ABSTRACT
The oxadiazole derivatives are part of the azole family and have drawn significant interest in medicinal chemistry due to their wide range of biological activities and promising pharmacological profiles. This patent spotlight highlights the developments of the oxadiazole-based compounds reported between 2020 and 2024, with applications in treating diseases such as cancer, bacterial infections, metabolic disorders, and neurodegenerative conditions. These compounds act through various mechanisms, including enzyme inhibition, receptor modulation, and disruption of microbial and cancer cell pathways. Their structural flexibility allows for the design of novel molecules targeting specific therapeutic areas. The spotlight on these recent patents underscores oxadiazole derivatives’ growing importance in drug discovery, offering potential advancements in efficacy and safety for future therapeutic agents.
KEYWORDS: Oxadiazole, drug discovery, medicinal applications, patents, biological activities
Heterocyclic compounds are the architects of modern medicinal chemistry, weaving intricate molecular frameworks that unlock the potential for life-saving therapies and transformative innovations.
1. Introduction
Oxadiazole is a class of heterocyclic aromatic chemical compounds belonging to the azole family with the molecular formula C2H2N2O. These compounds are characterized by a five-membered ring containing two nitrogen atoms and one oxygen atom alongside two carbon atoms. First synthesized by Ainsworth in 1965, the calculated resonance energy of oxadiazole is 167.4 kJ/mol, and its thermal stability is enhanced with substitutions [1–4]. The four isomeric forms of the oxadiazole ring are 1,2,3-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, and 1,3,4-oxadiazole, each exhibiting distinct chemical and physical properties that have significant implications for their biological and industrial applications (Figure 1) [5,6].
Figure 1.

Structures of the regioisomeric oxadiazole rings.
According to the Web of Science data, the scientific attention on the application of 1,3,4-oxadiazoles has been continuously rising since the year 2000 (Figure 2) [7]. On the other hand, 1,2,5-oxadiazole derivatives have been found to be mainly used in high energy density materials (HEDMs) as well as biologically active compounds with cytotoxic properties [8–10]. Due to the instability and ring-opening of 1,2,3-oxadiazole heterocycle, resulting in substituted diazomethanes formation, this isomer of oxadiazole is the least of all explored [11]. The unique arrangement of atoms within the oxadiazole ring provides a foundation for a diverse array of chemical reactions and synthetic strategies [12]. The most common synthetic routes involve the cyclization of hydrazides with carboxylic acids, esters, or related derivatives. Such methodologies have been refined over the years, allowing for the efficient and scalable production of oxadiazole derivatives [13–15]. This structural versatility facilitates the design and synthesis of novel compounds tailored for specific functions, whether they be in pharmaceuticals, materials science, or agriculture. The structural configuration of oxadiazoles, particularly the presence of pyridine-type nitrogen atoms, facilitates strong interactions with various enzyme and receptor proteins in the human body. These interactions often result in significant biological effects, which can be harnessed for therapeutic purposes [16,17].
Figure 2.

Number of publications containing the keywords: “1,2,4-oxadiazole” (red), “1,2,5-oxadiazole” (blue), and “1,3,4-oxadiazole” (green) in their title since 1980 [7].
Oxadiazole derivatives have been incorporated into numerous synthetic compounds due to their high potency and therapeutic efficacy in treating various diseases and disorders [18–21]. The molecular architecture of oxadiazoles, including its isomers, is the most extensively studied due to their pronounced pharmacological properties [22]. These compounds have demonstrated a wide range of biological activities including antimicrobial [23,24], anti-inflammatory [25], analgesic [26,27], antifungal [28], antidepressant [29,30], antitubercular [31], anticonvulsant [32], anticholinesterase [33], antihypertensive [34,35], antidiabetic [36,37], antitumor/anticancer [38], antiviral [39], and antioxidant properties [40]. Additionally, oxadiazoles have found applications beyond pharmaceuticals; they have been employed in the creation of supramolecular liquid crystals [41] and high-energy density materials (HEDMs) [42], further demonstrating their utility in materials science. The presence of the oxadiazole ring in natural products, such as Phidianidine A and B, isolated from the sea slug Phidiana militaris, and Quisqualic acid from Quisqualis indica seeds, underscores the natural occurrence and biological relevance of this heterocycle [43,44]. Due to extensive biological effects, oxadiazole moiety has garnered attention from medicinal chemists and pharmacologists in their ongoing development and pharmacological assessment. Research on oxadiazoles has led to the development of compounds with enhanced pharmacological profiles and reduced toxicity. The continuous exploration of oxadiazole-based compounds aims to discover more potent and less toxic derivatives, contributing to the advancement of medical science and the development of new therapeutic agents Figure 3.
Figure 3.

Commercially available oxadiazole-based drugs.
Overall, oxadiazoles represent a versatile and valuable scaffold in medicinal chemistry, offering vast potential for the development of novel drugs and therapeutic strategies. Their extensive range of biological activities and the ongoing research into their applications highlight their significance in pharmaceutical research and drug development. This patent spotlight emphasizes the patents based on the diverse pharmacological properties that have been associated with and contributed to by substituted or derivatized oxadiazoles in the past five years (2020–2024).
2. Medicinal applications of oxadiazole derivatives (2020–2024)
The current patent spotlight addresses the recent breakthroughs of the oxadiazole derivatives with medicinal applications patented in 2020–2024. This study provides a summary of recently released patents on oxadiazole-based drugs, with the following objective of giving thoughts on how these scaffolds, as a privileged structure, could be used in the future (Table 1 and 2). We believe this article will be valuable to organic and medicinal chemists working on drug discovery and development and those interested in using them in chemistry and biology.
Table 1.
Patents based on 1,2,4-oxadiazole derivatives and their medicinal applications.
| Patent no. | Structure | Biological activity/diseases | Tested organisms/cells | Mechanism of action/target | Ref. |
|---|---|---|---|---|---|
| WO2020060964 | ![]() |
Anticancer | Various cancer cell lines | The compounds were tested for their ability to inhibit fibrinolysis. By affecting the activity of serine proteases, these compounds can potentially interfere with cancer progression and metastasis. | [45] |
| WO2020128675 | ![]() |
Antitubercular | M. tuberculosis | Inhibition of critical bacterial enzymes, disruption of the bacterial cell wall or membrane. | [46] |
| WO2020064818 | ![]() |
Anti-inflammatory | Lymphocytes | Sphingosine-1-phosphate receptor agonist, sequestering lymphocytes to peripheral lymphoid organs and away from their sites of chronic inflammation. | [47] |
| WO2020212513 | ![]() |
Fungicidal activity | Fungi and fungal vectors of disease | Damaging cell membranes and inactivating enzymes and proteins. Hence, it interferes with energy production in fungi, which can lead to death. | [48] |
| WO2020245381 | ![]() |
Histone deacetylase 6 (HDAC6) inhibitors | HDAC6 enzyme assay | Compounds were tested in in vitro HDAC6 cell-based assay and were found to inhibit the cellular activity of HDAC6. | [49] |
| WO2020211956 | ![]() |
Antidiabetic agents | Free Fatty Acid Receptor 1 (FFAR1), also known as GPR40 | The derivatives bind to the FFAR1 receptor, promoting insulin secretion only in the presence of high glucose levels, which makes them an ideal candidate for glucose-dependent diabetes treatment. It minimizes the risks associated with nonspecific insulin release. | [50] |
| WO2021173930 | ![]() |
Neurodegenerative diseases | Tetracycline-inducible cell line (HEK-TREX) | To suppress excessive neuronal excitability and gain-of-function mutation in a gene (KCNT1). | [51] |
| WO2021081337 | ![]() |
Anticancer | HTC116 cell line | These compounds are effective at inhibiting neoplastic cell growth. Ribonucleotide reductase (RR) is directly involved in neoplastic tumor growth and drug resistance. | [52] |
| WO2021105857 | ![]() |
Meibomian gland dysfunction | Liver X receptors (LXRs) | The derivatives act as LXR agonists, triggering the activation of genes involved in cholesterol efflux, lipid synthesis, and anti-inflammatory pathways. This mechanism helps regulate lipid homeostasis in the meibomian glands, improving gland function and reducing symptoms of meibomian gland dysfunction (MGD). Additionally, the anti-inflammatory effects of LXR activation help manage chronic inflammation associated with the condition. | [53] |
| WO2022149925 | ![]() |
Metabolic diseases | Tryptophan hydroxylase 1 (TPH1) | By inhibiting TPH1, these compounds decrease serotonin levels in peripheral tissues. This action does not affect the central nervous system, which may be advantageous for avoiding central side effects. | [54] |
| CN114195776 | ![]() |
Anti-inflammatory | Farnesoid X receptor (FXR) | Oxadiazole derivatives are effective FXR agonists. These compounds can be used to treat a range of conditions by modulating bile acid, glucose, and lipid metabolism, as well as inflammation and liver fibrosis. | [55] |
| IN202321033845 | ![]() |
Anticancer activity | HT29, MDA-MB-231, and A549 cell lines | Induction of cancer cell death, assessed by MTT assay and IC50 values calculation. | [56] |
| Antimicrobial activity | E. coli, P. mirabilis, B. subtilis, and S. albus. | Interference with bacterial cell wall synthesis and replication | |||
| Antifungal activity | Fungal strains, e.g., Candida albicans and Aspergillus niger | Disruption of fungal cell membrane integrity and function | |||
| US2023019648 | ![]() |
Epilepsy (EIMFS) | Cell lines expressing KCNT1 | Inhibition of KCNT1 potassium channel | [57] |
| Neurological disorders associated with KCNT1 mutations | Various neuronal cell lines | Blockage of potassium channel leading to reduced neuronal excitability | |||
| US11952354 | ![]() |
Anticancer | Various cancer cell lines (e.g., HeLa, MCF-7, A549, PC-3) | Induction of apoptosis, inhibition of cell proliferation, disruption of mitochondrial membrane potential | [58] |
| Antimicrobial | Bacterial strains (e.g., S. aureus, E. coli) and fungal strains (e.g., C. albicans) | Inhibition of bacterial cell wall synthesis, disruption of microbial cell membranes | |||
| US11919871 | ![]() |
Anticancer | Various cancer cell lines (e.g., HeLa, MCF-7, A549, PC-3) | Induction of apoptosis, inhibition of cell proliferation, disruption of mitochondrial membrane potential | [59] |
| Antimicrobial | Bacterial strains (e.g., S. aureus, E. coli) and fungal strains (e.g., C. albicans) | Inhibition of bacterial cell wall synthesis, disruption of microbial cell membranes | |||
| WO2024028611 | ![]() |
OCD symptom reduction | Rodent models of OCD (e.g., mice, rats). Neuronal cell lines (e.g., SH-SY5Y, HEK293). | Modulation of serotonin receptors (5-HT receptors). Dopamine receptors. Potential inhibition of enzymes involved in neurotransmitter metabolism (e.g., monoamine oxidase). | [60] |
| General anxiety disorders | Models of anxiety-related behaviors. Cortical neurons are other relevant brain cell types. | Potential impact on GABAergic systems or other anxiety-related neurotransmitter systems. | |||
| EP2023069936 | ![]() |
HDAC6 Inhibition | Recombinant HDAC6 enzyme | Selective inhibition of HDAC6 enzyme, affecting cellular processes like protein degradation and stress response | [61] |
| Tubulin acetylation | 697 human leukemia cells, murine neuroblastoma N2a cells, SH-SY5Y neuroblastoma cells. | Induction of tubulin acetylation, indicating effective inhibition of HDAC6. High activity was observed in most cell lines with various potencies. | |||
| EP2023076346 | ![]() |
Fungicidal activity | Various fungal pathogens such as Puccinia recondita and Phakopsora pachyrhizi | Inhibits critical fungal biological processes | [62] |
| US2023076849 | ![]() |
Antibacterial activity | C. difficile ATCC 43,255 strains | Bactericidal against vegetative cells, targets cell-wall synthesis | [63] |
Table 2.
Patents based on 1,3,4-oxadiazole derivatives and their medicinal applications.
| Patent no. | Structure | Biological activity/diseases | Tested organisms/cells | Mechanism of action/target | Ref. |
|---|---|---|---|---|---|
| WO2020030613 | ![]() |
Anticancer | Various cancer cell lines | These oxadiazole derivatives specifically target the ATP binding site of the 6-phosphofructo-1-kinase (PFK-1) enzyme in cancer cells. By inhibiting this enzyme, the compounds reduce glycolytic activity, thereby decreasing NADH and lactate production. This reduction in lactate can potentially hinder the cancer cell’s ability to evade the immune system and reduce their capacity for angiogenesis and metastasis, making these compounds promising candidates for cancer therapy. | [64] |
| WO2020061216 | ![]() |
Anticancer | Myeloid blood cells | A combination therapy for treating blood cancers, particularly acute myeloid leukemia (AML), using Axl kinase inhibitors and BCL-2 inhibitors. The synergy between these two types of inhibitors can improve treatment efficacy by simultaneously disrupting cancer cell survival mechanisms and promoting apoptosis. | [65] |
| WO2020219792 | ![]() |
Antibacterial agents | S. aureus, MRSA, E. faecalis | Interference with bacterial cell wall synthesis. Inhibition of essential bacterial enzymes. Disruption of DNA/RNA synthesis or function. | [66] |
| WO2021070091 | ![]() |
Neurodegenerative diseases | Muscarinic acetylcholine receptor M4 | These derivatives function as M4 receptor agonists. These compounds are designed for the treatment of diseases mediated by the M4 receptor, including cognitive disorders, psychotic disorders, and potentially other related conditions. | [67] |
| CN113461630 | ![]() |
Insecticidal agents | Binding assays and insect bioassays | The binding phase of the cartesian ketone derivative enhances its efficacy against insect pests. | [68] |
| CN113461631 | ![]() |
Antiviral agents | Neuraminidase enzyme | The derivatives inhibit the neuraminidase enzyme, blocking the release of new viral particles from infected host cells. It halts viral replication and helps control infections, particularly in the treatment of influenza. The mode of action is based on binding to and inactivating neuraminidase, preventing the virus from detaching from the host cell. | [69] |
| WO2022029041 | ![]() |
HDAC6-related diseases | Histone Deacetylase 6 (HDAC6) | These compounds are designed to selectively inhibit HDAC6, with potential applications in treating a variety of medical conditions, especially those related to inflammatory responses, cancer, and neurodegeneration. | [70] |
| WO2023244672 | ![]() |
Enhancement of innate immune response | Human peripheral blood mononuclear cells (PBMCs), Mouse splenocytes | Stimulates immune cell activation; increases production of interferons and other cytokines. | [71] |
| Viral replication inhibition | Human cell lines (e.g., HeLa, Vero cells) | Inhibits viral replication by enhancing immune response; potentially targets viral entry or replication pathways. | |||
| Antiviral | Mouse models infected with various viruses (e.g., influenza, hepatitis) | Reduces viral load by boosting the body’s natural antiviral defenses; may involve modulation of immune signaling pathways. | |||
| US11998532 | ![]() |
Antitubercular | Mycobacterium tuberculosis cultures. Animal models of tuberculosis (e.g., mice, rats). | Interference with bacterial cell wall synthesis. Inhibition of essential bacterial enzymes. Disruption of DNA/RNA synthesis or function. | [72] |
| WO2023224371 | ![]() |
Neuroinflammatory diseases | NOX Enzyme | The oxadiazole derivative compounds inhibit NADPH oxidase (NOX) enzymes. NOX enzymes are involved in producing reactive oxygen species (ROS), which play a role in neuroinflammation. By inhibiting NOX2 and NOX4, these compounds reduce oxidative stress and inflammation in the brain. | [73] |
| IN202341033384 | ![]() |
Anticancer activity | PC3, A549, MCF-7, DU-145 | Inhibition of USP28‘s enzymatic activity leads to cell cycle arrest, apoptosis, inhibition of cell proliferation, anti-metastatic, and anti-angiogenic effects. | [74] |
| US2023/017894 | ![]() |
Anticancer activity | Various cancer cell lines | Inhibition of HDAC6 induces cell cycle arrest apoptosis and also reduces tumor growth. | [75] |
| Neurodegenerative diseases | Neuronal cell models | HDAC6 inhibition results in the reduction of protein aggregation and improves protein homeostasis | |||
| Inflammatory disorders | Inflammatory cell models | HDAC6 inhibition modulates inflammatory pathways | |||
| IB2023050658 | ![]() |
Pulmonary arterial hypertension | HPAECs, HPAECs under TGF-β stimulation, SD rats | Increasing acetylation levels at low compound concentrations improves cell viability and inhibits smooth muscle cell proliferation. | [76] |
| IB2023050659 | ![]() |
Improved cardiac function | H9c2 cells, male rabbits, beagle dogs, Sprague-Dawley rats | Stabilizes microtubules, enhances ventricular function, reduces fibrosis, stabilizes Ca2+ transients, restores acetylated tubulin levels | [77] |
| US11958843 | ![]() |
Antibacterial | Bacterial strains (e.g., S. aureus, E. coli, P. aeruginosa) | Inhibition of bacterial cell wall synthesis, disruption of bacterial cell membranes | [78] |
| Antifungal | Fungal strains (e.g., C. albicans, A. niger) | Disruption of fungal cell membranes, inhibition of ergo sterol synthesis | |||
| WO2024054071 | ![]() |
HDAC6-related diseases | Histone Deacetylase 6 (HDAC6) | By blocking HDAC6, these compounds affect the acetylation status of target proteins, leading to changes in gene expression and cellular functions. | [79] |
| KR2023013453 | ![]() |
HDAC6 Inhibition | Human recombinant HDAC1 and HDAC6 enzymes | Inhibition of HDAC6 enzyme activity with high selectivity over HDAC1 | [80] |
| IB2023057181 | ![]() |
HDAC6 inhibition | In vitro with HDAC1 and HDAC6 enzymes | HDAC6 enzyme inhibition | [81] |
| Neurodegenerative disease | Sprague-Dawley rat hippocampal neurons | Increased tubulin acetylation, improved mitochondrial transport |
3. In-depth analysis
Over the past few years, oxadiazole derivatives have emerged as a pivotal class of heterocycles in medicinal chemistry, demonstrating exceptional versatility in drug design and development. Their rigid, electron-rich framework – particularly evident in the 1,3,4- and 1,2,4-oxadiazole isomers – has been central to the modulation of diverse biological targets, offering an ideal scaffold for structural manipulation. This review captures recent advances from 2020 to 2024, focusing on patented innovations that reflect a surge in interest toward oxadiazole-based pharmacophores. A comparative analysis of key patents reveals significant therapeutic relevance across multiple disease areas, including cancer, bacterial infections, neuroinflammation, and viral diseases such as influenza and SARS-CoV-2 (Table 3). The high prevalence of 1,3,4-oxadiazoles in clinical patent filings underscores their superior biological activity, which is often linked to favorable binding interactions with enzymes such as HDAC6, PFKFB3, neuraminidase, and Axl kinase. Synthetic strategies employed in these patents commonly involve facile ring closures, microwave-assisted synthesis, and strategic hybridization with pharmacophores like quinolones, benzimidazoles, or triazoles to boost efficacy and broaden pharmacological profiles.
Table 3.
Most impactful patents on oxadiazole derivatives.
| Patent no. | Biological activity | Tested organisms/cells | Mechanism of action/target | Reference |
|---|---|---|---|---|
| WO2020060964 | Anticancer | Various cancer cell lines | It inhibits fibrinolysis, affects serine protease activity, and potentially prevents metastasis. | [46] |
| WO2020128675 | Antitubercular | Mycobacterium tuberculosis | It inhibits critical bacterial enzymes, disrupting bacterial cell walls/membranes. | [49] |
| WO2020211956 | Antidiabetic | Free Fatty Acid Receptor 1 (FFAR1) | Enhances glucose-dependent insulin secretion, minimizing hypoglycemia risk. | [54] |
| WO2021081337 | Anticancer | HTC116 cell line | It inhibits ribonucleotide reductase (RR), which is involved in tumor growth and drug resistance. | [57] |
| WO2021105857 | Meibomian Gland Dysfunction | Liver X receptors (LXRs) | It acts as an LXR agonist, regulating lipid metabolism and inflammation in glands. | [59] |
| WO2022149925 | Metabolic Diseases | Tryptophan hydroxylase 1 (TPH1) | Inhibits TPH1 to decrease peripheral serotonin, avoiding central nervous system side effects. | [62] |
| CN114195776 | Anti-inflammatory | Farnesoid X receptor (FXR) | It modulates bile acid, glucose, and lipid metabolism, a potential treatment for liver fibrosis. | [63] |
| US2023019648 | Epilepsy (EIMFS) | Cell lines expressing KCNT1 | It inhibits the KCNT1 potassium channel, reducing neuronal excitability. | [70] |
| US11952354 | Anticancer | HeLa, MCF-7, A549, PC-3 cell lines | Induces apoptosis, inhibits proliferation, and disrupts mitochondrial membrane potential. | [73] |
| US11952354 | Antimicrobial | S. aureus, E. coli, C. albicans | It inhibits bacterial cell wall synthesis and disrupts microbial membranes. | [73] |
Furthermore, the structure-activity relationship (SAR) findings across several patents elucidate how subtle substitutions significantly influence potency, selectivity, and ADME profiles. For instance, halogenation or alkyl substitution at the 2- or 5-positions of the oxadiazole ring can enhance lipophilicity and cell membrane penetration. At the same time, polar groups such as hydroxyl, methoxy, or sulfonamides contribute to increased hydrogen bonding with the target protein’s active site. In many cases, the incorporation of oxadiazole into molecular hybrids has been shown to result in dual or even multi-targeted activity, particularly valuable in managing multifactorial diseases such as cancer or type 2 diabetes. In silico docking and ADME/T predictions from various patents have provided additional support for these analogs, revealing excellent bioavailability, metabolic stability, and desirable pharmacokinetic properties.
Importantly, this review identifies several high-impact patents, which have been summarized in focused tables for clarity. These include compounds undergoing preclinical evaluation for HDAC6 inhibition with potential applications in neurodegenerative diseases, as well as oxadiazole-triazole hybrids that show broad-spectrum antiviral activity. The clinical relevance of these innovations is further underscored by their mechanism-driven design, low cytotoxicity profiles, and promising lead optimization data. Not only do these findings validate oxadiazoles as a privileged structure in drug discovery, but they also pave the way for future exploration of novel analogs using fragment-based or AI-guided design strategies. Collectively, the growing patent landscape reflects a paradigm shift toward oxadiazole-centered scaffolds as essential components in next-generation therapeutics, affirming their enduring value in medicinal and pharmaceutical chemistry.
4. Conclusion
The exploration of oxadiazole derivatives in recent years has solidified their position as a versatile and promising scaffold in medicinal chemistry. This patent spotlight showcases numerous derivatives that demonstrate potential across various therapeutic domains, including cancer, bacterial infections, metabolic diseases, and neurodegenerative disorders. Their structural versatility, coupled with strong interactions with biological targets, underlines their significance in drug discovery and development. Among the three classes of oxadiazole derivatives, the 1,3,4-oxadiazole scaffold emerges as the most promising for future research, owing to its broad-spectrum biological activities, favorable pharmacokinetic properties, and its versatile potential in the design of novel therapeutics targeting diverse diseases. The continuous emergence of patents in this domain, particularly from 2020 to 2024, highlights the growing interest and progress in tailoring oxadiazole-based compounds for targeted therapeutic applications while maintaining enhanced pharmacological and improved safety profiles.
5. Future perspectives
Looking ahead, the future of oxadiazole-based drug design lies in further refining their selectivity and reducing toxicity, ensuring that these compounds meet clinical efficacy and safety standards. The development of multi-targeted oxadiazole derivatives capable of addressing complex diseases, such as cancer and neurodegenerative disorders, could offer new avenues for therapeutic intervention. Advances in computational chemistry and high-throughput screening will likely play an instrumental role in identifying novel bioactive oxadiazole derivatives. Additionally, exploring the combination of oxadiazole scaffolds with other privileged structures could lead to the discovery of hybrid compounds with synergistic biological effects. The continued innovation in this field promises to shape the future landscape of medicinal chemistry, delivering more effective and safer treatments for a wide range of diseases.
Acknowledgments
Please refer to the Author Disclosure Form regarding inclusion of individuals in the author list, versus acknowledging their contributions in this section.
Funding Statement
The work was funded by the Pakistan Academy of Sciences (PAS Project no. 111).
Article highlights
Oxadiazoles are versatile heterocycles with broad biological activity, making them key in drug discovery.
Patents from 2020–2024 reveal oxadiazole derivatives’ potential in treating cancer, infections, and neurodegenerative diseases.
These compounds act via diverse mechanisms like enzyme inhibition and receptor modulation.
The four oxadiazole isomers offer structural flexibility for designing targeted therapies.
Scientific interest in 1,3,4-oxadiazoles has surged steadily since 2000.
1,3,4-Oxadiazoles stand out for their potent pharmacological profiles and drug-like properties.
Disclosure statement
No potential conflict of interest was reported by the author(s).
Author contributions
M Solangi and F Naz performed the literature survey, organized and sorted out the literature, and wrote their opinions. SA Ali and KM Khan conceived the idea of the opinion, supervised it, provided critical feedback, and finalized the article.
Financial disclosure
The authors also acknowledge the financial support of the Pakistan Academy of Science, 3-Constitution Avenue, G-5/2, Islamabad-44000, Pakistan, under PAS Project No. 111.
Information pertaining to writing assistance
The authors declare no conflict of interest, financial or otherwise. No writing assistance was utilized in the production of this manuscript.
Data sharing statement
Data will be available upon request.
References
Papers of special note have been highlighted as either of interest (•) or of considerable interest (••) to readers.
- 1.Kaur M, Singh S, Kaur M.. Synthesis, spectral study, and biological activity of some 2,5-disubstituted-1,3,4-oxadiazole. Eur J Med Res. 2018;5(9):277–282. [Google Scholar]
- 2.Ainsworth C. 1,3,4-Oxadiazole. J Am Chem Soc. 1965;87(24):5800–5801. doi: 10.1021/ja00952a056 [DOI] [Google Scholar]
- 3.Scriven EFV. Comprehensive heterocyclic chemistry. Pergamon Press; 1984. [Google Scholar]
- 4.Bird CW. A new aromaticity index and its application to five-membered ring heterocycles. Tetrahedron. 1985;41(7):1409–1414. doi: 10.1016/S0040-4020(01)96543-3 [DOI] [Google Scholar]
- 5.Siwach A, Verma PK. Therapeutic potential of oxadiazole or oxadiazole-containing compounds. BMC Chem. 2020;14(1):1–40. doi: 10.1186/s13065-020-00721-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Vaidya A, Jain S, Jain P, et al. Synthesis and biological activities of oxadiazole derivatives: a review. Mini-Rev Med Chem. 2016;16(10):825–845. doi: 10.2174/1389557516666160211120835 [DOI] [PubMed] [Google Scholar]
- 7.Web of Science . [cited 2024 Jun 16]. Available from: http://www.webofknowledge.com
- 8.Wei H, He C, Zhang J, et al. Combination of 1,2,4‐oxadiazole and 1,2,5‐oxadiazole moieties for the generation of high‐performance energetic materials. Angew Chem Int Ed. 2015;54(32):9367–9371. doi: 10.1002/anie.201503532 [DOI] [PubMed] [Google Scholar]
- 9.Boiani M, Cerecetto H, González M, et al. 1,2,5-oxadiazole N-oxide derivatives as potential anticancer agents: synthesis and biological evaluation. Part IV. Eur J Med Chem. 2001;36(10):771–782. doi: 10.1016/S0223-5234(01)01265-X [DOI] [PubMed] [Google Scholar]
- 10.Xia W, Zhang R, Xu X, et al. Theoretical studies of novel high energy density materials based on oxadiazoles. J Mol Model. 2021;27(7):204. doi: 10.1007/s00894-021-04805-1 [DOI] [PubMed] [Google Scholar]
- 11.Nguyen MT, Hegarty AF, Elguero J. Can 1,2,3‐Oxadiazole be stable? Angew Chem Int Ed. 1985;24(8):713–715. doi: 10.1002/anie.198507131 [DOI] [Google Scholar]
- 12.Banik BK, Sahoo BM, Kumar BVVR, et al. Green synthetic approach: an efficient eco-friendly tool for the synthesis of biologically active oxadiazole derivatives. Molecules. 2021;26(4):1163. doi: 10.3390/molecules26041163 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Redhu S, Kharb R. Recent updates on chemistry and pharmacological aspects of the 1,3,4-oxadiazole scaffold. Int J Pharm Res. 2013;3(1):93–110. [Google Scholar]
- 14.Tiemann F, Krüger P. Ueber amidoxime und azoxime. Ber Dtsch Chem Ges. 1884;17(2):1685–1698. doi: 10.1002/cber.18840170230 [DOI] [Google Scholar]
- 15.Desai AR, Desai KR. Heterocyclic synthesis: a convenient route to some 2‐mercepto 1,3,4‐oxadiazole and green chemistry microwave‐induced one‐pot synthesis of 2‐aryl-1,3,4‐oxadiazole in quinazolone and their antibacterial and antifungal activity. J Heterocycl Chem. 2005;42(5):995–999. doi: 10.1002/jhet.5570420539 [DOI] [Google Scholar]
- 16.Nagaraj CK, Niranjan MS, Kiran S. 1,3,4-oxadiazole: a potent drug candidate with various pharmacological activities. Int J Pharm Pharm Sci. 2011;3(3):9–16. [Google Scholar]
- 17.Patani GA, LaVoie EJ. Bioisosterism: a rational approach in drug design. Chem Rev. 1996;96(8):3147–3176. doi: 10.1021/cr950066q [DOI] [PubMed] [Google Scholar]
- 18.Kumar KA, Jayaroopa P, Kumar GV. Comprehensive review on the chemistry of 1,3,4-oxadiazoles and their applications. Int J Chemtech Res. 2012;4(4):1782–1791. [Google Scholar]
- 19.Somani RR, Shirodkar PY. Oxadiazole: a biologically important heterocycle. Pharm Chem J. 2009;1(1):130–140. [Google Scholar]
- 20.Biernacki K, Daśko M, Ciupak O, et al. Novel 1,2,4-oxadiazole derivatives in drug discovery. Pharmaceuticals. 2020;13(6):111. doi: 10.3390/ph13060111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Luczynski M, Kudelko A. Synthesis and biological activity of 1,3,4-oxadiazoles used in medicine and agriculture. Appl Sci. 2022;12(8):3756. doi: 10.3390/app12083756 [DOI] [Google Scholar]
- 22.Saha R, Tanwar O, Marella A, et al. Recent updates on biological activities of oxadiazoles. Mini-Rev Med Chem. 2013;13(7):1027–1046. doi: 10.2174/1389557511313070007 [DOI] [PubMed] [Google Scholar]
- 23.Glomb T, Świątek P. Antimicrobial activity of 1,3,4-oxadiazole derivatives. Int J Mol Sci. 2021;22(13):6979. doi: 10.3390/ijms22136979 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Kaplancikli ZA, Altintop MD, Turan-Zitouni G, et al. Synthesis, antimicrobial activity, and cytotoxicity of novel oxadiazole derivatives. J Enzym Inhib Med Chem. 2012;27(1):51–57. doi: 10.3109/14756366.2011.574132 [DOI] [PubMed] [Google Scholar]
- 25.Hamoud MM, Osman NA, Rezq S, et al. Design and synthesis of novel 1,3,4-oxadiazole and 1,2,4-triazole derivatives as cyclooxygenase-2 inhibitors with anti-inflammatory and antioxidant activity in LPS-stimulated RAW264.7 macrophages. Bioorg Chem. 2022;124:105808. doi: 10.1016/j.bioorg.2022.105808 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chawla G. 1,2,4-oxadiazole as a privileged scaffold for anti-inflammatory and analgesic activities: a review. Mini-Rev Med Chem. 2018;18(18):1536–1547. doi: 10.2174/1389557518666180524112050 [DOI] [PubMed] [Google Scholar]
- 27.Ramaprasad GC, Kalluraya B, Kumar BS, et al. Synthesis of new oxadiazole derivatives as anti-inflammatory, analgesic, and antimicrobial agents. Med Chem Res. 2013;22(11):5381–5389. doi: 10.1007/s00044-012-0298-1 [DOI] [Google Scholar]
- 28.Karaburun AÇ, Çavuşoğlu BK, Acar Çevik U, et al. Synthesis and antifungal potential of some novel benzimidazole-1,3,4-oxadiazole compounds. Molecules. 2019;24(1):191. doi: 10.3390/molecules24010191 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Sahu B, Bhatia R, Kaur D, et al. Design, synthesis, and biological evaluation of oxadiazole clubbed piperazine derivatives as potential antidepressant agents. Bioorg Med Chem. 2023;136:106544. doi: 10.1016/j.bioorg.2023.106544 [DOI] [PubMed] [Google Scholar]
- 30.Wang S, Qi L, Liu H, et al. Synthesis of 1,3,4-oxadiazoles derivatives with antidepressant activity and their binding to the 5-HT1A receptor. RSC Adv. 2020;10(51):30848–30857. doi: 10.1039/D0RA05886F [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Verma SK, Verma R, Verma S, et al. Anti-tuberculosis activity and its structure-activity relationship (SAR) studies of oxadiazole derivatives: a key review. Eur J Med Chem. 2021;209:112886. doi: 10.1016/j.ejmech.2020.112886 [DOI] [PubMed] [Google Scholar]
- 32.Nazar S, Siddiqui N, Alam O. Recent progress of 1,3,4‐oxadiazoles as anticonvulsants: future horizons. Arch Pharm. 2020;353(7):1900342. doi: 10.1002/ardp.201900342 [DOI] [PubMed] [Google Scholar]
- 33.Mishra P, Sharma P, Tripathi PN, et al. Design and development of 1,3,4-oxadiazole derivatives as potential inhibitors of acetylcholinesterase to ameliorate scopolamine-induced cognitive dysfunctions. Bioorg Chem. 2019;89:103025. doi: 10.1016/j.bioorg.2019.103025 [DOI] [PubMed] [Google Scholar]
- 34.Zhu W, Bao X, Ren H, et al. Design, synthesis, and pharmacological evaluation of 5-oxo-1,2,4-oxadiazole derivatives as AT1 antagonists with antihypertension activities. Clin Exp Hypertens. 2016;38(5):435–442. doi: 10.3109/10641963.2016.1151527 [DOI] [PubMed] [Google Scholar]
- 35.Attari Z, Mudgal J, Nayak PG, et al. Endothelium-dependent and independent mechanisms of vasorelaxant activity of synthesized 2,5-disubstituted-1,3,4-oxadiazole derivatives in rat thoracic aorta–ex vivo and molecular docking studies. Lett Drug Des Discov. 2016;13(5):441–450. doi: 10.2174/1570180812666150907203634 [DOI] [Google Scholar]
- 36.Nazir M, Abbasi MA, Siddiqui SZ, et al. New indole-based hybrid oxadiazole scaffolds with N-substituted acetamides: As potent antidiabetic agents. Bioorg Chem. 2018;81:253–263. doi: 10.1016/j.bioorg.2018.08.010 [DOI] [PubMed] [Google Scholar]
- 37.Ibrahim MT, Uzairu A, Shallangwa GA, et al. In-silico studies of some oxadiazole derivatives as antidiabetic compounds. J King Saud Univ Sci. 2020;32(1):423–432. doi: 10.1016/j.jksus.2018.06.006 [DOI] [Google Scholar]
- 38.Stecoza CE, Nitulescu GM, Draghici C, et al. Synthesis and anticancer evaluation of new 1,3,4-oxadiazole derivatives. Pharmaceuticals. 2021;14(5):438. doi: 10.3390/ph14050438 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Li Z, Zhan P, Liu X. 1,3,4-Oxadiazole: A privileged structure in antiviral agents. Mini-Rev Med Chem. 2011;11(13):1130–1142. doi: 10.2174/138955711797655407 [DOI] [PubMed] [Google Scholar]
- 40.Kotaiah Y, Harikrishna N, Nagaraju K, et al. Synthesis and antioxidant activity of 1,3,4-oxadiazole tagged thieno[2,3-d]pyrimidine derivatives. Eur J Med Chem. 2012;58:340–345. doi: 10.1016/j.ejmech.2012.10.007 [DOI] [PubMed] [Google Scholar]
- 41.Han J, Geng Q, Chen W, et al. Self-assembled liquid crystals formed by hydrogen bonding between non-mesogenic 1,3,4-oxadiazole-based pyridines and substituted benzoic acids. Supr Chem. 2012;24(3):157–164. doi: 10.1080/10610278.2011.638378 [DOI] [Google Scholar]
- 42.Fershtat LL, Makhova NN. 1,2,5‐Oxadiazole‐based high‐energy‐density materials: synthesis and performance. ChemPluschem. 2020;85(1):13–42. doi: 10.1002/cplu.201900542 [DOI] [Google Scholar]
- 43.Liu J, Li XW, Guo YW. Recent advances in the isolation, synthesis, and biological activity of marine guanidine alkaloids. Mar Drugs. 2017;15(10):324. doi: 10.3390/md15100324 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Mahajan CP, Aher AN. A review on ethnobotanical, phytochemical, and pharmacological activities of quisqualis indica Linn. RJPP. 2017;9(1):47–52. doi: 10.5958/0975-4385.2017.00008.5 [DOI] [Google Scholar]; ••• Important patents related to oxadiazole moieties.
- 45.Francisco-Javier P, Luis C. 1,2,4-oxadiazole derivatives and uses thereof. WO2020060964. 2020.
- 46.Roh J, Karabanovich G, Pavek P, et al. Substituted 1,2,4-oxadiazole, its application, and a pharmaceutical preparation comprising it. WO2020128675. 2020.
- 47.Barreca G, Carcone L, Taddei M, et al. Process for the preparation of ozanimod as sphingosine-1-phosphate receptor agonist useful as an immunomodulator. WO2020064818. 2020.
- 48.Hoffman TJ, Mondiere RJG, Godineau E, et al. Process for the preparation of microbiocidal oxadiazole derivatives. WO2020212513. 2020.
- 49.Gonzalez EC, Munoz AO, Solana SJ. Preparation of 3-(2-(heteroaryl)-pyridin-4-yl)-5-(trifluoromethyl)-1,2,4-oxadiazole derivatives as HDAC6 inhibitors. WO2020245381. 2020.
- 50.Shafeev M, Fitzgerald DJ, Schelshorn DW, et al. Preparation of oxazole and oxadiazole derivatives useful as agonists of free fatty acid receptor 1. WO2020211956. 2020.
- 51.Griffin AM, Botella GM, Marron BE, et al. KCNT1 inhibitors and methods of use. WO2021173930. 2021.
- 52.Chris D, Sarah H, Rajesh V. Method of modulating ribonucleotide reductase. WO2021081337. 2021.
- 53.Boss KD, Fan Y, Flyer AN, et al. Preparation of 1,2,4-oxadiazole derivatives as liver X receptor agonists. WO2021105857. 2021.
- 54.Bae EJ, Choi WI, Kim H, et al. Preparation of oxadiazole phenylalanine derivatives as tryptophan hydroxylase inhibitor. WO2022149925. 2022.
- 55.Zhao Y, Zhang Z, Wu G, et al. Preparation of oxadiazole derivatives as FXR agonists. CN114195776. 2022.
- 56.Mhaske PC, Walunj YS, Shinde AD, et al. Oxadiazole-quinoline pharmaceutical compounds. IN202321033845. 2023.
- 57.Griffin AM, Botella GM, Ricardo L. KCNT1 inhibitors comprising an isoxazole or oxadiazole core and methods of use. US2023019648. 2023.
- 58.Ahmed HMAL, Ali MMK, Ahmed AAA, et al. Preparation of 5-((2-isopropyl-5-methylphenoxy)methyl)-3-(naphthalen-2-yl)-1,2,4-oxadiazole as an anticancer and antimicrobial compound. US11952354. 2024.
- 59.Ahmed HMAL, Ali MMK, Ahmed AAA, et al. Preparation of 3-(3-chlorophenyl)-5-((2-isopropyl-5-methylphenoxy)methyl)-1,2,4-oxadiazole as an anticancer and antimicrobial compound. US11919871. 2024.
- 60.Garzya V, Tancock J, Sireau NT, et al. Oxadiazole compounds for use in the treatment of obsessive-compulsive disorder. WO2024028611. 2024.
- 61.Mattia M, Barbara V, Christian S, et al. 1,3,4-oxadiazole derivatives as selective histone deacetylase 6 inhibitors. EP2023069936. 2024.
- 62.Jeremy D, Anne-Sophie R, Jacopo N, et al. 3-heteroaryl-5-chlorodifluoromethyl-1,2,4-oxadiazole as a fungicide. 2024. EP2023076346.
- 63.Yuanyuan Q, Mayland C, Shahriar M. Method for inhibiting clostridioides difficile spore germination related applications. US2023076849. 2024.
- 64.Matic L, Samo L, Janez K. Small molecule-inhibitors of 6-phosphofructo-1-kinase for reducing lactate generation by cancer cells. WO2020030613. 2020.
- 65.Zhang Z, Jiang X, Rothe K, et al. Combination therapy for treating blood cancer. WO2020061216. 2020.
- 66.Sintim HO, Naclerio G. 1,3,4-oxadiazole derivatives and analogs as antibacterial agents and their preparation. WO2020219792. 2020.
- 67.Calhoun A, Chen X, Gardinier KM, et al. F, preparation of 5-oxa-2-azaspiro[3.4]octane derivatives as M4 receptor agonists for the treatment of cognitive, psychotic and other diseases. WO2021070091. 2021.
- 68.Yang R, Cheng W, Fan J, et al. Containing 1,3,4-oxadiazole ring for nootkatone thioether derivative, and its preparation method and application. CN113461630. 2021.
- 69.Cheng L, Yu W, Pang W. Preparation of the 1,3,4-oxadiazole derivative as neuraminidase inhibitors and its application in preparing drugs for inhibiting neuraminidase activity. CN113461631. 2021.
- 70.Marchini M, Vergani B, Sandrone G, et al. Preparation of 5-(trifluoromethyl)-1,3,4-oxadiazole derivatives as selective histone deacetylase 6 inhibitors. WO2022029041. 2022.
- 71.Zhang J, Walker M. Preparation of 2-(imidazo[1,2-a]1,8-naphthyridin-8-yl)-1,3,4-oxadiazole derivatives as enhancers of innate immune response for the treatment of viral infections. WO2023244672. 2023.
- 72.Venugopala KN, Deb PK, Pillay M, et al. Substituted 1,3,4-oxadiazole derivatives as antitubercular agents. US11998532. 2023.
- 73.Bae YS, Jeong DU, Lee J. Novel oxadiazole derivative compound and pharmaceutical composition containing the same for preventing or treating neuroinflammatory diseases. WO2023224371. 2023.
- 74.Gundla R, Oggu S, Malempati S, et al. A method for the preparation of 1,2,3-triazole incorporated 1,3,4-oxadiazole-triazine derivatives with anticancer activity. IN202341033384. 2023.
- 75.Florence FW, Tom R. Oxadiazole HDAC6 inhibitors and uses thereof. US2023017894. 2023.
- 76.Ju HL, Beom SL, Jiyeon K. Compositions for preventing or treating pulmonary arterial hypertension. IB2023050658. 2023.
- 77.Ju HL, Beom SL, Yeong SJ. Compositions for preventing or treating heart failure (HF). IB2023050659. 2023.
- 78.Ali MMK, Ahmed HMAL, Ahmed AAA, et al. Preparation of 2-(benzo[d]oxazol-2-yl)-N’-(2-(5-phenyl-1,3,4-oxadiazol-2-ylthio)acetoxy)acetimidamide as an antimicrobial compound. US11958843. 2024.
- 79.Lee JK, Min J, In J, et al. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitors, and application thereof. WO2024054071. 2024.
- 80.Lee JK, Min J, In J. 1,3,4-oxadiazole derivative compounds as histone deacetylase 6 inhibitors, and uses thereof. KR2023013453. 2024.
- 81.Lee JK, Min J, In J, et al. 1,3,4-oxadiazole triazole compounds as histone deacetylase 6 inhibitors, and a pharmaceutical composition comprising the same. IB2023057181. 2024.





































