. Recent advance in oxazole-based medicinal chemistry. Eur J Med Chem 2018; 144: 444-492
- 6 Neha K, Ali F, Haider K, Khasimbi S, Wakode S. Synthetic approaches for oxazole derivatives: a review. Synth Commun 2021; 51: 3501-3519
- 7 Zheng X, Liu W, Zhang D. Recent advances in the synthesis of oxazole-based molecules via van Leusen oxazole synthesis. Molecules 2020; 25 (07) 1594
- 8 Sisco E, Barnes KL. Design, synthesis, and biological evaluation of novel 1,3-oxazole sulfonamides as tubulin polymerization inhibitors. ACS Med Chem Lett 2021; 12 (06) 1030-1037
- 9 Kakkar S, Narasimhan B. A comprehensive review on biological activities of oxazole derivatives. BMC Chem 2019; 13 (01) 16
- 10 Kulkarni S, Kaur K, Jaitak V. Recent developments in oxazole derivatives as anticancer agents: Review on synthetic strategies, mechanism of action and SAR studies. Anticancer Agents Med Chem 2022; 22 (10) 1859-1882
- 11 Joshi S, Bisht AS, Juyal D. Systematic scientific study of 1, 3-oxazole derivatives as a useful lead for pharmaceuticals: a review. Pharma Innov 2017; 6: 109
- 12 Wasserman HH, Vinick FJ. Mechanism of the Robinson-Gabriel synthesis of oxazoles. J Org Chem 1973; 38: 2407-2408
- 13 Cornforth JW, Cornforth RH. 218. Mechanism and extension of the Fischer oxazole synthesis. J Chem Soc 1949; 1028-1030
- 14 Weitman M, Lerman L, Cohen S. et al. Facile structural elucidation of imidazoles and oxazoles based on NMR spectroscopy and quantum mechanical calculations. Tetrahedron 2010; 66: 1465-1471
- 15 Van Leusen AM, Hoogenboom BE, Siderius H. A novel and efficient synthesis of oxazoles from tosylmethylisocyanide and carbonyl compounds. Tetrahedron Lett 1972; 13: 2369-2372
- 16 Venkatesha NJ, Bhat YS, Prakash BJ. Manifestation of zeolitic pore characteristics of modified montmorillonite in oxazole synthesis by propargylation and cycloisomerization reactions. Appl Catal A Gen 2015; 496: 51-57
- 17 Alghamdi SS, Suliman RS, Alshehri RA, Almahmoud RS, Alhujirey RI. N-Heterocycle derivatives: an update on the biological activity in correlation with computational predictions. J Appl Pharm Sci 2022; 12: 59-77
- 18 Harale AB, Chaudhari SY, Baviskar SN. Novel nitric oxide-donating pyrazoline-coumarin as a cyclooxygenase-2 inhibitors for anti-inflammatory activity. Syst Rev Pharm 2022; 13 (04) 251-255
- 19 Hernández-Cortez E. Non-steroidal anti-inflammatory analgesics in children. Anest Mex 2006; 18: 162
- 20 Han CL, Qu CZ. Cardiovascular risk and safety evaluation of a dual peroxisome proliferator–activated receptor-alpha/gamma agonist, aleglitazar, in patients with type 2 diabetes: a Meta-analysis. J Cardiovasc Pharmacol 2020; 75 (04) 351-357
- 21 Sepúlveda CS, García CC, Damonte EB. Inhibitors of nucleotide biosynthesis as candidates for a wide spectrum of antiviral chemotherapy. Microorganisms 2022; 10 (08) 1631
- 22 Kakkar S, Kumar S, Lim SM. et al. Design, synthesis and biological evaluation of 3-(2-aminooxazol-5-yl)-2H-chromen-2-one derivatives. Chem Cent J 2018; 12 (01) 130
- 23 Bi Y, Gong L, Liu P, Xiong X, Zhao Y. Nuclear ErbB2 represses DEPTOR transcription to inhibit autophagy in breast cancer cells. Cell Death Dis 2021; 12 (04) 397
- 24 Jo SJ, Jiang R, Jung SI, Rhee JH, Kim YR. Restoration of oxacillin susceptibility in MRSA strains by NPPB. Sci Rep 2025; 15 (01) 23739
- 25 Mahwish N, Bairy LK, Srinivasamurthy S. Antivitamins: A silver lining in the era of antimicrobial resistance. J Pharmacol Pharmacother 2022; 13: 5-13
- 26 Roux H, Touret F, Rathelot P, Vanelle P, Roche M. From the “One-Molecule, One-Target, One-Disease” concept towards looking for multi-target therapeutics for treating non-polio enterovirus (NPEV) infections. Pharmaceuticals (Basel) 2024; 17 (09) 1218
- 27 Zheng K, Chen Y, Liu S. et al. Leflunomide: traditional immunosuppressant with concurrent antiviral effects. Int J Rheum Dis 2023; 26 (02) 195-209
- 28 Meyer JH, Matveychuk D, Holt A, Santhirakumar A, Baker GB. Monoamine oxidase inhibitors in depressive disorders. NeuroPsychopharmacotherapy 2021; 26: 1-33
- 29 Tanwar S, Dhingra G, Goyal S, Chaturvedi V, Tanwar K. Beta-2-agonists in the management of asthma and chronic obstructive pulmonary disease. J Pharmaceut Educ Res 2022; 11: 4
- 30 Shahin AI, Zaraei SO, Alzuraiqi S. et al. Evaluation of 2,3-dihydroimidazo[2,1-b]oxazole and imidazo[2,1-b]oxazole derivatives as chemotherapeutic agents. Future Med Chem 2023; 15 (20) 1885-1901
- 31 Guo R, Dowlati A, Dagogo-Jack I. et al. 603O first clinical results from a phase I trial of PRT3789: a first-in-class intravenous SMARCA2 degrader, in patients with advanced solid tumors with a SMARCA4 mutation. Ann Oncol 2024; 35: S483-S484
- 32 Yap TA, Gutierrez M, Iams WT. et al. A phase 2 safety and efficacy study of PRT3789 in combination with pembrolizumab in patients with advanced or metastatic solid tumors and a SMARCA4 mutation. J Clin Oncol 2025; 43: TPS8667-TPS8667
- 33 Kaur R, Sidhu P, Singh S. What failed BIA 10-2474 Phase I clinical trial? Global speculations and recommendations for future phase I trials. J Pharmacol Pharmacother 2016; 7 (03) 120-126
- 34 Epp-Ducharme B, Dunne M, Fan L. et al. Heat-activated nanomedicine formulation improves the anticancer potential of the HSP90 inhibitor luminespib in vitro . Sci Rep 2021; 11 (01) 11103
- 35 Seggewiss-Bernhardt R, Bargou RC, Goh YT. et al. Phase 1/1B trial of the heat shock protein 90 inhibitor NVP-AUY922 as monotherapy or in combination with bortezomib in patients with relapsed or refractory multiple myeloma. Cancer 2015; 121 (13) 2185-2192
- 36 Jin Z. Muscarine, imidazole, oxazole, and thiazole alkaloids. Nat Prod Rep 2011; 28 (06) 1143-1191
- 37 Schwartsmann G, Da Rocha AB, Mattei J, Lopes R. Marine-derived anticancer agents in clinical trials. Expert Opin Investig Drugs 2003; 12 (08) 1367-1383
- 38 Nagle A, Biggart A, Be C. et al. Discovery and characterization of clinical candidate LXE408 as a kinetoplastid-selective proteasome inhibitor for the treatment of leishmaniases. J Med Chem 2020; 63 (19) 10773-10781
- 39 Khare S, Nagle AS, Biggart A. et al. Proteasome inhibition for treatment of leishmaniasis, Chagas disease and sleeping sickness. Nature 2016; 537 (7619) 229-233
- 40 Gujjarappa R, Sravani S, Kabi AK. et al. An overview on biological activities of oxazole, isoxazoles and 1,2,4-oxadiazoles derivatives. In: Swain BP. eds. Nanostructured Biomaterials. Materials Horizons: From Nature to Nanomaterials. Springer, Singapore; 2022: 379-400
- 41 Chiacchio MA, Lanza G, Chiacchio U. et al. Oxazole-based compounds as anticancer agents. Curr Med Chem 2019; 26 (41) 7337-7371
- 42 Padmavathi V, Prema Kumari C, Venkatesh BC, Padmaja A. Synthesis and antimicrobial activity of amido linked pyrrolyl and pyrazolyl-oxazoles, thiazoles and imidazoles. Eur J Med Chem 2011; 46 (11) 5317-5326
- 43 Yan Z, Liu A, Huang M. et al. Design, synthesis, DFT study and antifungal activity of the derivatives of pyrazolecarboxamide containing thiazole or oxazole ring. Eur J Med Chem 2018; 149: 170-181
- 44 Reddy PR, Seenaiah D, Padmaja A, Padmavathi V, Krishna NS. Synthesis, antioxidant, and cytotoxic activities of bis (oxazolyl/thiazolyl/imidazolyl) amidomethanesulfonyl Acetamides. Med Chem Res 2015; 24: 86-98
- 45 Abhale YK, Sasane AV, Chavan AP. et al. Synthesis and antimycobacterial screening of new thiazolyl-oxazole derivatives. Eur J Med Chem 2017; 132: 333-340
- 46 Chavan AP, Deshpande RR, Borade NA. et al. Synthesis of new 1, 3, 4-oxadiazole and benzothiazolylthioether derivatives of 4-arylmethylidene-3-substituted-isoxazol-5 (4H)-one as potential antimycobacterial agents. Med Chem Res 2019; 28: 1873-1884
- 47 Tan Y, He X, Rao B, Cheng B, Song M, Deng X. Synthesis and evaluation of the anticonvulsant activities of triazole-containing benzo [d] oxazoles. Youji Huaxue 2016; 36: 2449
- 48 Mariappan G, Saha BP, Datta S, Kumar D, Haldar PK. Design, synthesis and antidiabetic evaluation of oxazolone derivatives. J Chem Sci 2011; 123: 335-341
- 49 Ferrer Flegeau E, Popkin ME, Greaney MF. Suzuki coupling of oxazoles. Org Lett 2006; 8 (12) 2495-2498
- 50 Anagnostopoulou K, Nannou C, Evgenidou E, Lambropoulou D. Overarching issues on relevant pesticide transformation products in the aquatic environment: a review. Sci Total Environ 2022; 815: 152863
- 51 Carayon C, Fery-Forgues S. 2-Phenylbenzoxazole derivatives: a family of robust emitters of solid-state fluorescence. Photochem Photobiol Sci 2017; 16 (07) 1020-1035
- 52 Ponomarenko SA, Surin NM, Skorotetcky MS. et al. Ultrafast intramolecular energy transfer in a nanostructured organosilicon luminophore based on p-terphenyl and 1,4-bis(5-phenyloxazol-2-yl)benzene. J Mater Chem C Mater Opt Electron Devices 2019; 7: 14612-14624
- 53 Singh BS, Lobo HR, Pinjari DV, Jarag KJ, Pandit AB, Shankarling GS. Ultrasound and deep eutectic solvent (DES): a novel blend of techniques for rapid and energy efficient synthesis of oxazoles. Ultrason Sonochem 2013; 20 (01) 287-293
- 54 Lamberth C. Oxazole and isoxazole chemistry in crop protection. J Heterocycl Chem 2018; 55: 2035-2045
- 55 Kant K, Patel CK, Banerjee S. et al. Recent advancements in strategies for the synthesis of imidazoles, thiazoles, oxazoles, and benzimidazoles. ChemistrySelect 2023; 8: e202303988
- 56 Salam MA, Al-Amin MY, Salam MT. et al. Antimicrobial resistance: a growing serious threat for global public health. Healthcare (Basel) 2023; 11 (13) 1946
- 57 Puri B, Vaishya R, Vaish A. Antimicrobial resistance: current challenges and future directions. Med J Armed Forces India 2025; 81 (03) 247-258
- 58 Grinev VS, Demeshko IA, Evstigneeva SS, Yegorova AY. Synthesis, optical properties, and antibacterial activity of oxazol-5 (4H)-one arylhydrazones. Russ Chem Bull 2023; 72: 1654-1660
- 59 Singagari S, Sundararajan R. Novel pyrazole substituted oxazole derivatives: design, in silico studies, synthesis & biological activities. J Res Pharm 2022; 26: 625-640
- 60 Keivanloo A, Abbaspour S, Sepehri S, Bakherad M. Synthesis, antibacterial activity and molecular docking study of a series of 1,3-oxazole-quinoxaline amine hybrids. Polycycl Aromat Compd 2022; 42: 2378-2391
- 61 Seelam N, Shrivastava SP, Prasanthi S. Synthesis and antimicrobial activity of some novel fused heterocyclic moieties. Org Commun 2013; 6: 78
- 62 Mohanty SK, Khuntia A, Harika MS, Sarangi SP, Susmitha DJ. Synthesis, characterization and antimicrobial activity of some oxazole and thiazole derivatives. Int J Pharm Pharm Sci 2015; 2: 60-66
- 63 Chilumula NR, Gudipati R, Ampati S, Manda S, Gadhe D. Synthesis of some novel methyl-2-(2-(arylideneamino)oxazol-4-ylamino)benzoxazole-5-carboxylate derivatives as antimicrobial agents. Int J Chemtech Res 2010; 1 (02) 1-6
- 64 Dabholkar VV, Syed SA. Synthesis of novel oxazoles and their hydrazones. Rasayan J Chem 2010; 3: 761-765
- 65 Reddy AB, Hymavathi RV, Swamy GN. A new class of multi-substituted oxazole derivatives: synthesis and antimicrobial activity. J Chem Sci 2013; 125: 495-509
- 66 Kamble VS, Habade BM, Patil GK, Agasimundin Y. Synthesis and evaluation of 4-(1-benzofuran-2-yl)-1,3-oxazole-2-amine and its derivatives. Int J Res Pharm Chem 2012; 2: 32-36
- 67 Richardson MD, Warnock DW. Fungal Infection: Diagnosis and Management. John Wiley & Sons; 2012
- 68 Mallikarjuna Rao R, Sreeramulu J, Ravindranath LK. et al. Synthesis and biological screening of some pyridine and pyrrole derivatives of pyrazolo [3,4-c] pyrazoles. J Chem Pharm Res 2012; 4: 17
- 69 Rabaan AA, Sulaiman T, Al-Ahmed SH. et al. Potential strategies to control the risk of antifungal resistance in humans: a comprehensive review. Antibiotics (Basel) 2023; 12 (03) 608
- 70 Kumar S, Jain T, Banerjee D. Fungal diseases and their treatment: a holistic approach. In: Hameed S, Fatima Z. eds. Pathogenicity and Drug Resistance of Human Pathogens: Mechanisms and Novel Approaches. Springer Singapore; 2019: 111-134
- 71 Bąchor U, Brożyna M, Junka A, Chmielarz MR, Gorczyca D, Mączyński M. Novel isoxazole-based antifungal drug candidates. Int J Mol Sci 2024; 25 (24) 13618
- 72 Tlapale-Lara N, López J, Gómez E. et al. Synthesis, in silico study, and in vitro antifungal activity of new 5-(1,3-diphenyl-1H-pyrazol-4-yl)-4-tosyl-4,5-dihydrooxazoles. Int J Mol Sci 2024; 25 (10) 5091
- 73 Ryu CK, Lee RY, Kim NY, Kim YH, Song AL. Synthesis and antifungal activity of benzo[d]oxazole-4,7-diones. Bioorg Med Chem Lett 2009; 19 (20) 5924-5926
- 74 Rawat BS, Shukla SK. Synthesis and evaluation of some new thiazole/oxazole derivatives for their biological activities. World J Pharm Pharm Sci 2016; 5: 1473-1482
- 75 Al-Asady IN, Ali JF. Virulence factors of Mycobacterium tuberculosis . J Res Appl Sci Biotechnol 2023; 2: 221-237
- 76 Yadav K, Prakash S. Tuberculosis: an airborne disease. Int J Microbiol Res 2017; 5: 225-243
- 77 Gupta R, Sharma S, Singh R, Vishwakarma RA, Mignani S, Singh PP. Functionalized nitroimidazole scaffold construction and their pharmaceutical applications: a 1950–2021 comprehensive overview. Pharmaceuticals (Basel) 2022; 15 (05) 561
- 78 Thakare VM, Kadam AB, Shaha A. et al. Design, synthesis, evaluation of benzoxazole derivatives for anti-tubercular and anti-bacterial activity. Cuest Fisioter 2025; 54: 2666-2676
- 79 Shinde SR, Inamdar SN, Obakachi VA. et al. Discovery of oxazole-dehydrozingerone-based hybrid molecules as potential anti-tubercular agents and their docking for MTB DNA gyrase. Results Chem 2022; 4: 100374
- 80 Moraski GC, Franzblau SG, Miller MJ. Utiliztion of the Suzuki coupling to enhance the antituberculosis activity of aryl oxazoles. Heterocycles 2010; 80 (02) 977-988
- 81 Zwawiak J, Olender D, Zwolska Z, Augustynowicz-Kopeć E, Zaprutko L. Synthesis of 2,3-dihydro-7-nitroimidazo[5,1-b]oxazoles as potential tuberculostatic agents. Acta Pol Pharm 2008; 65 (02) 229-233
- 82 Liu J. The “life code”: a theory that unifies the human life cycle and the origin of human tumors. Semin Cancer Biol 2020; 60: 380-397
- 83 Albuquerque TAF, Drummond do Val L, Doherty A, de Magalhães JP. From humans to hydra: patterns of cancer across the tree of life. Biol Rev Camb Philos Soc 2018; 93 (03) 1715-1734
- 84 García JJS, Flores-Álamo M, Martínez-Klimova E, Apan TR, Klimova EI. Diferrocenyl (areno) oxazoles, spiro (arenooxazole) cyclopropenes, quinolines and areno [1,4-] oxazines: synthesis, characterization and study of their antitumor activity. J Organomet Chem 2018; 867: 312-322
- 85 Gillet JP, Calcagno AM, Varma S. et al. Redefining the relevance of established cancer cell lines to the study of mechanisms of clinical anti-cancer drug resistance. Proc Natl Acad Sci U S A 2011; 108 (46) 18708-18713
- 86 Rixe O, Ortuzar W, Alvarez M. et al. Oxaliplatin, tetraplatin, cisplatin, and carboplatin: spectrum of activity in drug-resistant cell lines and in the cell lines of the National Cancer Institute's Anticancer Drug Screen panel. Biochem Pharmacol 1996; 52 (12) 1855-1865
- 87 Khowdiary MM, Khan S, Iqbal T. et al. Anti-leukemic profiling of oxazole-linked oxadiazole derivatives: a computational and kinetic approach. Pharmaceuticals (Basel) 2025; 18 (05) 625
- 88 Komirishetti D, Mittapelli V. Synthesis and biological evaluation of 4-(1H-imidazol-2-yl)-2-(pyrimidin-5-yl) oxazoles as potent anticancer agents. Results Chem 2025; 13: 101938
- 89 Premakumari C, Muralikrishna A, Padmaja A. et al. Synthesis, antimicrobial and anticancer activities of amido sulfonamido methane linked bis heterocycles. Arab J Chem 2014; 7: 385-395
- 90 Mathew B, Hobrath JV, Connelly MC, Guy RK, Reynolds RC. Oxazole and thiazole analogs of sulindac for cancer prevention. Future Med Chem 2018; 10 (07) 743-753
- 91 Kachaeva MV, Hodyna DM, Obernikhina NV. et al. Dependence of the anticancer activity of 1,3-oxazole derivatives on the donor/acceptor nature of his substitutes. J Heterocycl Chem 2019; 56: 3122-3134
- 92 Romagnoli R, Baraldi PG, Prencipe F. et al. Synthesis and biological evaluation of 2-methyl-4, 5-disubstituted oxazoles as a novel class of highly potent antitubulin agents. Sci Rep 2017; 7: 46356
- 93 Ranjith R. Synthesis, characterization, and anticancer activity of benzoxazole derivatives. J Chem Pharm Res 2015; 7: 380-387
- 94 Denova LD, Ivanov DD. Influence of oxidative, carbonyl, and nitrosative stresses on the course of chronic kidney disease (analytical review). Kidneys 2022; 11: 53-61
- 95 Saxena P, Selvaraj K, Khare SK, Chaudhary N. Superoxide dismutase as multipotent therapeutic antioxidant enzyme: role in human diseases. Biotechnol Lett 2022; 44 (01) 1-22
- 96 Rao AT, Naveen K, Mutyalanaidu G, Venkanna B, Shree AJ. Design, synthesis, and characterization of novel substituted dihydrooxazoles: biological evaluation and molecular docking. Russ J Org Chem 2022; 202 (58) 125-135
- 97 Sagud I, Skoric I, Burcul F. Naphthoxazoles and heterobenzoxazoles: cholinesterase inhibition and antioxidant activity. Turk J Chem 2019; 43: 118-124
- 98 Kuş C, Uğurlu E, Özdamar ED, Can-Eke B. Synthesis and antioxidant pr operties of new oxazole-5 (4H)-one derivatives. Turk J Pharm Sci 2017; 14 (02) 174-178
- 99 Severin OO, Kachaeva MV, Pilyo SG. et al. Synthesis, characterization, and study of anti-HPV activity and cell cytotoxicity of novel 1,3-oxazole-4- carbonitrile and 4-sulfonylamide-5-phenyl-1, 3- thiazole derivatives in vitro . Lett Appl NanoBioScience 2024; 13: 1-28
- 100 Kachaeva M, Pilyo S, Kornienko A. et al. In vitro activity of novel 1, 3-oxazole derivatives against human papillomavirus. Ibnosina J Med Biomed Sci 2017; 9: 111-118
- 101 Makarov VA, Riabova OB, Granik VG, Wutzler P, Schmidtke M. Novel [(biphenyloxy)propyl]isoxazole derivatives for inhibition of human rhinovirus 2 and coxsackievirus B3 replication. J Antimicrob Chemother 2005; 55 (04) 483-488
- 102 Barry JJ, Huynh N, Lembke A. Depression in individuals with epilepsy. Curr Treat Options Neurol 2000; 2 (06) 571-585
- 103 Löscher W, Klitgaard H, Twyman RE, Schmidt D. New avenues for anti-epileptic drug discovery and development. Nat Rev Drug Discov 2013; 12 (10) 757-776
- 104 Srilakshmi S, Sundararajan R. Design, in silico studies, synthesis, characterization, and anticonvulsant activities of novel thiazole-substituted oxazole derivatives. Rasayan J Chem 2022; 15: 711-725
- 105 Abdellatif KR, Noha H, Asma A, Mohammed A. Synthesis of some benzoxazole derivatives and their anti-inflammatory evaluation. J Chem Pharm Res 2016; 8: 1253-1261
- 106 Song MX, Rao BQ, Cheng BB. et al. Design, synthesis, and evaluation of the antidepressant and anticonvulsant activities of triazole-containing benzo [d] oxazoles. CNS Neurol Disord Drug Targets 2017; 16 (02) 187-198
- 107 Singh BK, Singh UK, Goswami D. Synthesis and anticonvulsant activity (Chemo-Shock) of some potent benzoxazole semicarbazone derivatives as GABA agonists. Saudi J Med Pharm Sci 2018; 4: 806-814
- 108 Marabathuni VJ, Mariyamma K, Sravani K, Ramaiah S, Ravindra P, Srihari R. Synthesis, characterization, anti-microbial, analgesic activities of 3-(3-chlorophenyl)-5-phenyl-4,5-dihydro-1,2- oxazole derivatives. Asian J Pharm Res 2017; 7: 171-174
- 109 Rao P, Knaus EE. Evolution of nonsteroidal anti-inflammatory drugs (NSAIDs): cyclooxygenase (COX) inhibition and beyond. J Pharm Pharm Sci 2008; 11 (02) 81s-110s
- 110 Garg AK, Singh RK, Saxena V, Sinha SK, Rao S. Synthesis, characterization, and anti-inflammatory activity of some novel oxazole derivatives. Drug Delivery Therapeut 2023; 13: 26-28
- 111 Meenakshi M, Kannan A, Jothimani M. et al. Evaluation of dual potentiality of 2,4,5-trisubstituted oxazole derivatives as aquaporin-4 inhibitors and anti-inflammatory agents in lung cells. RSC Adv 2023; 13 (37) 26111-26120
- 112 Shakya AK, Kaur A, Al-Najjar BO, Naik RR. Molecular modeling, synthesis, characterization and pharmacological evaluation of benzo[d]oxazole derivatives as non-steroidal anti-inflammatory agents. Saudi Pharm J 2016; 24 (05) 616-624
- 113 Sarkate AP, Shinde DB. Synthesis and docking studies of 2-(nitrooxy)-ethyl-2-(substituted-2, 5-diphenyl-oxazole)-acetate as anti-inflammatory, analgesic and nitric oxide releasing agents. Int J Pharm Pharm Sci 2015; 7: 128-135
- 114 Lotfy M, Adeghate J, Kalasz H, Singh J, Adeghate E. Chronic complications of diabetes mellitus: a mini review. Curr Diabetes Rev 2017; 13 (01) 3-10
- 115 Jenkins AJ, O'Neal DN, Nolan CJ, Januszewski AS. The pathobiology of diabetes mellitus. Pancreatic Islet Biology 2016; 1-48
- 116 Kumar A, Ahmad P, Maurya RA, Singh AB, Srivastava AK. Novel 2-aryl-naphtho[1,2-d]oxazole derivatives as potential PTP-1B inhibitors showing antihyperglycemic activities. Eur J Med Chem 2009; 44 (01) 109-116
- 117 Husain A, Joshi S, Choudhary AN. et al. Computational design and toxicity prediction of oxazole derivatives targeting PPARγ as potential therapeutics for diabetes mellitus in comparison to rosiglitazone and pioglitazone. J Chil Chem Soc 2024; 69: 6056-6064
























