Abstract
Pyrazole or 1H-pyrazole, a five-membered 1,2-diazole, is found in several approved drugs and some bioactive natural products. A myriad number of derivatives of this small molecule have been reported in clinical and preclinical studies for the potential treatment of several diseases. The number of drugs containing a pyrazole nucleus has increased significantly in the last 10 years. Some of the best-selling drugs in this class are ibrutinib, ruxolitinib, axitinib, niraparib and baricitinib, and are used to treat different types of cancers; lenacapavir to treat HIV; riociguat to treat pulmonary hypertension; and sildenafil to treat erectile dysfunction. Several aniline-derived pyrazole compounds have been reported as potent antibacterial agents with selective activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci. Here, we discuss the pyrazole-derived drugs reported up to September 2023.
Keywords: antibacterial, anticancer, indazole, kinase inhibitor, lenacapavir, MRSA, pyrazole, sildenafil, zavegepant
Graphical abstract
Pyrazole or 1H-pyrazole (1) is a five-membered 1,2-diazole (Figure 1). Substituted pyrazole and its derivatives are known for a wide spectrum of biological and therapeutic activities. Mono-, di- and tri-substituted pyrazole derivatives as well as their quaternary salts are integral parts of several drugs [1,2]. Pyrazole nucleus fused with a benzene ring such as 1H-indazole (indazole or 1,2-benzopyrazole, 2) is found in some natural products, several drugs and a large number of bioactive derivatives [3–5]. Similarly, 2H-indazole (3) and pyrazolo[1,5-a]pyridine (4) are an integral part of several drugs and numerous bioactive compounds such as antimalarials [6], antineoplastic [7], antibacterials [8] and so on [9–13].
Figure 1. . Structure of pyrazole (1) and its fused variants: 1H-indazole (2), 2H-indazole (3) and pyrazolo[1,5-a]pyridine (4).
Several pyrazole-derived drugs have been reported in recent decades. Surprisingly, most pyrazole-containing drugs have been reported after 2016, as can be seen in the timeline (Figure 2). Here we will discuss the pyrazole-derived drugs reported up to September 2023.
Figure 2. . Timeline of approval of pyrazole containing in the USA.
Pirtobrutinib (5) or Jaypirca® is one of the most recently approved drugs that received accelerated approval in January 2023. This tetra-substituted pyrazole derivative is a Bruton's tyrosine kinase inhibitor (TKI) approved to treat ibrutinib-resistant mantle cell lymphoma [14,15]. Zavegepant (6, Zavzpret™) is an indazole-substituted drug approved in 2023 to treat migraine. It is an antagonist of calcitonin gene-related peptide receptor (Figure 3) and used as a nasal spray [16]. This indazole derivative is the first intranasal third-generation gepant [17]. Lenacapavir (7, Sunlenca®) is a fused pyrazole-containing antiretroviral drug approved in 2022 to treat HIV/AIDS. It is the first capsid inhibitor – blocking the HIV-1 protein shell – that is approved to treat any disease to date. Lenacapavir (7) is being used to treat multidrug-resistant HIV-1 infection [18,19]. One of the unique properties of this polyfluorinated pyrazole-containing drug is its extra-long half-life, which needs a dosing interval of every 6 months [20]. Futibatinib (8, Lytgobi®), a kinase inhibitor, was approved in late 2022 to treat locally advanced and metastatic bile duct cancer (cholangiocarcinoma). This pyrimidine-fused pyrazole derivative (8) is being used to handle previously treated cholangiocarcinoma harboring FGF receptor 2-mutated or -rearranged genes [21,22]. Omidenepag isopropyl (9, Eybelis® and Omlonti®) has been approved to treat glaucoma and ocular hypertension. This drug was approved in Japan followed by the USA in 2018 and 2022, respectively. Omidenepag isopropyl (9) is a prodrug that is hydrolyzed to its active form, omidenepag.
Figure 3. . Pyrazole-derived drugs approved in recent years.
Chronic myelogenous leukemia arises from the fusion of BCR and ABL1 genes on the Philadelphia chromosome from a reciprocal translocation between chromosomes 9 and 12. Some adult acute lymphatic leukemia cases arise from the same mechanism. Asciminib (10) has been discovered as the first allosteric BCR-ABL1 inhibitor to reach the clinic and has been approved to treat Philadelphia-positive chronic myelogenous leukemia (Figure 3). This pyrazole-substituted pyridine derivative (10) is a kinase inhibitor [23,24]. Vericiguat (11, Verquvo®) is a fused pyrazole-derived drug used to treat patients with heart failure. This small-molecule drug is a soluble guanylate cyclase stimulator that was approved in 2021 by the US FDA [25]. Belumosudil (12, Rezurock®) is a 1,2-benzopyrazole (indazole) drug that has been approved to treat chronic graft-versus-host disease caused by the complications of allogeneic hematopoietic cell transplantation. This pyrazole indazole-pyrimidine hybrid drug (12) has a dual and unique mechanism of action: it targets ROCK2 in T helper follicular cells and inhibits tissue fibrosis by inhibiting the Rho-ROCK-MRTF pathway [26].
Umbralisib (13, Ukoniq®, TG Therapeutics, NC, USA) is pyrazole-fused pyrimidine derivative that was approved on 5 February 2020 to treat refractory or relapsed marginal zone lymphoma and follicular lymphoma. It inhibits several kinases including casein kinase CK1-ε and PI3K-δ [27,28]. Berotralstat (14, Orladeyo®) was approved in 2020 to treat hereditary angioedema. This trifluoromethyl-attached trisubstituted pyrazole derivative (14) is a potent plasma kallikrein inhibitor and highly selective over related serine proteases [29,30]. Pralsetinib (15, Gavreto®) is a potent and selective inhibitor of RET receptor tyrosine kinase, which has been approved to treat non-small cell lung carcinoma (NSCLC) and thyroid cancer [31–33]. This small molecule drug contains two pyrazole units with fluorine and methyl substitutions. Selpercatinib (16, Retevmo®) has been approved by the FDA in 2020 to treat three types of cancer: NSCLC, medullary thyroid cancer and other thyroid cancers with RET gene altercations. This pyrazolo–pyridine hybrid is the first drug approved to treat cancer with a RET gene mutation or fusion, which accounts for approximately 2% of all human cancers [34]. These two pyrazole-based RET inhibitors, pralsetinib (15) and selpercatinib (16), have opened up a new paradigm of RET precision oncology [34]. Avapritinib (17, Ayvakit™) is an N-methyl pyrazole-substituted drug used to treat advanced systemic mastocytosis including aggressive systemic mastocytosis, systemic mastocytosis with an associated hematological neoplasm and mast cell leukemia (MCL). This drug is an orally available, potent and selective inhibitor of KIT (a receptor tyrosine kinase proto-oncogene) and PDGF receptor-α [35,36]. Voxelotor (18, Oxbryta) is an N-isopropyl substituted pyrazole-containing small-molecule drug approved to treat sickle cell disease. It inhibits the polymerization of deoxygenated sickle hemoglobin, polymerization of sickle hemoglobin drives sickle cell disease [37,38]. Voxelotor (18) is one of the rare drugs with an aldehyde functional group. Zanubrutinib (19, Brukinsa®) is an anticancer drug built on the core structure of the pyrazole nucleus. It is an irreversible, potent and highly selective covalent inhibitor of Bruton's tyrosine kinase [39]. This orally available small molecule has been approved to treat different types of B-cell malignancies [40]. Zanubrutinib (19) is a pyrazole–pyrimidine hybrid molecule containing a N-acryl warhead for covalent binding. Elexacaftor (20) is a cystic fibrosis transmembrane conductance regulator modulator dipyrazole-containing drug approved to treat cystic fibrosis. It is used along with ivacaftor and tezacaftor as a combination therapy (Trikafta®) [41]. Enterectinib (21, Rozlytrek®) is an indazole-containing drug used to treat several types of cancer. It is an ATP-competitive TKI with activity against ALK, ROS1, TRKA, TRKB and TRKC [42].
Darolutamide (22, Nubeqa™) is a small-molecule anticancer drug containing two pyrazole units (Figure 4). This dipyrazole-containing drug is used to treat castration-resistant prostate cancer. It is a structurally unique nonsteroidal androgen receptor antagonist [43]. Erdafitinib (23, Balversa™) is a TKI specifically targeting FGF receptor to treat different types of cancers. This small molecule has been approved to treat metastatic urothelial (bladder) cancer [44] and it (23) contains an N-methyl pyrazole moiety. Baricitinib (24, Olumiant™) is a disubstituted pyrazole derivative that has been approved to treat several diseases such as alopecia, COVID-19 and rheumatoid arthritis and is an inhibitor of several subtypes of Janus kinases. This molecule is also effective against adult-onset Still's disease, an idiopathic systemic inflammatory disease of unknown etiology [45–47]. Encorafenib (25, Braftovi®) is a trisubstituted pyrazole derivative that acts as a BRAF inhibitor to target MAP signaling pathways. This small-molecule pyrazole derivative (25) was approved in 2018 to treat BRAF V600E- or V600K-mutated melanoma [48]. Larotrectinib (26, Vitrakvi®) is a fused pyrimidine-pyrazole derivative approved in 2016 to treat metastatic solid tumors and is an inhibitor of tropomyosin kinase receptors (Trk A, B and C) [49]. This is the first drug specifically approved for tumors based on their specific mutation rather than their origin [50]. Lorlatinib (27, Lorbrena®) is a pyrazole-substituted macrolactam derivative that is an orally available ALK and ROS1 inhibitor used to treat ALK-positive metastatic NSCLC [51]. Edaravone (28, Radicava®) is a pyrazolone derivative approved to treat stroke and amyotrophic lateral sclerosis that was approved in 2016. Interestingly, this small molecule has been used as a biochemical reagent for a long time to analyze monosaccharides in carbohydrate complex media [52]. Niraparib (29, Zejula®) is a 2H-indazole derivative showing potent inhibition of PARP inhibitors with selective activity against PARP1 and PARP2. The FDA approved this drug in 2017 to treat recurrent epithelial ovarian and fallopian tube cancer [53,54]. Telotristat ethyl (30, Xermelo®) is an ester prodrug approved to treat diarrhea caused by carcinoid syndrome (Figure 4); this disubstituted pyrazole derivative (30) is a tryptophan hydroxylase inhibitor [55,56].
Figure 4. . Recently approved pyrazole-based drugs.
Two pyrazolium-containing antibiotics, cefoselis (31) and ceftolozane (32), have been approved to treat bacterial infections (Figure 5). The zwitterion ion structure of these antibiotics helps to traverse the outer membrane of Gram-negative bacteria. Cefoselis (31) is a fourth-generation cephalosporin antibiotic used to treat respiratory and urinary tract infections [57]. Ceftolozane (32), a fifth-generation cephalosporin antibiotic containing a tetra-substituted pyrozolium moiety, is one of the most widely used antibacterial agents to treat drug-resistant bacterial infections including methicillin-resistant Staphylococcus aureus (MRSA), enterobacterales and Pseudomonas aeruginosa [58]. Ceftolozane (32), in combination with a bacterial β-lactamase inhibitor (tazobactam), is used to treat urinary tract infection, bacterial pneumonia and intra-abdominal infection. The pyrazolium nucleus is the key component of these antibiotics to prevent hydrolysis of the lactam moiety by AmpC β-lactamase [59]. Ibrutinib (33, Imbruvica®) is an irreversible inhibitor of BRTK approved in 2013 to treat mantle cell lymphoma. This pyrazole–pyrimidine hybrid molecule has been approved to treat mantle cell lymphoma, chronic lymphocytic leukemia and Waldenström's macroglobulinemia as a first-generation BRTK inhibitor [60]. Apixaban (34, Eliquis) is a fused pyrazole compound approved in 2012 in the USA as an anticoagulant medication to prevent blood clots; it is an inhibitor of factor Xa and an alternative to warfarin. This orally available drug was the 48th most prescribed medicine in the USA in 2020 [61,62]. Crizotinib (35, Xalkori®) is a disubstituted pyrazole derivative that was approved in 2011 to treat NSCLC and its mode of action is similar to that of lorlatinib (27) [63,64]. Ruxolotinib (36, Jakafi®) is a disubstituted pyrazole-derived drug approved in 2011 to treat myelofibrosis. This Janus kinase inhibitor has also been approved to treat other diseases such as intolerance of hydroxy urea and steroid-refractory acute graft-versus-host disease [65,66]. Ruxolitinib (36) is the first drug approved to treat alopecia and repigmentation of vitiligo [67].
Figure 5. . Pyrazole-derived compounds approved as drugs prior to 2017.
Eltrombopag (37, Promacta®) is a pyrazolone-derived hydrazone derivative used to treat thrombocytopenia. This unique thrombopoietin receptor agonist was approved in the USA in 2008 [68,69]. Celecoxib (38, Celebrex®) was approved in 1999 as a COX-2 inhibitor to treat several inflammatory diseases. This trisubstituted pyrazole derivative is one of the top 100 bestselling drugs in the USA [70,71]. Sildenafil (39, Viagra®) is a pyrimidine-fused pyrazole derivative used to treat erectile dysfunction and pulmonary heart disease. This blockbuster drug is a phosphodiesterase-5 blocker used to regulate blood in the penis. This orally available, serendipitously discovered drug was approved in 1998 [72]. Granisetron (40, Kytril® or Sancuso®) is a selective 5-HT3R antagonist and a longer acting drug used to treat chemotherapy-induced nausea and vomiting. This indazole drug (40) was approved in the 1990s and can be administered by mouth, intravenously or transdermally [73,74]. Benzydamine (41) is a selective prostaglandin synthetase inhibitor that was approved in the 1990s for prophylactic treatment of oral mucositis caused by radiation therapy for head and neck cancer [75]. This indazole drug also works as a central nervous system stimulant and is often used as recreational drug [76]. Bendazac (42) is an indazole-containing oxyacetic acid drug that inhibits the denaturation of proteins [77] and is used for the treatment of joint and muscular pain [78]. This fused pyrazole derivative has the therapeutic potential to treat cataracts [79]. Tepoxalin (43) is a nonsteroidal anti-inflammatory drug and a dual inhibitor of cyclooxygenase and lipoxygenase approved in 1998 to treat osteoarthritic pain in animals [80,81]. This trisubstituted pyrazole derivative (43) contains a hydroxamic acid functional group [82]. Fomepizole (44, Antizole®) is an alcohol dehydrogenase inhibitor that has been approved to treat methanol or ethylene glycol poisoning [83,84] and is one of the smallest of the small-molecule drugs. In addition to the above-approved drugs, there are several pyrazole drugs such as lonazolac (nonsteroidal anti-inflammatory drug), romonabant (antiobesity), metamizole (pain relief), fezolamine (antidepressant), betazole (diagnostic agent), pyrazofurin (a rare pyrazole-containing natural product with antibacterial, anticancer and antiviral properties) and sulfaphenazole (antibacterial sulfonamide) that are in different stages of clinical trials or withdrawn from clinical use due to severe side effects. Several recent reviews can be consulted describing these obsolete or under clinical trial drugs [6–8].
Pyrazole derivatives as antibacterial agents
In addition to the numerous approved drugs discussed earlier, a large number of pyrazole derivatives are at different stages of preclinical development to treat different types of diseases. Several review articles have been published recently explaining the importance of the pyrazole nucleus in drug discovery. These compounds have α-glucosidase [85], carbonic anhydrase inhibitor [86], antineoplastic [87] and anti-inflammatory [88] properties, as well as several other therapeutic uses [89–91].
In our quest to find azole derivatives to be used as therapeutic agents [92–95], we have reported the various classes of pyrazole derivatives that can act as potent antibacterial agents [96–104], also summarized in our recent mini-review article [2]. In continuation of our research to discover novel antibacterial agents, we have found 1-[4-(trifluoromethyl)phenyl]pyrazole derivatives (44) to be potent methicillin-resistant S. aureus (MRSA) and vancomycin-resistant enterococci (VRE) agents with minimum inhibitory concentration (MIC) values less than 1 μg/ml (Figure 6). These compounds are effective against the biofilms of S. aureus, and persisters; metabolically inactive variant of the homogenous population of bacteria [105]. The corresponding hydrazone derivatives (45) of this class of compounds showed activity against Acinetobacter baumannii, in addition to MRSA and VRE bacteria, with an MIC value as low as 1.56 μg/ml [106]. A series of trifluoromethyl phenyl-substituted derivatives of pyrazole (46) has been found as potent antibacterial agents with selective activity against Gram-positive bacteria. These compounds (e.g., 46) are effective in inhibiting and eradicating the biofilms of S. aureus and Enterococcus faecalis at two-times the MIC value [107]. Similarly, we have found 3,5-bis(trifluoromethyl)phenyl-substituted pyrazole derivatives (e.g., 47) to be potent growth inhibitors of drug-resistant bacteria [108]. A number of in vitro mechanistic assays such as membrane permeability, scanning electron microscopy and CRISPRi studies have led the determination of these compounds as cell membrane-disrupting agents, most likely by inhibiting fatty acid biosynthesis [109]. Several reviews have been reported on the synthesis and therapeutic properties of pyrazole derivatives [6,85,86,110–113].
Figure 6. . Pyrazole derivatives as potent antibacterial agents.
MIC: Minimum inhibitory concentration; MRSA: Methicillin-resistant Staphylococcus aureus; VRE: Vancomycin-resistant enterococci.
Conclusion
Pyrazole derivatives have been approved to treat different various diseases such as bacterial infections (cefoselis and ceftolozane), cancer (pirtobrutinib, futibatinib, asciminib, etc.), migraine (zavegepant), HIV (Lenacapavir), glaucoma (omidenepag), heart disease (vericiguat and umbralisib) and chronic graft-versus-host disease (belumosudil), as well as several other anomalies. Several pyrazole-derived drugs such as celecoxib and sildenafil have been among the most widely used medicines in the world. A myriad number of pyrazole derivatives are at different stages of preclinical and clinical development. Thus, based on these facts pyrazole is a privileged scaffold in drug discovery.
Future perspective
The pyrazole nucleus is the cornerstone of several approved drugs to treat different types of diseases. Recent approval of novel drugs has seen an uptick of this metabolically stable heterocycle. In many preclinical studies, it has been found that replacing other five-membered heterocycles with a pyrazole nucleus increases the antibacterial spectrum of the compounds. We can see more pyrazole derivatives among approved drugs and novel small compounds in the future. Metabolic stability of pyrazole derivatives is one of the important factors for its surge in newly approved drugs and novel compounds reported in literature.
Executive summary.
Introduction
Substituted pyrazole and fused-pyrazoles – 1H-indazole, 2H-indazole and pyrazolo[1,5-a]pyridine – are the common variants of this class of drugs.
Most the pyrazole-containing anticancer drugs are kinase inhibitors.
A myriad number of pyrazole derivatives are at different stages of clinical and preclinical trials.
Pyrazole-derived important drugs
Sildenafil is one of the blockbuster drugs that contains a fused-pyrazole moiety.
Zavegepant is one of the latest indazole-substituted drugs approved in 2023 to treat migraine.
Lenacapavir has been approved to treat multidrug-resistant HIV-1 infection. This unique drug has an extra-long half-life, which needs a dosing interval of 6 months.
Pralsetinib and selpercatinib have opened a new paradigm of RET precision oncology, which accounts for 2% of all cancers.
Baricitinib, a disubstituted pyrazole derivative, has been approved to treat several diseases such as alopecia, COVID-19, rheumatoid arthritis and Still's disease, an idiopathic systemic inflammatory disease of unknown etiology.
Larotrectinib is the first drug specifically approved to treat tumors based on their specific mutation rather than their origin.
Conclusion
The majority of pyrazole-containing drugs have been reported since 2016.
Pyrazole has emerged as an important heterocycle in drug discovery.
Supplementary Material
Footnotes
Supplementary data
To view the supplementary data that accompany this paper please visit the journal website at: www.future-science.com/doi/suppl/10.4155/fmc-2023-0207
Financial disclosure
This review was made possible by Arkansas INBRE Research Grant supported by a grant from the National Institute of General Medical Sciences (NIGMS), P20 GM103429 from the NIH. The author has no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.
Competing interests disclosure
The author has no competing interests or relevant affiliations with any organization or entity with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.
Writing disclosure
No writing assistance was utilized in the production of this manuscript.
References
Papers of special note have been highlighted as: • of interest; •• of considerable interest
- 1.Naim MJ, Alam O, Nawaz F, Alam MJ, Alam P. Current status of pyrazole and its biological activities. J. Pharm. Bioallied. Sci. 8(1), 2–17 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Alam MA. Antibacterial pyrazoles: tackling resistant bacteria. Future Med. Chem. 14(5), 343–362 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Review article describing antibacterial pyrazole derivatives.
- 3.Chevalier A, Ouahrouch A, Arnaud A, Gallavardin T, Franck X. An optimized procedure for direct access to 1H-indazole-3-carboxaldehyde derivatives by nitrosation of indoles. RSC Adv. 8(24), 13121–13128 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Mal S, Malik U, Mahapatra M, Mishra A, Pal D, Paidesetty SK. A review on synthetic strategy, molecular pharmacology of indazole derivatives, and their future perspective. Drug Develop. Res. 83(7), 1469–1504 (2022). [DOI] [PubMed] [Google Scholar]
- 5.Qin J, Cheng W, Duan Y-T, Yang H, Yao Y. Indazole as a privileged scaffold: the derivatives and their therapeutic applications. Anti-Cancer Agents Med. Chem. 21(7), 839–860 (2021). [DOI] [PubMed] [Google Scholar]
- 6.Ravindar L, Hasbullah SA, Rakesh KP, Hassan NI. Pyrazole and pyrazoline derivatives as antimalarial agents: a key review. Eur. J. Pharm. Sci. 183, 106365 (2023). [DOI] [PubMed] [Google Scholar]; • Latest review article describing antimalarial pyrazole derivatives.
- 7.Alam MJ, Alam O, Naim MJ et al. Recent advancement in drug design and discovery of pyrazole biomolecules as cancer and inflammation therapeutics. Molecules 27(24), 8708 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Verma R, Verma SK, Rakesh KP et al. Pyrazole-based analogs as potential antibacterial agents against methicillin-resistance Staphylococcus aureus (MRSA) and its SAR elucidation. Eur. J. Med. Chem. 212, 113134 (2021). [DOI] [PubMed] [Google Scholar]
- 9.Ghosh S, Pyne P, Ghosh A, Hajra A. Ortho C–H functionalizations of 2-Aryl-2H-indazoles. Chem. Record. 22(11), e202200158 (2022). [DOI] [PubMed] [Google Scholar]
- 10.An R, Guo M, Zang Y, Xu H, Hou Z, Guo C. Recent advances in synthesis of benzazoles via imines. Curr. Org. Chem. 24(17), 1897–1942 (2020). [Google Scholar]
- 11.Behbehani H, Ibrahim HM. Synthetic strategy for pyrazolo[1,5-a]pyridine and pyrido[1,2-b]indazole derivatives through AcOH and O2-promoted cross-dehydrogenative coupling reactions between 1,3-dicarbonyl compounds and N-amino-2-iminopyridines. ACS Omega 4(12), 15289–15303 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Lusardi M, Spallarossa A, Brullo C. Amino-pyrazoles in medicinal chemistry: a review. Int. J. Mol. Sci. 24(9), 7834 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Important review article describing the medicinal importance of pyrazole derivatives.
- 13.Ebenezer O, Shapi M, Tuszynski JA. A review of the recent development in the synthesis and biological evaluations of pyrazole derivatives. Biomedicines 10(5), 1124 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Aslan B, Kismali G, Iles LR et al. Pirtobrutinib inhibits wild-type and mutant Bruton's tyrosine kinase-mediated signaling in chronic lymphocytic leukemia. Blood Cancer J. 12(5), 80 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Pirtobrutinib (Jaypirca): A fourth Bruton's tyrosine kinase inhibitor for mantle cell lymphoma. The Medical Letter Med. Lett. Drug. Ther. 65(1670), e35–e36 (2023). [DOI] [PubMed] [Google Scholar]
- 16.Croop R, Madonia J, Stock DA et al. Zavegepant nasal spray for the acute treatment of migraine: a phase 2/3 double-blind, randomized, placebo-controlled, dose-ranging trial. Headache 62(9), 1153–1163 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Scuteri D, Tarsitano A, Tonin P, Bagetta G, Corasaniti MT. Focus on zavegepant: the first intranasal third-generation gepant. Pain Manage. 12(8), 879–885 (2022). [DOI] [PubMed] [Google Scholar]
- 18.Link JO, Rhee MS, Tse WC et al. Clinical targeting of HIV capsid protein with a long-acting small molecule. Nature 584(7822), 614–618 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Paik J. Lenacapavir: first approval. Drugs 82(14), 1499–1504 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Tuan J, Ogbuagu O. Lenacapavir: a twice-yearly treatment for adults with multidrug-resistant HIV infection and limited treatment options. Expert Rev. Anti-infect. Ther. 21(6), 565–570 (2023). [DOI] [PubMed] [Google Scholar]
- 21.De KS. Futibatinib: a potent and irreversible inhibitor of fibroblast growth factor receptors for treatment of the bile duct cancer. Curr. Med. Chem. 30, 1–7 (2023). [DOI] [PubMed] [Google Scholar]
- 22.Goyal L, Meric-Bernstam F, Hollebecque A et al. Futibatinib for FGFR2-rearranged intrahepatic cholangiocarcinoma. N. Engl. J. Med. 388(3), 228–239 (2023). [DOI] [PubMed] [Google Scholar]
- 23.Schoepfer J, Jahnke W, Berellini G et al. Discovery of asciminib (ABL001), an Allosteric inhibitor of the tyrosine kinase activity of BCR-ABL1. J. Med. Chem. 61(18), 8120–8135 (2018). [DOI] [PubMed] [Google Scholar]
- 24.De SK. Asciminib: first FDA approved allosteric inhibitor of BCR-ABL1 for the treatment of chronic myeloid leukemia. Med. Chem. Res. 32(3), 424–433 (2023). [Google Scholar]
- 25.Siddiqi AK, Greene SJ, Fudim M, Mentz RJ, Butler J, Khan MS. Vericiguat for the treatment of heart failure with reduced ejection fraction. Expert Rev. Cardiovasc. Ther. 21(4), 245–257 (2023). [DOI] [PubMed] [Google Scholar]
- 26.Salhotra A, Sandhu K, O'hearn J, Ali H, Nakamura R, Modi BG. A critical review of belumosudil in adult and pediatric patients with chronic graft-versus-host disease. Expert Rev. Clin. Immunol. 19(3), 241–251 (2023). [DOI] [PubMed] [Google Scholar]
- 27.US FDA. FDA grants accelerated approval to umbralisib for marginal zone lymphoma and follicular lymphoma. www.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-umbralisib-marginal-zone-lymphoma-and-follicular-lymphoma
- 28.Schweitzer J, Hoffman M, Graf SA. The evidence to date on umbralisib for the treatment of refractory marginal zone lymphoma and follicular lymphoma. Expert Opin. Pharmacother. 23(5), 535–541 (2022). [DOI] [PubMed] [Google Scholar]
- 29.Kotian PL, Wu M, Vadlakonda S et al. Berotralstat (BCX7353): structure-guided design of a potent, selective, and oral plasma kallikrein inhibitor to prevent attacks of hereditary angioedema (HAE). J. Med. Chem. 64(17), 12453–12468 (2021). [DOI] [PubMed] [Google Scholar]
- 30.Farkas H, Balla Z. A review of berotralstat for the treatment of hereditary angioedema. Expert Rev. Clin. Immunol. 19(2), 145–153 (2023). [DOI] [PubMed] [Google Scholar]
- 31.Subbiah V, Cassier PA, Siena S et al. Pan-cancer efficacy of pralsetinib in patients with RET fusion-positive solid tumors from the phase 1/2 ARROW trial. Nat. Med. 28(8), 1640–1645 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Gainor JF, Curigliano G, Kim DW et al. Pralsetinib for RET fusion-positive non-small-cell lung cancer (ARROW): a multi-cohort, open-label, phase 1/2 study. Lancet Oncol. 22(7), 959–969 (2021). [DOI] [PubMed] [Google Scholar]
- 33.Subbiah V, Hu MI, Wirth LJ et al. Pralsetinib for patients with advanced or metastatic RET-altered thyroid cancer (ARROW): a multi-cohort, open-label, registrational, phase 1/2 study. Lancet Diabetes Endocrinol. 9(8), 491–501 (2021). [DOI] [PubMed] [Google Scholar]
- 34.Addeo A, Miranda-Morales E, Den Hollander P et al. RET aberrant cancers and RET inhibitor therapies: current state-of-the-art and future perspectives. Pharmacol. Ther. 242, 108344 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Wu C-P, Lusvarghi S, Wang J-C et al. Avapritinib: a selective inhibitor of KIT and PDGFRα that reverses ABCB1 and ABCG2-mediated multidrug resistance in cancer cell lines. Mol. Pharmaceut. 16(7), 3040–3052 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Gotlib J, Reiter A, Deangelo DJ. Avapritinib for advanced systemic mastocytosis. Blood 140(15), 1667–1673 (2022). [DOI] [PubMed] [Google Scholar]
- 37.Vichinsky E, Hoppe CC, Ataga KI et al. A phase 3 randomized trial of voxelotor in sickle cell disease. N. Engl. J. Med. 381(6), 509–519 (2019). [DOI] [PubMed] [Google Scholar]
- 38.Konte K, Gaartman AE, Beuger BM et al. Voxelotor treatment leads to improved viability of sickle erythrocytes and altered neutrophil phenotype. Blood 140, 11114–11115 (2022). [Google Scholar]
- 39.Xu W, Yang S, Zhou K et al. Zanubrutinib in patients with relapsed/refractory chronic lymphocytic leukemia/small lymphocytic lymphoma: final results and correlative analysis of lymphocytosis. Leukemia Lymphoma 64(3), 712–716 (2023). [DOI] [PubMed] [Google Scholar]
- 40.Syed YY. Zanubrutinib: first approval. Drugs 80(1), 91–97 (2020). [DOI] [PubMed] [Google Scholar]
- 41.Ridley K, Condren M. Elexacaftor-tezacaftor-ivacaftor: the first triple-combination cystic fibrosis transmembrane conductance regulator modulating therapy. J. Pediatr. Pharmacol. Ther. 25(3), 192–197 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Pacenta HL, Macy ME. Entrectinib and other ALK/TRK inhibitors for the treatment of neuroblastoma. Drug Des. Devel. Ther. 12, 3549–3561 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Markham A, Duggan S. Darolutamide: first approval. Drugs 79(16), 1813–1818 (2019). [DOI] [PubMed] [Google Scholar]
- 44.Loriot Y, Necchi A, Park SH et al. Erdafitinib in locally advanced or metastatic urothelial carcinoma. N. Engl. J. Med. 381(4), 338–348 (2019). [DOI] [PubMed] [Google Scholar]
- 45.Sun Z, Li R, Wang Y, Han F, Wei W, Li X. Efficacy of baricitinib in patients with refractory adult-onset Still's disease. Drugs R&D 23(2), 109–120 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Sampath A, Banerjee A, Atal S, Jhaj R. Use of baricitinib in treatment of COVID-19: a systematic review. Med. Res. Rev. 43(5), 1322–1345 (2023). [DOI] [PubMed] [Google Scholar]
- 47.Obeime I, Larrondo J, Mcmichael AJ. Alopecia areata in skin of color patients: new considerations sparked by the approval of baricitinib. Cutis 111(1), 10–12 (2023). [DOI] [PubMed] [Google Scholar]
- 48.Koelblinger P, Thuerigen O, Dummer R. Development of encorafenib for BRAF-mutated advanced melanoma. Curr. Opin. Oncol. 30(2), 125–133 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Federman N, Mcdermott R. Larotrectinib, a highly selective tropomyosin receptor kinase (TRK) inhibitor for the treatment of TRK fusion cancer. Expert Rev. Clin. Pharmacol. 12(10), 931–939 (2019). [DOI] [PubMed] [Google Scholar]
- 50.Scott LJ. Larotrectinib: first global approval. Drugs 79(2), 201–206 (2019). [DOI] [PubMed] [Google Scholar]
- 51.Syed YY. Lorlatinib: first global approval. Drugs 79(1), 93–98 (2019). [DOI] [PubMed] [Google Scholar]
- 52.Bailly C, Hecquet P-E, Kouach M, Thuru X, Goossens J-F. Chemical reactivity and uses of 1-phenyl-3-methyl-5-pyrazolone (PMP), also known as edaravone. Bioorg. Med. Chem. 28(10), 115463 (2020). [DOI] [PubMed] [Google Scholar]
- 53.Van Andel L, Zhang Z, Lu S et al. Human mass balance study and metabolite profiling of 14C-niraparib, a novel poly(ADP-Ribose) polymerase (PARP)-1 and PARP-2 inhibitor, in patients with advanced cancer. Invest. New Drugs 35(6), 751–765 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lee A. Niraparib: a review in first-line maintenance therapy in advanced ovarian cancer. Target. Oncol. 16(6), 839–845 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Dillon JS, Kulke MH, Horsch D et al. Time to sustained improvement in bowel movement frequency with telotristat ethyl: analyses of phase iii studies in carcinoid syndrome. J. Gastrointest. Cancer 52(1), 212–221 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Kasi PM. Telotristat ethyl for the treatment of carcinoid syndrome diarrhea not controlled by somatostatin analogues. Drugs Today 54(7), 423–432 (2018). [DOI] [PubMed] [Google Scholar]
- 57.Liu YB, Lv XJ, Yu RJ et al. Multicenter, double-blind, randomized clinical trial of parenterally administered cefoselis versus cefepime for the treatment of acute bacterial infections. Eur. Rev. Med. Pharm. Sci. 18(14), 2006–2012 (2014). [PubMed] [Google Scholar]
- 58.Yusuf E, Bax HI, Verkaik NJ, Van Westreenen M. An update on eight “new” antibiotics against multidrug-resistant Gram-negative bacteria. J. Clin. Med. 10(5), 1068 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Murano K, Yamanaka T, Toda A et al. Structural requirements for the stability of novel cephalosporins to AmpC β-lactamase based on 3D-structure. Bioorg. Med. Chem. 16(5), 2261–2275 (2008). [DOI] [PubMed] [Google Scholar]
- 60.Ran F, Liu Y, Wang C et al. Review of the development of BTK inhibitors in overcoming the clinical limitations of ibrutinib. Eur. J. Med. Chem. 229, 114009 (2022). [DOI] [PubMed] [Google Scholar]
- 61.Frost C, Wang J, Nepal S et al. Apixaban, an oral, direct factor Xa inhibitor: single dose safety, pharmacokinetics, pharmacodynamics and food effect in healthy subjects. Br. J. Clin. Pharmacol. 75(2), 476–487 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Granger CB, Alexander JH, Mcmurray JJV et al. Apixaban versus warfarin in patients with atrial fibrillation. N. Engl. J. Med. 365(11), 981–992 (2011). [DOI] [PubMed] [Google Scholar]
- 63.Shaw AT, Bauer TM, De Marinis F et al. First-line lorlatinib or crizotinib in advanced ALK-positive lung cancer. N. Engl. J. Med. 383(21), 2018–2029 (2020). [DOI] [PubMed] [Google Scholar]
- 64.Shaw AT, Yeap BY, Solomon BJ et al. Effect of crizotinib on overall survival in patients with advanced non-small-cell lung cancer harbouring ALK gene rearrangement: a retrospective analysis. Lancet Oncol. 12(11), 1004–1012 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mesa RA, Yasothan U, Kirkpatrick P. Ruxolitinib. Nat. Rev. Drug Discov. 11(2), 103–104 (2012). [DOI] [PubMed] [Google Scholar]
- 66.Harrison C, Mesa R, Ross D et al. Practical management of patients with myelofibrosis receiving ruxolitinib. Expert Rev. Hematol. 6(5), 511–523 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Falto-Aizpurua L, Choudhary S, Tosti A. Emerging treatments in alopecia. Expert. Opin. Emerg. Drug. 19(4), 545–556 (2014). [DOI] [PubMed] [Google Scholar]
- 68.Serebruany VL, Eisert C, Sabaeva E, Makarov L. Eltrombopag (Promacta), a thrombopoietin receptor agonist for the treatment of thrombocytopenia: current and future considerations. Am. J. Ther. 17(1), 68–74 (2010). [DOI] [PubMed] [Google Scholar]
- 69.Panzer S, Pabinger I. Eltrombopag in chronic idiopathic thrombocytopenic purpura. Lancet 373(9664), 607–608 (2009). [DOI] [PubMed] [Google Scholar]
- 70.Mor A, Aizman E, Kloog Y. Celecoxib enhances the anti-inflammatory effects of farnesylthiosalicylic acid on T cells independent of prostaglandin E(2) production. Inflammation 35(5), 1706–1714 (2012). [DOI] [PubMed] [Google Scholar]
- 71.Clemett D, Goa KL. Celecoxib: a review of its use in osteoarthritis, rheumatoid arthritis and acute pain. Drugs 59(4), 957–980 (2000). [DOI] [PubMed] [Google Scholar]
- 72.Goldstein I, Burnett AL, Rosen RC, Park PW, Stecher VJ. The serendipitous story of sildenafil: an unexpected oral therapy for erectile dysfunction. Sexual Med. Rev. 7(1), 115–128 (2019). [DOI] [PubMed] [Google Scholar]
- 73.Neufeld SM, Newburn-Cook CV. The efficacy of 5-HT3 receptor antagonists for the prevention of postoperative nausea and vomiting after craniotomy: a meta-analysis. J. Neurosurg. Anesthes. 19(1), 10–17 (2007). [DOI] [PubMed] [Google Scholar]
- 74.Satiamurthy R, Yaakob SN, Shah MN, Azmi N, Omar SM. Potential roles of 5-HT receptor antagonists in reducing chemotherapy-induced peripheral neuropathy (CIPN). Curr. Mol. Med. 23(4), 341–349 (2023). [DOI] [PubMed] [Google Scholar]
- 75.Müller-Peddinghaus R. [New pharmacologic and biochemical findings on the mechanism of action of the non-steroidal antiphlogistic, benzydamine. A synopsis]. Arzneimittelforschung 37(5a), 635–645 (1987). [PubMed] [Google Scholar]
- 76.Ősz BE, Jîtcă G, Sălcudean A, Rusz CM, Vari CE. Benzydamine –an affordable over-the-counter drug with psychoactive properties –from chemical structure to possible pharmacological properties. Pharmaceuticals 16(4), 566 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Balfour JA, Clissold SP. Bendazac lysine. Drugs 39(4), 575–596 (1990). [DOI] [PubMed] [Google Scholar]
- 78.Gaikwad DD, Chapolikar AD, Devkate CG et al. Synthesis of indazole motifs and their medicinal importance: an overview. Eur. J. Med. Chem. 90, 707–731 (2015). [DOI] [PubMed] [Google Scholar]
- 79.Balfour JA, Clissold SP. Bendazac lysine. A review of its pharmacological properties and therapeutic potential in the management of cataracts. Drugs 39(4), 575–596 (1990). [DOI] [PubMed] [Google Scholar]
- 80.Charlton AN, Benito J, Simpson W, Freire M, Lascelles BD. Evaluation of the clinical use of tepoxalin and meloxicam in cats. J. Feline Med. Surg. 15(8), 678–690 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Sunaga T, Oh N, Hosoya K, Takagi S, Okumura M. Pro-apoptotic effects of tepoxalin, a cyclooxygenase/lipoxygenase dual inhibitor, on canine synovial fibroblasts. J. Vet. Med. Sci. 74(6), 745–750 (2012). [DOI] [PubMed] [Google Scholar]
- 82.Alam AM. Methods for hydroxamic acid synthesis. Curr. Org. Chem. 23(9), 978–993 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.US FDA. Search orphan drug designations and approvals. www.accessdata.fda.gov/scripts/opdlisting/oopd/detailedIndex.cfm?cfgridkey=33388
- 84.Mégarbane B. Treatment of patients with ethylene glycol or methanol poisoning: focus on fomepizole. Open Access Emerg. Med. 2, 67–75 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Firdaus JU, Siddiqui N, Alam O, Manaithiya A, Chandra K. Pyrazole scaffold-based derivatives: a glimpse of α-glucosidase inhibitory activity, SAR, and route of synthesis. Archiv. Pharmazie 356(5), 2200421 (2023). [DOI] [PubMed] [Google Scholar]
- 86.Dorbabu A. Pyrazole/pyrazoline as an excellent pharmacophore in the design of carbonic anhydrase inhibitors (2018–2022). Archiv. Pharmazie 356(4), 2200562 (2023). [DOI] [PubMed] [Google Scholar]
- 87.Ardevines S, Marqués-López E, Herrera PR. Heterocycles in breast cancer treatment: the use of pyrazole derivatives. Curr. Med. Chem. 30(10), 1145–1174 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]; • Pyrazole derivatives as potential treatment of different anomalies.
- 88.Priya D, Gopinath P, Dhivya LS et al. Structural insights into pyrazoles as agents against anti-inflammatory and related disorders. ChemistrySelect 7(5), e202104429 (2022). [Google Scholar]
- 89.Atukuri D. Pyrazolopyridine: an efficient pharmacophore in recent drug design and development. Chem. Biol. Drug Design 100(3), 376–388 (2022). [DOI] [PubMed] [Google Scholar]
- 90.Burnett CL, Bergfeld WF, Belsito DV et al. Safety assessment of 1-hydroxyethyl 4,5-diamino pyrazole sulfate as used in cosmetics. Int. J. Toxicol. 41(Suppl. 1), 69S–79S (2022). [DOI] [PubMed] [Google Scholar]
- 91.Sharma T, Singh J, Singh B, Kataria R, Kumar V. Methyl-linked pyrazoles: synthetic and medicinal perspective. Mini-Rev. Med. Chem. 22(5), 770–804 (2022). [DOI] [PubMed] [Google Scholar]
- 92.Ali MA, Okolo C, Alsharif ZA et al. Benign synthesis of thiazolo-androstenone derivatives as potent anticancer agents. Org. Lett. 20(18), 5927–5932 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Okolo C, Ali MA, Newman M et al. Hexafluoroisopropanol-mediated domino reaction for the synthesis of thiazolo-androstenones: potent anticancer agents. ACS Omega 3(12), 17991–18001 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chambers SA, Newman M, Frangie MM, Savenka AV, Basnakian AG, Alam MA. Antimelanoma activities of chimeric thiazole–androstenone derivatives. R. Soc. Open Sci. 8(8), 210395 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Alnufaie R, Ali MA, Alkhaibari IS, Roy S, Day VW, Alam MA. Benign synthesis of fused-thiazoles with enone-based natural products and drugs for lead discovery. New J. Chem. 45(13), 6001–6017 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Allison D, Delancey E, Ramey H et al. Synthesis and antimicrobial studies of novel derivatives of 4-(4-formyl-3-phenyl-1H-pyrazol-1-yl)benzoic acid as potent anti-Acinetobacter baumannii agents. Bioorg. Med. Chem. Lett. 27(3), 387–392 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Discusses rare pyrazole derivatives as anti-Acinetobacter baumannii agents.
- 97.Alnufaie R, Raj Kc H, Alsup N et al. Synthesis and antimicrobial studies of coumarin-substituted pyrazole derivatives as potent anti-Staphylococcus aureus agents. Molecules 25(12), 2758 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Zakeyah AA, Whitt J, Duke C et al. Synthesis and antimicrobial studies of hydrazone derivatives of 4-[3-(2,4-difluorophenyl)-4-formyl-1H-pyrazol-1-yl]benzoic acid and 4-[3-(3,4-difluorophenyl)-4-formyl-1H-pyrazol-1-yl]benzoic acid. Bioorg. Med. Chem. Lett. 28(17), 2914–2919 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Brider J, Rowe T, Gibler DJ et al. Synthesis and antimicrobial studies of azomethine and N-arylamine derivatives of 4-(4-formyl-3-phenyl-1H-pyrazol-1-yl)benzoic acid as potent anti-methicillin-resistant Staphylococcus aureus agents. Med. Chem. Res. 25(11), 2691–2697 (2016). [Google Scholar]
- 100.Whitt J, Duke C, Sumlin A et al. Synthesis of hydrazone derivatives of 4-[4-formyl-3-(2-oxochromen-3-yl)pyrazol-1-yl]benzoic acid as potent growth inhibitors of antibiotic-resistant Staphylococcus aureus and Acinetobacter baumannii. Molecules 24(11), 2051 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Whitt J, Duke C, Ali MA et al. Synthesis and antimicrobial studies of 4-[3-(3-fluorophenyl)-4-formyl-1h-pyrazol-1-yl]benzoic acid and 4-[3-(4-fluorophenyl)-4-formyl-1h-pyrazol-1-yl]benzoic acid as potent growth inhibitors of drug-resistant bacteria. ACS Omega 4(10), 14284–14293 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Hansa RKC, Khan MMK, Frangie MM et al. 4-4-(Anilinomethyl)-3-[4-(trifluoromethyl)phenyl]-1H-pyrazol-1-ylbenzoic acid derivatives as potent anti-Gram-positive bacterial agents. Eur. J. Med. Chem. 219, 113402 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Alnufaie R, Alsup N, Kc HR et al. Design and synthesis of 4-[4-formyl-3-(2-naphthyl)pyrazol-1-yl]benzoic acid derivatives as potent growth inhibitors of drug-resistant Staphylococcus aureus . J. Antibiot. 73(12), 818–827 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Delancey E, Allison D, Kc HR et al. Synthesis of 4,4′-(4-formyl-1h-pyrazole-1,3-diyl)dibenzoic acid derivatives as narrow spectrum antibiotics for the potential treatment of Acinetobacter baumannii infections. Antibiotics 9(10), 650 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Saleh I, Raj Kc H, Roy S et al. Design, synthesis, and antibacterial activity of N-(trifluoromethyl)phenyl substituted pyrazole derivatives. RSC Med. Chem. 12(10), 1690–1697 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Alkhaibari I, Kc HR, Angappulige DH, Gilmore D, Alam MA. Novel pyrazoles as potent growth inhibitors of staphylococci, enterococci and Acinetobacter baumannii bacteria. Future Med. Chem. 14(4), 233–244 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Raj Kc H, Gilmore DF, Alam MA. Development of 4-[4-(anilinomethyl)-3-phenyl-pyrazol-1-yl] benzoic acid derivatives as potent anti-staphylococci and anti-enterococci agents. Antibiotics 11(7), 939 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Alkhaibari IS, Kc HR, Roy S, Abu-Gazleh MK, Gilmore DF, Alam MA. Synthesis of 3,5-bis(trifluoromethyl)phenyl-substituted pyrazole derivatives as potent growth inhibitors of drug-resistant bacteria. Molecules 26(16), 5083 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Roy S, Kc HR, Roberts J et al. Development and antibacterial properties of 4-[4-(anilinomethyl)-3-phenylpyrazol-1-yl]benzoic acid derivatives as fatty acid biosynthesis inhibitors. J. Med. Chem. 66(19), 13622–13645 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]; •• Discusses the important mode of action of pyrazole derivatives as potent fatty acid biosynthesis inhibitors.
- 110.Xu Z, Zhuang Y, Chen Q. Current scenario of pyrazole hybrids with in vivo therapeutic potential against cancers. Eur. J. Med. Chem. 257, 115495 (2023). [DOI] [PubMed] [Google Scholar]
- 111.Zhang S, Ye Y, Zhang Q et al. Current development of pyrazole–azole hybrids with anticancer potential. Future Med. Chem. 15(16), 1527–1548 (2023). [DOI] [PubMed] [Google Scholar]
- 112.Sabnis RW. Fused pyrazole urea analogs as glucosylceramide synthase inhibitors for treating diseases. ACS Med. Chem. Lett. 14(8), 1031–1032 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Kaur P, Arora V. Pyrazole as an anti-microbial scaffold: a comprehensive review. Mini-Rev. Org. Chem. 20(6), 578–592 (2023). [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.