International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Quinazolinone and its derivatives are important heterocyclic compounds commonly found in various bioactive molecules (1). They represent a major class of biologically active compounds, with the quinazolinone nucleus attracting substantial attention due to its broad pharmacological activities (2). Studies have shown that quinazolinone and its derivatives possess diverse biological effects, including anti-HIV (3), anti-tumor (4), anti-bacterial (5), anti-inflammatory (6), anti-malarial (7), anti-convulsant (8), anti-diabetic (9), anti-oxidant (10), dihydrofolate reductase inhibition (11) and kinase inhibitory activity (12).
Heterocyclic compounds represent an important class of organic chemicals. These compounds contain one or more heteroatoms, such as nitrogen, oxygen or sulfur, along with carbon atoms, forming a ring structure. Their properties are largely influenced by the number and type of heteroatoms within the ring (13). The cyclic structure strongly affects the compound's characteristics, which are primarily determined by the heteroatoms present.
The term 'cyclic' refers to the presence of at least one ring, whereas 'hetero' indicates that non-carbon atoms, or heteroatoms, are part of that ring (14). In general, heterocyclic compounds resemble cyclic organic compounds composed only of carbon atoms. However, the inclusion of heteroatoms imparts distinct physical and chemical properties, differentiating them from their all-carbon counterparts (15). These compounds are highly versatile in organic chemistry and play a key role in medicinal chemistry. They are widely used in developing bioactive synthetic compounds, pharmaceutical agents and synthetic intermediates (16). In the pharmaceutical industry, heterocyclic structures are particularly common, with >60% of top-selling drugs containing at least one heterocyclic ring (17).
The basic structure of quinazolinone consists of a quinazoline ring and a ketone group. This bicyclic compound contains a pyrimidine ring fused with a benzene ring at the 4 and 8 positions (18). The quinazoline ring comprises six carbon atoms and two nitrogen atoms located at the 1 and 4 positions (19) (Fig. 1). As an oxidized form of quinazoline, quinazolinone retains the nitrogen atoms and is categorized as a nitrogen-containing heterocyclic compound. Quinazolinone forms the core structure of nearly 150 naturally occurring alkaloids found in various plant families, as well as in animals and microorganisms (20), including compounds such as glycyrrhizin, quinazolinone, deoxycannabinoid ketone and rutaecarpine (21). The first quinazoline alkaloid, vasicine, was isolated in 1888 from the Indian medicinal plant Adhatoda vasica and continues to demonstrate significant pharmacological value (22). In recent years, studies have shown that vasicine and its derivatives have potential anti-inflammatory, antioxidant, and anti-Alzheimer's disease effects (23). For instance, vasicine has been shown to alleviate atopic dermatitis induced by 2,4-dinitrochlorobenzene in BALB/c mice by inhibiting pro-Th 2 and Th 2 cytokines in both serum and skin tissues (24). Additionally, vasicine inhibits acetylcholinesterase (AChE) by specifically binding to the AChE catalytic site, such as Trp 84 and Ser 200, positioning it as a promising lead compound for Alzheimer's disease treatment (25). Furthermore, in a rat model of lung injury, in vitro assays of lung tissue homogenates revealed that vasicine inhibits lipid peroxidation triggered by excessive reactive oxygen species (ROS), while simultaneously restoring the activity of the endogenous antioxidant enzyme system (26). Thus, it is both a historical milestone in drug development and a starting point for research on new drugs. Later, vasicine and related quinazolinone alkaloids such as vasicinone and deoxyvasicinone were also identified in plants within the Acanthaceae family (27). Numerous additional natural products containing quinazoline or quinazolinone structures have since been isolated, analyzed and synthesized. The first quinazolinone compound was synthesized in the late 1860s by reacting anthranilic acid with cyanogen to produce 2-cyanoquinazolinone (28). This compound is a key precursor in quinazolinone chemistry, with its cyano group providing synthetic flexibility, making it a central molecule for constructing structurally diverse and complex quinazolinone derivatives. Systematic modification of its cyano group and the quinazolinone ring-particularly at the N-3 and C-6/C-7 positions-can efficiently generate a large library of structurally diverse compounds for high-throughput screening and structure-activity relationship (SAR) studies, supporting the discovery of new lead compounds and optimization of drug activity (29,30). It plays a pivotal role as a synthetic building block in drug discovery, particularly in the development of kinase inhibitors and related compounds. The unique structural features of quinazolinone, particularly its fused benzene and pyrimidine rings, increase the possibility and flexibility of structural modification (31). In addition, the carbonyl group at the 4-position is a strong electron-withdrawing group, markedly reducing the electron density of the quinazolinone ring and facilitating nucleophilic substitution reactions (32). A molecular docking study revealed that certain quinazolinone derivatives can form strong hydrogen bonds with LYS 630 and HIS 775 in topoisomerase II and stack π-π interactions with DT15, supporting the compound's ability to inhibit DNA replication and repair (33). Furthermore, the quinazolinone core adapts to the hydrophobic channel of cyclooxygenase-2, where the carbonyl group can form hydrogen bonds with Arg 121 and Tyr 356, thereby inhibiting enzyme activity (34). These properties enhance selectivity and biological activity during interactions with biomolecules, making quinazolinone a key scaffold in drug development (35).
Quinazolinones can be classified by structural features into six main groups: Those substituted at the 2- and/or 3-positions, simple 2-substituted quinazolin-4-ones and quinazolinones fused with pyrrole, pyrroloquinoline, piperidine or piperazine ring systems (36). They can also be categorized by the position of the keto or oxo group into 2(1H)-quinazolinones, 4(3H)-quinazolinones and 2,4(1H,3H)-quinazolinediones (37). Among these, 4(3H)-quinazolinones are the most common and often serve as intermediates or natural products in proposed biosynthetic pathways (38).
The pharmacological properties of quinazolinone-based compounds can be optimized by modifying their central structure with various functional groups. For instance, introducing alkyl (39), hydrazone (40,41) or other substituents at different positions on the ring can significantly influence target interactions and therapeutic efficacy. This structural adaptability has made quinazolinones and their derivatives a central focus in research on anti-tumor, anti-bacterial and anti-inflammatory drug development.
In synthetic and medicinal chemistry, the quinazoline scaffold has attracted considerable interest due to its simple synthesis, versatile reactivity and broad pharmacological potential (42). Several quinazolinone-derived drugs, such as Idelalisib and Canertinib, demonstrate the therapeutic potential of this class in treating hematological malignancies and other cancers. In addition, prazosin is used to treat hypertension, Albaconazole for fungal infections, Balaglitazone for diabetes and Dictyoquinazol A as both a neuroprotective agent and a glutamate receptor antagonist (43). Over the past two decades, >20 drugs containing a quinazoline or quinazolinone core structure have been approved by the Food and Drug Administration (FDA) for anti-tumor use. A prominent example is Dacomitinib, approved in 2018 for the treatment of non-small-cell lung carcinoma. These agents act through various mechanisms to inhibit cancer cell growth, mainly by targeting kinases, tubulin, kinesin spindle proteins and other related molecules (44).
Natural quinazolinone compounds, derived from plants, animals and microorganisms and widely used in traditional medicine, often have complex structures that have been refined through natural selection and evolutionary processes to produce unique biological activities (45). Quinazolinone alkaloids are present in the traditional Chinese medicinal herb Dichroae Radix, forming the core structure of febrifugine and isofebrifugine. These compounds have been used as antimalarial agents in China for >2,000 years (46). In recent years, increasing evidence has confirmed the significant anti-tumor activity of natural quinazolinone compounds, including their ability to overcome tumor resistance (47). Several derivatives based on the quinazolinone core nucleus, such as Bouchardatine, Luotonin F and Isofebrifugine, have been widely used in cancer treatment, underscoring the broad distribution and biological importance of this class of compounds in nature (43,48).
By contrast, the structural design of chemically synthesized quinazolinone-based drugs often involves modifications to the natural structure, with researchers selectively altering specific molecular sites (such as positions 2, 4, 6, 7 and 8) through chemical functionalization (49). These modifications can markedly influence pharmacological activity by introducing various functional groups, thereby optimizing drug efficacy, selectivity, pharmacological properties, toxicity, stability and pharmacokinetics. The choice between natural and synthetic quinazolinone drugs in therapy depends on their origins, pharmacodynamics and development strategies. Natural quinazolinones are better suited for chronic diseases that require multi-target synergy and low toxicity, such as asthma and chronic inflammation. By contrast, synthetic derivatives are more appropriate for conditions requiring targeted precision, adjustable structure and large-scale production, such as cancer therapy, anti-diabetic treatments and central nervous system disorders (50-52).
Although quinazolinones display a wide range of biological activities, this review focuses specifically on their anti-tumor properties, particularly the mechanisms through which they induce tumor cell death. It summarizes key quinazolinone derivatives known to induce cell death and aims to provide insights for designing structures that help elucidate the mechanisms involved in tumor cell death.
Apoptosis is a gene-regulated, self-controlled and orderly form of cell death that maintains internal homeostasis (53). It is regulated by multiple genes, many of which are highly conserved across species. Apoptosis enables multicellular organisms to remove damaged or unnecessary cells, playing a critical role in tissue remodeling during embryonic development and in maintaining tissue stability throughout life (54). There are two major apoptotic pathways: Intrinsic and extrinsic (55).
The intrinsic pathway is mainly triggered by internal signals such as cellular stress, DNA damage or oxidative stress. It involves the release of pro-apoptotic factors, including cytochrome c, from mitochondria, which activate caspases that execute cell death. Proteins of the Bcl-2 family are central regulators of this pathway, controlling mitochondrial membrane permeability (56). The extrinsic pathway is activated by external stimuli through membrane-bound death receptors, the most well-known being Fas and tumor necrosis factor (TNF) receptors. Ligand binding to these receptors activates a signaling cascade, including caspases, that leads to apoptosis (57).
Over the past decade, most quinazolinone derivatives have been shown to induce apoptosis through intrinsic pathways, including mitochondrial and endoplasmic reticulum (ER) stress pathways (Table I, Fig. 2). Regarding targeting the intrinsic pathway, Liang et al (58) synthesized four novel series of quinazolinone-based compounds by adding alkynyl functional groups at the C8 position. Among them, compound 9b showed the highest affinity for PI3Kγ and induced apoptosis in leukemic cells by simultaneously modulating the PI3K-AKT and MAPK signaling pathways. Similarly, Kim et al (59) modified substituents at the C-5/6 positions and optimized side-chain positioning within a hydrophobic binding region to develop a selective PI3Kδ inhibitor. This compound suppressed the PI3K pathway in SUDHL-5 and MOLT-4 cell models, reduced phosphorylation of AKT, S6 and eukaryotic translation initiation factor 4E-binding protein 1, and triggered apoptosis in malignant cells.
Wani et al (60) developed a new quinazolinonechalcone derivative, 3-(3-((E)-3-(4-hydroxy-3-methoxyphenyl)-2-propenoyl)phenyl)-2-methyl-3,4 dihydro-4-quinazolinone (QC). QC suppressed the mitochondrial anti-apoptotic protein Bcl-2. At the same time, it promoted the translocation of Bax from the cytoplasm to the mitochondria. Bax forms oligomers on the mitochondria, leading to an increase in mitochondrial membrane permeability. This results in the release of cytochrome C from the mitochondria into the cytoplasm. Cytochrome C binds to apoptotic protease activating factor 1 (Apaf-1) to form an apoptosome, which recruits and activates caspase-9. Activated caspase-9 then further activates downstream caspases, including caspase-3, caspase-6 and caspase-7, ultimately triggering cell apoptosis (60). Similarly, Madbouly et al (61) synthesized a related quinazolinone-chalcone compound [(E)-2-((4-acetylphenoxy) methyl)-3-phenylquinazolin-4(3H)-one], which induced apoptosis by promoting caspase-3 and poly(ADP-ribose) polymerase 1 (PARP-1) cleavage in A431 carcinoma cells. Some quinazolinone derivatives regulate apoptosis by modulating Bcl-2 family proteins. Xie et al (62) synthesized a series of MITC [4-((α-L-rhamnose oxy)benzyl)] quinazolinone derivatives. Among them, MITC-12 induced apoptosis in U251 cells by increasing caspase-3 expression and elevating the Bax/Bcl-2 ratio. Qiu et al (63) introduced alkoxy substituents and showed that compound 5k induced concentration-dependent apoptosis in HepG2 cells by increasing pro-apoptotic proteins Bad and Bax and decreasing anti-apoptotic proteins Bcl-2 and Bcl-xl. El-Shafey et al (64) demonstrated that novel compounds with a 2-thioquinazolinone scaffold triggered mitochondrial apoptosis by increasing ROS accumulation, elevating the Bax/Bcl-2 ratio and activating caspases 6, 7 and 9.
A smaller group of quinazolinone derivatives induce apoptosis via the extrinsic pathway. HMJ-38 [2-(3'-methoxy phenyl)-6-pyrrolidinyl-4-quinazolinone], a quinazolinone derivative, inhibits tubulin polymerization. Hour et al (65) showed that HMJ-38 induces both autophagy and apoptosis in gemcitabine-resistant pancreatic cancer cells, with the recruitment of pro-caspase-9 to the apoptosome by the Apaf-1 complex activating caspase-9 auto-catalytically, thereby enhancing apoptosis through subsequent activation of caspase-3, caspase-6 and caspase-7. Chiang et al (66) reported that HMJ-38 induces apoptosis in human umbilical vein endothelial cells through ROS generation and activation of the Fas/death receptor-mediated caspase-8 pathway, regulated by p53/ATM signaling. In WEHI-3 cells, MJ-29 activates key markers of ER stress by increasing the protein levels of calpain 1 and C/EBP homologous protein (CHOP), playing an important role in regulating cell apoptosis (67).
Ferroptosis is a regulated form of cell death first identified in cancer cells with oncogenic Ras mutations. It is characterized by iron-dependent lipid peroxidation (68,69). Unlike classical forms of cell death such as apoptosis and necrosis, ferroptosis involves intracellular iron accumulation and ROS generation. These processes lead to polyunsaturated fatty acid peroxidation in cell membranes, resulting in membrane damage and cell death (70). Ferroptosis is regulated by multiple metabolic pathways, including those involved in redox balance, iron metabolism, mitochondrial function, and amino acid, lipid and carbohydrate metabolism. Several disease-related signaling pathways also contribute to its regulation (71). The three primary pathways that control ferroptosis are the system Xc−/glutathione (GSH) peroxidase 4 (GPX4) axis, lipid metabolism and iron metabolism (72). In addition to small molecules and drugs, external stressors such as heat, cold, hypoxia and radiation can also induce ferroptosis (73) (Table II, Fig. 3).
Erastin, a piperazinyl-quinazolinone compound, is a well-known ferroptosis inducer (74). It inhibits the system Xc−, leading to reduced intracellular GSH levels and impaired GPX4 function. This results in increased lipid peroxidation, mitochondrial damage and other ferroptotic features (75). Erastin remains in the preclinical stage and is not yet marketed. Although it induces ferroptosis in various tumor cells, its therapeutic effect is limited by a feedback mechanism involving degradation of voltage-dependent anion channel (VDAC)2 and VDAC3. In melanoma cells, Erastin activates forkhead box M1, which transcriptionally upregulates NEDD4 E3 ubiquitin protein ligase, leading to VDAC2/3 ubiquitination and proteasomal degradation. This reduces intracellular ROS levels and modulates ferroptosis. Silencing Nedd4 restores VDAC2/3 levels and enhances cancer cell sensitivity to Erastin (76). Furthermore, Erastin induces mitochondrial dysfunction and ROS accumulation in HGC-27 gastric cancer and endometrial stromal cells, thereby promoting ferroptosis and suppressing malignancy (77,78). This process is also marked by iron accumulation and decreased ferroportin expression. Huang et al (79) found that Erastin induces both ferroptosis and apoptosis in A549 lung cancer cells by increasing ROS and activating p53. Erastin may also enhance the therapeutic effects of programmed cell death 1/programmed cell death ligand 1 inhibitors by influencing tumor-associated macrophage polarization (80). In another study, aspirin induced ferroptosis in HepG2 and Huh7 cells, and its effect was enhanced by Erastin co-treatment (81,82).
Organelle-targeted photosensitizers (PSs) are promising in enhancing photodynamic therapy, as they generate ROS under light exposure in the presence of oxygen. Zhao et al (83) developed a series of PSs based on the coumarin-quinazolinone (CQ) structure. The mitochondria-targeted CQ-Mito compound inhibits GPX4 upon light exposure, induces lipid peroxidation and activates Nrf2 while reducing Keap1 expression, which normally deubiquitinates Nrf2. Treatment with the ferroptosis inhibitor Fer-1 restores ferroptosis-related protein expression, confirming the light-induced ferroptotic effect of CQ-Mito (83). Additionally, BODIQPy-triphenylamine (BODIQPy-TPA), a lipophilic quinazolinone-based probe, can directly induce lipid peroxidation upon light exposure. In B16 and HepG2 cells, BODIQPy-TPA triggers ferroptosis by inhibiting GPX4, depleting GSH, and promoting cystine starvation, thereby activating the GPX4-GSH-cysteine axis (84). It also emits strong near-infrared fluorescence in live cells, making it useful for real-time imaging of the ferroptosis process.
3-(2,4-Dichloro-5-methoxyphenyl)-2,3-dihydro-2-thioxo-4(1H)-quinazolinone (Mdivi-1) is a quinazolinone derivative and a selective inhibitor of dynamin-related protein 1 (Drp1). In cold stress-induced liver injury, Mdivi-1 reduces malondialdehyde levels, increases GSH and GPX4 levels, inhibits mitochondrial fission and effectively mitigates ferroptosis (85). It also decreases ferrous ion and lipid peroxide levels. In models of PM2.5 exposure, Mdivi-1 significantly reduces the expression of acyl-CoA synthetase long chain family member 4, ferritin, ferritin heavy chain 1 and ferritin light chain 1, while increasing GPX4 protein levels, thereby inhibiting ferroptosis (86).
Ferroptosis suppressor protein 1 (FSP1), in conjunction with mitochondrial-derived ubiquinone, exogenous vitamin K and NAD(P)H/H+ as electron donors, has been identified as a second ferroptosis-suppressing system (87). A recent study indicated that 3-phenyl-quinazolinone, an FSP1 inhibitor, promotes FSP1 aggregation in tumors. This aggregation synergizes with ferroptosis inducers to enhance iron-dependent cell death and suppress tumor progression in vivo (88).
In the search for new therapies for uveal melanoma, coumarin-quinazolinone-phenylboronic acid has been developed as a prodrug to induce ferroptosis by targeting elevated ROS and tyrosinase levels in melanoma cells (89). QD394, a quinazolinone-dione compound, promotes lipid peroxidation and destabilizes GPX4 protein, thereby triggering GPX4-mediated ferroptosis (90).
Autophagy is a conserved cellular process that plays an essential role in maintaining homeostasis by degrading damaged or unnecessary organelles and proteins (91). It begins with the formation of autophagosomes, which enclose the targeted materials and deliver them to lysosomes for degradation. The term 'autophagy-dependent cell death (ADCD)' was introduced in 2018 by the Nomenclature Committee on Cell Death (92). ADCD is a regulated form of cell death characterized by the activation of autophagy markers, such as enhanced degradation of autophagosomal substrates and lipidation of microtubule-associated-proteinlight-chain-3 (LC3) (93). During autophagy, LC3 is converted from the cytoplasmic form LC3-I to the membrane-associated form LC3-II, and the LC3-II/LC3-I ratio is widely used as a marker of autophagy activity (94). Autophagy and apoptosis may act interchangeably under certain conditions. Excessive autophagy, referred to as 'autophagic death', can lead to cell death resembling apoptosis. These two processes are regulated by overlapping signaling pathways, including mTOR, Bcl-2 and Beclin-1 (95). Quinazolinone compounds modulate the initiation and progression of cell autophagy (Table III, Fig. 4).
Kumar et al (96) investigated a novel quinazolinone derivative, 2,3-dihydro-2-(quinoline-5-yl)quinazolin-4(1H)-one (DQQ). DQQ induced the formation of acidic vacuolar organelles (AVOs) in MOLT-4 cells and significantly increased the expression of autophagy-related proteins, including LC3-II, autophagy-related 7 (ATG7), ATG5 and Beclin-1 (96). Another study showed that a fused quinazolinone compound [compound 6, based on 7,8,9,10-tetrahydroazepino (2,1-b) quinazolin-12(6H)-one] increased AVO formation, as detected by acridine orange staining, indicating enhanced autophagic flux (97). Halofuginone (HF), a quinazolinone alkaloid derived from Dichroa febrifuga, promoted autophagy while inhibiting stathmin 1 and p53 expression and activity. HF increased LC3 expression, promoted ATG5-ATG12 complex formation and reduced sequestosome 1 expression in MCF-7 cells (98).
Certain quinazolinone derivatives can also inhibit autophagy. For instance, MJ-33 initiates autophagy at the vesicle nucleation stage by activating ATG proteins but decreases the LC3/LC3-II ratio and increases p62 levels, suggesting suppression of autophagic flux. In HT-29/5FUR cells, this suppression enhances MJ-33-induced apoptosis (99). ElZahabi et al (100) designed a series of quinazolin-4-one derivatives, among which compound 7 reduced autophagy, leading to increased apoptosis and cancer cell death.
Cellular senescence is generally an irreversible arrest of cell proliferation in damaged normal cells that have exited the cell cycle. It is associated with widespread macromolecular alterations and a secretory phenotype linked to chronic inflammation (101). Senescent cells show characteristic morphological changes, including flattened cell shape, vacuolization, cytoplasmic granularity and organelle abnormalities (102). Key features of senescence include persistent cell cycle arrest, altered transcriptional activity, a pro-inflammatory secretome, accumulated macromolecular damage and metabolic dysregulation (103). Increasing evidence indicates a complex relationship between aging and cancer (Table IV, Fig. 4). Initially, senescence functions as a protective mechanism by limiting abnormal cell proliferation and reducing cancer risk. However, with aging, immune decline and accumulated DNA damage may contribute to cancer development (104,105).
Table IVSenescence, necrosis and pyroptosis pathway changes following quinazolinone derivative treatment. |
Kamal et al (106) developed a series of 3-diarylacetylene quinazolinone compounds that activate p53, p21, p16, telomeric repeat binding factor 1 (TRF1) and protection of telomeres 1 (POT1) proteins. These compounds exhibited significant telomerase inhibitory activity, suggesting their potential as senescence inducers. Similarly, certain 3-(2-(hydroxymethyl) phenyl)-2-methylquinazolin-4(3H)-ones induced a senescence phenotype in HeLa cells (107). The application of quinazolinone and its derivatives in aging and cancer therapy remains an area of active investigation.
Necrosis is an abnormal form of cell or tissue death caused by external factors such as trauma, hypoxia, infection or injury. Receptor-interacting protein kinase 1 has been identified as a key regulator of necroptosis, a programmed form of necrosis activated by stimuli such as TNF, TNF superfamily member 10 (also known as TRAIL), lipopolysaccharide, oxidative stress or DNA damage (108). During necrosis, membrane breakdown leads to the release of cytoplasmic contents into the extracellular environment (109).
Pyroptosis is a regulated form of cell death mediated by gasdermins and characterized by continuous cell swelling, plasma membrane rupture and the release of intracellular contents that trigger inflammatory and immune responses (110). It is initiated by inflammasome activation in response to various triggers (111). Pyroptosis is involved in cancer, neurodegenerative diseases and ischemia-reperfusion injury. Research on the effects of quinazolinone derivatives in necrosis and pyroptosis is still at an early stage and the specific mechanisms remain elusive. The available literature is limited. Quinazolinone compounds can induce cell death through necrosis and pyroptosis (Table IV, Fig. 4).
In BALB/c mice, administration of two new quinazolinone compounds, 4(3H)-quinazolinone-2-propyl-2-phenylethyl and 4(3H)-quinazolinone-2-ethyl-2-phenylethyl, caused abnormal kidney function. Quinazolinones can affect sulfhydryl groups, disrupt protein structures, generate reactive metabolites and free radicals, and damage organelles such as tubular membranes, mitochondria and peroxisomes, ultimately leading to cell necrosis (112).
Mdivi-1, a quinazolinone-derived compound, has been shown to reduce necroptosis- and pyroptosis-related protein expression, including NLR family pyrin domain containing 3 (NLRP3) and gasdermin D N-terminus (GSDMD-NT), in cardiomyocytes of rats with myocardial infarction, thereby improving cardiac function in post-MI rats (113). Li et al (114) were the first to study the effect of Mdivi-1 on primary human keratinocytes in an in vitro model of atopic dermatitis-related inflammation induced by an inflammatory cocktail. Their results showed that Mdivi-1 inhibited NLRP3 inflammasome activation and pyroptosis, as indicated by reduced levels of NLRP3, apoptosis-associated speck-like protein containing a CARD, cleaved caspase-1, GSDMD-NT, and mature interleukins IL-1β and IL-18 in keratinocytes. In cellular and animal models of septic acute kidney injury, Mdivi-1 suppressed NLRP3-driven pyroptosis and improved mitochondrial function (115,116). Further research is needed to fully understand the roles and mechanisms of quinazolinone and its derivatives in necrosis and pyroptosis.
Quinazolinone and its derivatives have attracted considerable attention in recent years for their ability to induce cell death in various cancer types, representing a promising therapeutic approach. As single agents, these compounds act through multiple mechanisms, including apoptosis, autophagy and ferroptosis. To enhance therapeutic efficacy and address challenges such as drug resistance and toxicity, increasing efforts have focused on combining quinazolinone derivatives with other pharmacological agents. This strategy not only amplifies their anti-tumor effects but also enables more targeted interventions, potentially reducing adverse effects. The following section discusses the clinical prospectssss of quinazolinone and its derivatives in combination therapy, emphasizing their synergistic effects when used with conventional chemotherapeutic agents and molecular targeted therapies.
Studies have shown that co-treatment with cisplatin and Mdivi-1 synergistically enhances apoptotic cell death in both intrinsically and acquired cisplatin-resistant tumor cells, including MDA-MB-231 breast cancer, H1299 non-small cell lung cancer, glioblastoma, melanoma, cholangiocarcinoma and A2780cis cisplatin-resistant ovarian cancer cells (117,118). In A2780cis cells, Mdivi-1 enhances cisplatin sensitivity through a Drp1-independent pathway. The combination increases DNA damage, upregulates phorbol-12-myristate-13-acetate-induced protein 1 and disrupts mitochondrial function, ultimately triggering Bax- and Bak-independent mitochondrial outer membrane permeabilization and activating the mitochondrial apoptotic pathway (117). Tusskorn et al (118) demonstrated that Mdivi-1 increases the sensitivity of cholangiocarcinoma cells to cisplatin by promoting oxidative stress and autophagosome formation, thereby inducing cell death through mitochondrial pathways. Furthermore, Mdivi-1 protects against cisplatin-induced hair cell death in zebrafish by modulating mitochondrial dynamics, suggesting its potential for reducing ototoxicity. Further studies in mammalian models are required to clarify the underlying protective mechanisms (119).
MJ-33, a quinazolinone derivative, exhibits anti-tumor activity by reducing the viability of 5-FU-resistant HT-29 colon cancer cells. It induces apoptosis and autophagy through the AKT/mTOR signaling pathway, offering potential therapeutic value in 5-FU-resistant colorectal cancer (CRC) (76).
Lai et al (120) reported that 2-(3-fluorophenyl)-6-morpholinoquinazolin-4(3H)-one induces mitotic arrest, subsequently leading to apoptosis. This compound also synergizes with 5-FU to enhance cytotoxic effects against oral squamous cell carcinoma (OSCC).
HMJ-38 promotes both autophagy and apoptosis in MIA-RG100 pancreatic cancer cells and demonstrates cytotoxic effects against gemcitabine-resistant cells. The mechanism involves activation of the EGFR-AKT-mTOR signaling pathway, which induces autophagy, and the mitochondrial pathway, which facilitates apoptosis (65). These findings provide new perspectives for overcoming gemcitabine resistance in pancreatic cancer. Another quinazolinone derivative, QD232, suppresses the growth of gemcitabine-resistant MIA PaCa-2 cells by inhibiting STAT3 and Src phosphorylation, offering a potential strategy for treating drug-resistant pancreatic cancer associated with STAT3 activation (121).
Targeting the catalytic domain of PARP-1, a series of quinazolin-2,4(1H,3H)-dione derivatives have been synthesized as PARP-1 inhibitors. Zhou et al (122) developed novel compounds containing a 3-amino-pyrrolidine group, which enhanced the cytotoxic effects of temozolomide in the MX-1 xenograft breast cancer model. Similarly, 1-substituted benzyl quinazoline-2,4(1H,3H)-dione derivatives demonstrated comparable efficacy (123). Another study confirmed that the 2-propionyl-3H-quinazoline-4-one scaffold acts as a novel PARP-1 inhibitor, exhibiting synergistic effects with temozolomide in MX1 cells (124).
Ucleotide binding oligomerization domain containing 1 (NOD1) and NOD2 receptors, which contain nucleotide-binding oligomerization domains, are emerging as potential immune checkpoints. In a study by Ma et al (125), a quinazolinone derivative [6-(3-chlorophenyl)-3-(2-(3,3-difluoropiperidin-1-yl)-2-oxoethyl)-4-oxo-N-(3-(4-(trifluoromethyl)phenoxy)propyl)-3,4-dihydro quinazoline-7-carboxamide] was designed as a dual antagonist of NOD1/2. By inhibiting NOD1/2-mediated NF-κB and MAPK signaling pathways, this compound enhanced the response of B16 melanoma-bearing mice to paclitaxel therapy (125).
Idelalisib (Zydelig™; CAL-101; Gilead Sciences), with the chemical name 5-fluoro-3-phenyl-2-((1S)-1-(9H-purin-6-yl-amino)propyl)quinazolin-4(3H)-one, has the molecular formula C22H18FN7O and functions as a lipid kinase inhibitor, specifically targeting the p110δ isoform of class I phosphatidylinositol-3 kinase (PI3Kδ) (126). Idelalisib inhibits PI3Kδ by competitively binding to the ATP-binding site of the p110δ subunit. This inhibition disrupts key PI3Kδ signaling functions, including chemokine secretion, cell migration and receptor-driven kinase phosphorylation, ultimately reducing cell survival and promoting apoptosis (127,128).
Idelalisib has shown clinical efficacy in indolent B-cell non-Hodgkin lymphoma and was approved by the FDA in 2014 for monotherapy in patients with chronic lymphocytic leukemia (CLL), follicular lymphoma and small lymphocytic lymphoma. It is also approved in combination with rituximab for the treatment of CLL (129,130). In patients with previously treated indolent non-Hodgkin lymphoma, idelalisib as monotherapy achieved an objective response rate (ORR) of 57%, with a median duration of response (mDOR) of 12.5 months and a median progression-free survival (PFS) of 20.3 months (131,132). When combined with rituximab, idelalisib further improved overall response rates and 12-month overall survival (OS) in patients with CLL (133).
Despite its clinical benefits, idelalisib carries a black box warning from the FDA due to the risk of severe or fatal adverse events, including hepatotoxicity, diarrhea, colitis, pneumonia and intestinal perforation (134). Patients who developed colitis or transaminitis after idelalisib treatment exhibited elevated plasma chemokine levels and reduced T-regulatory cell (Treg) populations, suggesting that Treg depletion may contribute to the adverse effects of p110δ inhibition by releasing the suppression of cytotoxic T cells (135). In a Phase III clinical trial, the combination of idelalisib and rituximab improved PFS in patients with relapsed/refractory CLL from 11.1 to 20.8 months; however, long-term follow-up revealed a 5-year overall survival rate of only 40%, indicating disease progression in certain patients and suggesting the development of resistance (136,137). In CLL, resistance to idelalisib is closely associated with the activation of insulin-like growth factor 1 receptor (IGF1R). Specifically, the upregulation of IGF1R enhances the MAPK signaling pathway, bypassing PI3Kδ inhibition and thereby promoting tumor cell survival (138). To overcome Idelalisib resistance, researchers have explored various combination therapy strategies. For instance, idelalisib combined with bortezomib blocks drug resistance properties of Epstein-Barr virus-related B-cell origin cancer cells via regulation of NF-κB (139). Nevertheless, further investigation into the molecular mechanisms of resistance is necessary to develop more precise therapeutic strategies.
Ispinesib, a quinazolinone-derived compound, is a selective inhibitor of kinesin spindle protein (KSP) and functions as an allosteric modulator of KSP motor ATPase activity. By inhibiting KSP, ispinesib disrupts mitotic spindle formation, leading to cell cycle arrest and apoptosis (140). Developed by Cytokinetics, ispinesib entered clinical trials in 2004 for multiple indications, becoming the first potent and selective KSP inhibitor to undergo clinical testing in human diseases (20). According to data from the National Institutes of Health, ispinesib has been evaluated as monotherapy in 13 Phase I/II clinical trials for various cancer types, including relapsed renal cell cancer (RCC), breast cancer, recurrent or metastatic head and neck squamous cell carcinoma (HNSCC), ovarian cancer (OC), hepatocellular carcinoma (HCC) and CRC, with the best responses observed in breast cancer and OC (141,142). As a combination therapy, ispinesib has been tested in three clinical trials: With docetaxel for HCC, with capecitabine for HNSCC and with carboplatin for metastatic or recurrent malignant melanoma (142,143).
In clinical studies of breast cancer, Ispinesib has shown some antitumor activity, with an ORR of 9% (144). However, its ORR was relatively poor in other tumor types. In a phase II clinical trial for advanced RCC, no complete responses or partial responses were observed, and only 6 patients had stable disease (145). A phase II trial of Ispinesib was conducted in patients with advanced HCC who had not undergone chemotherapy. At the 8-week evaluation, 46% of patients had stable disease as the best response, with an mDOR of 3.9 months and a median time to tumor progression of 1.61 months (146). No significant objective response was achieved. The most common adverse reactions associated with ispinesib treatment include neutropenia, anemia, elevated alanine aminotransferase and aspartate aminotransferase, and diarrhea, while no neuropathy, mucositis or alopecia have been observed (147).
In glioblastoma, resistance to ispinesib is closely associated with the activation of STAT3. STAT3 mediates anti-apoptotic and metabolic effects through dual phosphorylation by Src and EGFR, thereby promoting drug resistance that can be reversed by simultaneously inhibiting Src and EGFR (148). In addition, drug efflux mediated by P-glycoprotein (P-gp) can pump ispinesib out of the cells, reducing its intracellular concentration and leading to drug resistance, which can be overcome by inhibiting P-gp activity to enhance the cytotoxicity of ispinesib (149). Clinical trials have reported a favorable safety profile, with no significant neurotoxicity, alopecia or gastrointestinal toxicity. The most common adverse event was reversible neutropenia (150). Ispinesib is currently in Phase I/II clinical development and further studies on its mechanisms and optimized clinical protocols may establish it as an effective anti-tumor therapy.
Nolatrexed [2-amino-6-methyl-5-(4-pyridylthio)-4(3H)-quinazolinone] is a thymidylate synthase inhibitor primarily developed for the treatment of HCC. In patients with unresectable HCC, nolatrexed achieved a median overall survival of 22.3 weeks, with an ORR of 1.4% and a median PFS of 12 weeks (151). Initially developed by Agouron Pharmaceuticals in the US, nolatrexed advanced to Phase II/III trials for liver cancer and head and neck tumors through a partnership with Roche. In 1999, Agouron transferred global oncology rights for nolatrexed to Eximias Pharmaceutical Company, which currently leads global development of nolatrexed. Ongoing Phase II clinical trials are being conducted for CRC, HNSCC, prostate cancer (PCA), pancreatic cancer and lung cancer in countries including the US, UK, Canada, Italy and South Africa. Phase III trials for liver cancer are also in progress (152,153).
Nolatrexed, a 4(3H)-quinazolinone derivative, has shown promising efficacy in clinical trials, exhibiting typical antimetabolite-related side effects such as short duration of action and low toxicity, along with mucositis, vomiting, diarrhea and thrombocytopenia (151). Thymidylate synthase (TS) is the primary target of nolatrexed and its overexpression is one of the key mechanisms of resistance. In CRC cells with p53 mutations, both TS mRNA and protein expression levels are significantly elevated, thereby increasing the cells' resistance to nolatrexed (154). Furthermore, being lipophilic, nolatrexed passively diffuses into cells without the need for specific membrane transport proteins. Since inhibition of transporter-mediated uptake is a mechanism of tumor cell resistance, this property may allow nolatrexed to be effective against drug-resistant tumors and reduce the likelihood of resistance development (155). Although granted orphan drug status by the European Medicines Agency, it was not approved by the FDA in 2005 (20).
Halofuginone is a synthetic derivative of a quinazolinone alkaloid with the chemical name 7-bromo-6-chloro-3-(3-(3-hydroxy-2-piperidinyl)-2-oxopropyl)-4(3H)-quinazolinone (156). It was developed by Collgard Biopharmaceuticals and received FDA approval in 2000 as an orphan drug for the treatment of scleroderma (20). Halofuginone inhibits epithelial-to-mesenchymal transition in tumor cells, reducing migration and invasiveness. It also selectively suppresses Th17 cell differentiation and inhibits Smad3 phosphorylation, a key downstream mediator of the TGF-β pathway. These effects reduce tumor-associated fibrosis and enhance immune surveillance (157,158). As a broad-spectrum anti-tumor agent, it has demonstrated efficacy against Pancreatic adenocarcinoma, CRC, breast cancer and lung cancer (159). Halofuginone has demonstrated promising results in a Phase II study for recurrent superficial transitional cell carcinoma of the bladder. In addition, a sustained-release formulation of fluorofebrin is under Phase II evaluation for safety, tolerability and pharmacokinetics in patients with Duchenne muscular dystrophy (20).
Up to date, clinical investigations of halofuginone have primarily focused on exploring its safety and tolerability rather than establishing overall therapeutic efficacy, and no specific data on ORR, PFS or DOR have yet been reported. Nonetheless, its antitumor potential has been demonstrated in several experimental models. For instance, in anaplastic thyroid carcinoma (ATC), halofuginone significantly suppresses ATC cell proliferation and tumor growth by inhibiting the enzyme-prolyl-tRNA synthetase-activating transcription factor 4-collagen type I signaling axis (160). In OSCC, halofuginone suppressed collagen synthesis and myofibroblast activation, thereby attenuating tumor invasiveness and growth (161). Common adverse effects include nausea, vomiting, a possible increased risk of bleeding, and decreased red and white blood cell counts (162). A study suggested that halofuginone can overcome drug resistance in cancer therapy with a relatively low risk of inducing resistance. For instance, it restores sensitivity to EGFR-tyrosine kinase inhibitors by targeting phosphoserine aminotransferase 1 downregulation (163), reverses paclitaxel resistance in basal-like breast cancer by modulating the BRCA1/TGF-β signaling axis (164) and serves as a potential therapeutic agent for 5-FU-resistant CRC by targeting microRNA-132-3p in vitro (165).
Febrifugine is the active antimalarial compound isolated from the traditional Chinese herb Chang Shan, which has been used for centuries to treat malaria-induced fevers (166). Febrifugine also demonstrates anti-tumor activity and has shown potential therapeutic effects of PCA and bladder cancer (BLCA), likely through inhibition of focal adhesion kinase (167-169). It also suppresses steroidogenesis and promotes apoptosis, contributing to its anti-tumor effects by inhibiting DNA synthesis (170). The effectiveness of febrifugine in clinical treatment is currently focused on basic research and animal models, with no specific data reported directly on clinical patients. A related study revealed its anticancer efficacy. In the BLCA model, febrifugine effectively inhibited the proliferation of T24 and SW780 BLCA cells, with IC50 values of 0.02 and 0.018 μM, respectively, and demonstrated good anticancer effects by reducing steroidogenesis and promoting apoptosis (168). It was also shown to enhance the anticancer effects of cisplatin in vivo (171). Due to significant side effects, there has been no further research on febrifugine for a long period of time and there is limited literature or clinical data that discuss its resistance in different cancer types in detail. Some potential mechanisms of resistance may include cancer cells rejecting febrifugine, mutations in target genes or cancer cells altering their sensitivity to the drug through metabolic pathways. The specific mechanisms of resistance may be gradually revealed in future studies. Common adverse effects include dizziness, dry mouth and persistent vomiting (172,173). A summary of the aforementioned quinazolinone derivatives is presented in Table V.
SAR studies of quinazolinone derivatives reveal a close association between molecular structure and biological activity. Structural optimization and the introduction of various substituents can effectively modulate anti-tumor activity, pharmacokinetic stability and targeting specificity. The substitution sites of quinazolinone are shown in Fig. 1.
The 2- and 3-positions are key sites for SAR studies, as they directly influence selectivity, potency and cellular activity. In anticancer compounds, substituents at positions 2 and 3 are predominantly sulfur ethers and aryl ketones, as seen in the investigational drug ispinesib. Electron-withdrawing groups at the meta position, such as halogens and trifluoromethyl, generally enhance inhibitory activity against EGFR, as exemplified by the quinazoline-based drugs gefitinib and erlotinib, which contain −Cl and −CF3 groups (174).
Substituents at the 6- and 7-positions typically target the solvent-exposed regions or entry channels of the ATP-binding pocket (175). Modifications at these positions significantly affect solubility, membrane permeability and pharmacokinetics. Halogen atoms are frequently introduced at these positions to increase lipophilicity and stability, thereby improving membrane penetration and bioactivity, as observed with -Cl and -Br in halofuginone (176). However, methoxy groups generally have the opposite effect, reducing the reactivity of the ring toward various reactions (50). The 6,7-dimethoxyquinazolinone derivative forms an additional hydrogen bond with Thr918 of VEGFR2 via the oxygen atom of the methoxy group. A molecular docking study showed that it can strongly bind to the hydrophobic site of VEGFR2 kinase, making it a potent VEGFR2 inhibitor (174). Furthermore, Kurogi et al (177) found from structure-activity relationship studies that introducing methoxy groups at the 6- and 7-positions of quinazolinone enhanced its hypolipidemic activity. Therefore, the addition or removal of such groups in vivo can serve as a tool for modulating the toxicity of quinazoline derivatives.
Substitution at the 5-position is relatively uncommon, as introducing substituents at this site may increase steric hindrance. However, it shows specificity for cytotoxic agents, such as the protein kinase inhibitor idelalisib and the dihydrofolate reductase inhibitor nolatrexed (20). These positions can also serve as linkers for synthesizing dual-target inhibitors. In the study on RAF kinase inhibitors, Huestis et al (178) designed the 5-fluoro-substituted quinazolinone compound GNE-0749. By masking the adjacent polar NH group, they enhanced the molecule's interaction with RAF kinase and significantly improved its solubility and permeability, thereby endowing it with properties suitable for oral administration (178). Table VI summarizes the SAR of several common quinazolinone derivatives. Fig. 5 shows the specific structure of these substituents.
This review examined the structure, applications and mechanisms of cell death induced by quinazolinone and its derivatives. Quinazolinone is a multifunctional heterocyclic compound that has attracted considerable attention for its promising biological activity (179). Quinazolinone derivatives act through multiple cell death pathways, including classical apoptosis, autophagy and ferroptosis associated with metabolic stress and damage, as well as inflammatory responses triggered by senescence, necrosis and pyroptosis. Importantly, quinazolinone and its derivatives offer distinct advantages in overcoming tumor resistance and immune evasion. Cancer cells often develop resistance to chemotherapy through mechanisms such as drug efflux and altered drug metabolism (180,181). Quinazolinone derivatives have demonstrated the ability to counteract some of these mechanisms by sensitizing cancer cells to chemotherapeutic agents. Several quinazolinone-based compounds have received FDA approval for cancer treatment, while others have shown encouraging efficacy in preclinical studies and in Phase I and II clinical trials (182). Despite these advances, further rigorous clinical studies are needed to fully evaluate the safety, efficacy and optimal dosing strategies of quinazolinone derivatives.
Although quinazolinone and its derivatives show considerable potential in cancer therapy, several challenges and unresolved issues remain. First, the chemical synthesis of quinazolinones still relies heavily on high temperatures and harsh reaction conditions, and generates numerous by-products (183). A team from South China Normal University developed a covalent organic framework, TAPP-Cu-An, which enables efficient dehydrogenative cross-coupling reactions under mild conditions (room temperature and light exposure), facilitating the photochemical synthesis of 4-quinazolinone (184). Furthermore, a team from Guilin University of Technology developed a 4-DPAIPN catalyst, using acetonitrile as the solvent and blue LED light irradiation, achieving a metal-free synthesis of tetracyclic quinazolinones and avoiding the use of precious metals (185). Second, although it is established that these compounds act through multiple cell death pathways, the interactions and regulatory mechanisms among these pathways are not yet fully understood. Future research should investigate the crosstalk between these pathways and determine how quinazolinone modulates them in different cancer types to optimize its anti-tumor effects. Most current studies have examined combinations of quinazolinone derivatives with chemotherapy agents; however, their synergistic potential with immunotherapy also warrants exploration (4,186). Finally, most quinazolinone derivatives suffer from poor water solubility, low oral bioavailability and short half-life, which limit their therapeutic efficacy in vivo (187). Future research should consider the use of nanocarriers (such as nanoparticles, liposomes and polymeric microparticles) to encapsulate the drug, improve solubility in aqueous solutions and enable targeted delivery, thereby maximizing therapeutic potential (188). Additionally, prodrug strategies can be employed, incorporating solubilizing moieties such as phosphate esters or polyethylene glycol (PEG) groups, such as PEG-400, to improve bioavailability (189).
Quinazolinone molecules can interact with fluorescent dyes through covalent coupling, electrostatic adsorption, or coordination bonding. These interactions allow real-time tracking of drug delivery in vivo, enable observation of drug absorption, transport and dynamic changes at the cellular level, and can be applied in fluorescent imaging of tumors (190). With advances in synthetic chemistry and drug design, structural modifications of quinazolinone compounds could further enhance specificity, selectivity and therapeutic efficacy. Designing new quinazolinone derivatives and investigating their roles in cancer treatment will be critical for advancing this field. By optimizing molecular structures to improve selectivity and cytotoxicity toward cancer cells, significant progress in therapeutic outcomes may be achieved.
Not applicable.
JL and YuY wrote the manuscript. YuY conceived and supervised the study. XK and YaY revised the manuscript. LW and QL finalized the figures and checked the grammar of the text. All authors contributed to the article and approved the submitted version. Data authentication is not applicable. All authors have read and agreed to the submitted version of the manuscript.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
Not applicable.
This work was supported by grants from the Natural Science Foundation of China (grant no. 82374273), the Natural Science Foundation of Henan (grant no. 252300420155), the Key Scientific Research Project of Higher Education of Henan Province (grant no. 25CY031), the Innovation Project of Graduate Students at Xinxiang Medical University (grant no. YJSCX202417Y) and the National College Students' Innovation and Entrepreneurship Training Program (grant nos. 202310472034 and 202410472002).
|
Tiwary BK, Pradhan K, Nanda AK and Chakraborty R: Implication of quinazoline-4(3H)-ones in medicinal chemistry: A brief review. J Chem Biol Ther. 1:1042016. | |
|
Rakesh KP, Manukumar HM and Gowda DC: Schiff's bases of quinazolinone derivatives: Synthesis and SAR studies of a novel series of potential anti-inflammatory and antioxidants. Bioorg Med Chem Lett. 25:1072–1077. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Sulthana MT, Chitra K, Alagarsamy V, Saravanan G and Solomon VR: Anti-HIV and antibacterial activities of novel 2-(3-Substituted-4-oxo-3, 4-dihydroquinazolin-2-yl)-2, 3-dihydrophthalazine-1,4-diones. Russ J Bioorg Chem. 47:112–121. 2021. View Article : Google Scholar | |
|
Salfi R, Hakim F, Bhikshapathi D and Khan A: Anticancer evaluation of novel quinazolinone acetamides: Synthesis and characterization. Anticancer Agents Med Chem. 22:926–932. 2022. View Article : Google Scholar | |
|
Gatadi S, Lakshmi TV and Nanduri S: 4(3H)-Quinazolinone derivatives: Promising antibacterial drug leads. Eur J Med Chem. 170:157–172. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Zayed MF: Medicinal Chemistry of quinazolines as analgesic and anti-inflammatory agents. ChemEngineering. 6:942022. View Article : Google Scholar | |
|
Mhetre UV, Haval NB, Bondle GM, Rathod SS, Choudhari PB, Kumari J, Sriram D and Haval KP: Design, synthesis and molecular docking study of novel triazole-quinazolinone hybrids as antimalarial and antitubercular agents. Bioorg Med Chem Lett. 108:1298002024. View Article : Google Scholar : PubMed/NCBI | |
|
Yaduwanshi PS, Singh S, Sahapuriya P, Dubey P, Thakur J and Yadav S: Synthesis of some noval qunazolinone derivatives for their anticonvulsant activity. Orient J Chem. 40:369–373. 2024. View Article : Google Scholar | |
|
Khalifa MM, Sakr HM, Ibrahim A, Mansour AM and Ayyad RR: Design and synthesis of new benzylidene-quinazolinone hybrids as potential anti-diabetic agents: In vitro α-glucosidase inhibition, and docking studies. J Mol Struct. 1250:1317682022. View Article : Google Scholar | |
|
Soliman AM, Karam HM, Mekkawy MH and Ghorab MM: Antioxidant activity of novel quinazolinones bearing sulfonamide: Potential radiomodulatory effects on liver tissues via NF-κB/PON1 pathway. Eur J Med Chem. 197:1123332020. View Article : Google Scholar | |
|
Osman EO, Emam SH, Sonousi A, Kandil MM, Abdou AM and Hassan RA: Design, synthesis, anticancer, and antibacterial evaluation of some quinazolinone-based derivatives as DHFR inhibitors. Drug Dev Res. 84:888–906. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
El-Karim SS, Syam YM, El Kerdawy AM and Abdel-Mohsen HT: Rational design and synthesis of novel quinazolinone N-acetohydrazides as type II multi-kinase inhibitors and potential anticancer agents. Bioorg Chem. 142:1069202024. View Article : Google Scholar | |
|
Kabir E and Uzzaman M: A review on biological and medicinal impact of heterocyclic compounds. Results Chem. 4:1006062022. View Article : Google Scholar | |
|
Almulla AF: A review: Biological importance of heterocyclic compounds. Der Pharma Chemica. 9:141–147. 2017. | |
|
Arora P, Arora V, Lamba HS and Wadhwa D: Importance of heterocyclic chemistry:A review. Int J Pharm Sci Res. 3:2947–2954. 2012. | |
|
Kumar B, Babu NJ and Chowhan RL: Sustainable synthesis of highly diastereoselective & fluorescent active spirooxindoles catalyzed by copper oxide nanoparticle immobilized on microcrystalline cellulose. Appl Organomet Chem. 36: View Article : Google Scholar : 2022. View Article : Google Scholar | |
|
Khan I, Ibrar A, Abbas N and Saeed A: Recent advances in the structural library of functionalized quinazoline and quinazolinone scaffolds: Synthetic approaches and multifarious applications. Eur J Med Chem. 76:193–244. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Borah B, Swain S, Patat M and Chowhan LR: Recent advances and prospects in the organocatalytic synthesis of quinazolinones. Front Chem. 10:9910262022. View Article : Google Scholar : PubMed/NCBI | |
|
Faisal M and Saeed A: Chemical insights into the synthetic chemistry of quinazolines: Recent advances. Front Chem. 8:5947172021. View Article : Google Scholar : PubMed/NCBI | |
|
Alsibaee AM, Al-Yousef HM and Al-Salem HS: Quinazolinones, the winning horse in drug discovery. Molecules. 28:9782023. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma PC, Kaur G, Pahwa R, Sharma A and Rajak H: Quinazolinone analogs as potential therapeutic agents. Curr Med Chem. 18:4786–4812. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Khandelwal P, Wadhwani BD, Rao RS, Mali D, Vyas P, Kumar T and Nair R: Exploring the pharmacological and chemical aspects of pyrrolo-quinazoline derivatives in Adhatoda vasica. Heliyon. 10:e257272024. View Article : Google Scholar : PubMed/NCBI | |
|
Padmanabhan D, Manimekalai R, Senthil-Nathan S, Suganthi M and Palanisamy S: Biosynthesis, therapeutic characteristics, origin and strategies to improve the yield of vasicine in plants. Vegetos. 186:2025. | |
|
Zhang Y, Du W, Zhu D, Li M, Qu L, Rao G, Lin Y, Tong X, Sun Y and Huang F: Vasicine alleviates 2,4-dinitrochlorobenzene-induced atopic dermatitis and passive cutaneous anaphylaxis in BALB/c mice. Clin Immunol. 244:1091022022. View Article : Google Scholar : PubMed/NCBI | |
|
Ali SK, Hamed AR, Soltan MM, El-Halawany AM, Hegazy UM and Hussein AA: Kinetics and molecular docking of vasicine from Adhatoda vasica: An acetylcholinesterase inhibitor for Alzheimer's disease. S Afr J Bot. 104:118–124. 2016. View Article : Google Scholar | |
|
Srinivasarao D, Jayarraj IA, Jayraaj R and Prabha ML: A study on antioxidant and anti-inflammatory activity of vasicine against lung damage in rats. Indian J Allergy Asthma Immunol. 20:1–7. 2006. | |
|
Eguchi S: Quinazoline alkaloids and related chemistry. Bioactive Heterocycles I. 6:113–156. 2006. View Article : Google Scholar | |
|
Ghosh P, Ganguly B and Das S: C-H functionalization of quinazolinones by transition metal catalysis. Org Biomol Chem. 18:4497–4518. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Rakesh KP, Darshini N, Shubhavathi T and Mallesha N: Biological applications of quinazolinone analogues: A review. Org Med Chem Int J. 2:41–45. 2017. | |
|
Kalogirou AS, Kourtellaris A and Koutentis PA: Synthesis of 2-Cyanoquinazolin-4-ones from 3',5'-Dichloro-1H-spiro (quinazoline-2,4'-[1,2,6]thiadiazin)-4(3H)-ones. ChemistrySelect. 5:1884–1889. 2020. View Article : Google Scholar | |
|
Zeng R, Huang C, Wang J, Zhong Y, Fang Q, Xiao S, Nie X, Chen S and Peng D: Synthesis, crystal structure, and antifungal activity of quinazolinone derivatives. Crystals. 13:12542023. View Article : Google Scholar | |
|
Ghoneim MM, Abdelgawad MA, Elkanzi NAA, Parambi DGT, Alsalahat I, Farouk A and Bakr RB: A literature review on pharmacological aspects, docking studies, and synthetic approaches of quinazoline and quinazolinone derivatives. Arch Pharm (Weinheim). 357:e24000572024. View Article : Google Scholar : PubMed/NCBI | |
|
Haneen DSA, Abdalha AA, Alkhatib MM, Kamal M, Youssef ASA, Abou-Elmagd WSI and Samir SS: Synthesis, comprehensive in silico studies, and cytotoxicity evaluation of novel quinazolinone derivatives as potential anticancer agents. Sci Rep. 15:236972025. View Article : Google Scholar : PubMed/NCBI | |
|
Borik RM and Hussein MA: A novel quinazoline-4-one derivatives as a promising cytokine inhibitors: Synthesis, molecular docking, and structure-activity relationship. Curr Pharm Biotechnol. 23:1179–1203. 2022. View Article : Google Scholar | |
|
Samotrueva MA, Starikova AA, Bashkina OA, Tsibizova AA, Borisov AV, Merezhkina DV, Tyurenkov IN and Ozerov AA: Biochemical basis of the antimicrobial activity of quinazolinone derivatives in the light of insights into the features of the chemical structure and ways of binding to target molecules. A review. Dokl Chem. 510:107–129. 2023. View Article : Google Scholar | |
|
Mhaske SB and Argade NP: The chemistry of recently isolated naturally occurring quinazolinone alkaloids. Tetrahedron. 62:9787–9826. 2006. View Article : Google Scholar | |
|
Mahato AK, Srivastava B and Shanthi CN: Chemistry structure activity relationship and biological activity of quinazoline-4(3H)-one derivatives. Inventi Rapid: MedChem. 1:0976–7541. 2011. | |
|
Asif M: Chemical characteristics, synthetic methods, and biological potential of quinazoline and quinazolinone derivatives. Int J Med Chem. 2014:1–27. 2014. | |
|
Garofalo A, Goossens L, Baldeyrou B, Lemoine A, Ravez S, Six P, David-Cordonnier MH, Bonte JP, Depreux P, Lansiaux A and Goossens JF: Design, synthesis, and DNA-binding of N-Alkyl(anilino)quinazoline derivatives. J Med Chem. 53:8089–8103. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
El-Malah A, Malebari AM, Khayyat AN, Mohammad KA, Gineinah MM and Mahmoud Z: Design, synthesis, and antiproliferative activities of novel substitutedhydrazone/triazolo-linked quinazoline derivatives. J Mol Struct. 1306:1378222024. View Article : Google Scholar | |
|
Haghighijoo Z, Firuzi O, Hemmateenejad B, Emami S, Edraki N and Miri R: Synthesis and biological evaluation of quinazolinone-based hydrazones with potential use in Alzheimer's disease. Bioorg Chem. 74:126–133. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Khan I, Ibrar A, Ahmed W and Saeed A: Synthetic approaches, functionalization and therapeutic potential of quinazoline and quinazolinone skeletons: The advances continue. Eur J Med Chem. 90:124–169. 2015. View Article : Google Scholar | |
|
Kaur J, Kaur S, Muskan, Kaur N, Kumar V and Anand A: Unveiling the therapeutic potential of quinazolinone derivatives in cancer treatment: A comprehensive exploration. ChemistrySelect. 9:e2024013662024. View Article : Google Scholar | |
|
Haider K, Das S, Joseph A and Yar MS: An appraisal of anticancer activity with structure-activity relationship of quinazoline and quinazolinone analogues through EGFR and VEGFR inhibition: A review. Drug Dev Res. 83:859–890. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Reddy MM and Sivaramakrishna A: Remarkably flexible quinazolinones-synthesis and biological applications. J Heterocycl Chem. 57:942–954. 2019. View Article : Google Scholar | |
|
He D, Wang M, Zhao S, Shu Y, Zeng H, Xiao C, Lu C and Liu Y: Pharmaceutical prospects of naturally occurring quinazolinone and its derivatives. Fitoterapia. 119:136–149. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Shankar GM, Alex VV, Nisthul AA, Bava SV, Sundaram S, Retnakumari AP, Chittalakkottu S and Anto RJ: Pre-clinical evidences for the efficacy of tryptanthrin as a potent suppressor of skin cancer. Cell Prolif. 53:e127102020. View Article : Google Scholar | |
|
Li H, Fu G and Zhong W: Natural quinazolinones: From a treasure house to promising anticancer leads. Eur J Med Chem. 245:1149152023. View Article : Google Scholar | |
|
Kushwaha N, Sahu A, Mishra J, Soni A and Dorwal D: An insight on the prospect of quinazoline and quinazolinone derivatives as anti-tubercular agents. Curr Org Synth. 20:838–869. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Jaiswal S, Verma K, Srivastva A, Arya N, Dwivedi J and Sharma S: Green synthetic and pharmacological developments in the hybrid quinazolinone moiety: An updated review. Curr Top Med Chem. 25:493–532. 2025. View Article : Google Scholar | |
|
Wu C, Wang Y, Yang F, Shi W, Wang Z, He L, He Y and Shen J: Synthesis and biological evaluation of five-atom-linker-based arylpiperazine derivatives with an atypical antipsychotic profile. ChemMedChem. 14:2042–2051. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Wei M, Chai WM, Wang R, Yang Q, Deng Z and Peng Y: Quinazolinone derivatives: Synthesis and comparison of inhibitory mechanisms on alpha-glucosidase. Bioorg Med Chem. 25:1303–1308. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Obeng E: Apoptosis (programmed cell death) and its signals-A review. Braz J Biol. 81:1133–1143. 2021. View Article : Google Scholar | |
|
Nair P, Lu M, Petersen S and Ashkenazi A: Apoptosis initiation through the cell-extrinsic pathway. Methods Enzymol. 544:99–128. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Lossi L: The concept of intrinsic versus extrinsic apoptosis. Biochem J. 479:357–384. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Hongmei Z: Extrinsic and intrinsic apoptosis signal pathway review. Apoptosis and Medicine. 2012. View Article : Google Scholar | |
|
Ashkenazi A: Targeting the extrinsic apoptosis pathway in cancer. Cytokine Growth Factor Rev. 19:325–331. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Liang Y, Zheng Y, Yang J, Ke J and Cheng K: Design, synthesis and bioactivity evaluation of a series of quinazolinone derivatives as potent PI3Kγ antagonist. Bioorg Med Chem. 84:1172612023. View Article : Google Scholar | |
|
Kim YS, Cheon MG, Boggu PR, Koh SY, Park GM, Kim G, Park SH, Park SL, Lee CW, Kim JW and Jung YH: Synthesis and biological evaluation of novel purinyl quinazolinone derivatives as PI3Kδ-specific inhibitors for the treatment of hematologic malignancies. Bioorg Med Chem. 45:1163122021. View Article : Google Scholar | |
|
Wani ZA, Guru SK, Rao AVS, Sharma S, Mahajan G, Behl A, Kumar A, Sharma PR, Kamal A, Bhushan S and Mondhe DM: A novel quinazolinone chalcone derivative induces mitochondrial dependent apoptosis and inhibits PI3K/Akt/mTOR signaling pathway in human colon cancer HCT-116 cells. Food Chem Toxicol. 87:1–11. 2016. View Article : Google Scholar | |
|
Madbouly EA, Lashine ESM, Al-Karmalawy AA, Sebaiy MM, Pratsinis H, Kletsas D and Metwally K: Design and synthesis of novel quinazolinone-chalcone hybrids as potential apoptotic candidates targeting caspase-3 and PARP-1:In vitro, molecular docking, and SAR studies. New J Chem. 46:22013–22029. 2022. View Article : Google Scholar | |
|
Xie J, Yang MR, Hu X, Hong ZS, Bai YY, Sheng J, Tian Y and Shi CY: Moringa oleifera Lam. Isothiocyanate quinazolinone derivatives inhibit U251 glioma cell proliferation through cell cycle regulation and apoptosis induction. Int J Mol Sci. 24:113762023. View Article : Google Scholar : PubMed/NCBI | |
|
Qiu J, Zhou Q, Zhang Y, Guan M, Li X, Zou Y, Huang X, Zhao Y, Chen W and Gu X: Discovery of novel quinazolinone derivatives as potential anti-HBV and anti-HCC agents. Eur J Med Chem. 205:1125812020. View Article : Google Scholar : PubMed/NCBI | |
|
El-Shafey HW, Gomaa RM, El-Messery SM and Goda FE: Synthetic approaches, anticancer potential, HSP90 inhibition, multitarget evaluation, molecular modeling and apoptosis mechanistic study of thioquinazolinone skeleton: Promising antibreast cancer agent. Bioorg Chem. 101:1039872020. View Article : Google Scholar : PubMed/NCBI | |
|
Hour MJ, Tsai FJ, Lai IL, Tsao JW, Chiang JH, Chiu YJ, Lu HF, Juan YN, Yang JS and Tsai SC: Efficacy of HMJ-38, a new quinazolinone analogue, against the gemcitabine-resistant MIA-PaCa-2 pancreatic cancer cells. Biomedicine (Taipei). 13:20–31. 2023. View Article : Google Scholar | |
|
Chiang JH, Yang JS, Lu CC, Hour MJ, Chang SJ, Lee TH and Chung JG: Newly synthesized quinazolinone HMJ-38 suppresses angiogenetic responses and triggers human umbilical vein endothelial cell apoptosis through p53-modulated Fas/death receptor signaling. Toxicol Appl Pharmacol. 269:150–162. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Lu CC, Yang JS, Chiang JH, Hour MJ, Lin KL, Lin JJ, Huang WW, Tsuzuki M, Lee TH and Chung JG: Novel quinazolinone MJ-29 triggers endoplasmic reticulum stress and intrinsic apoptosis in murine leukemia WEHI-3 cells and inhibits leukemic mice. PLoS One. 7:e368312012. View Article : Google Scholar : PubMed/NCBI | |
|
Pope LE and Dixon SJ: Regulation of ferroptosis by lipid metabolism. Trends Cell Biol. 33:1077–1087. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Dixon SJ and Olzmann JA: The cell biology of ferroptosis. Nat Rev Mol Cell Biol. 25:424–442. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Stockwell BR and Conrad M: Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M, Guo M, Gao Y, Wu C, Pan X and Huang Z: Mechanisms and therapeutic targets of ferroptosis: Implications for nanomedicine design. J Pharm Anal. 14:1009602024. View Article : Google Scholar : PubMed/NCBI | |
|
Tang D and Kroemer G: Ferroptosis. Curr Biol. 30:R1292–R1297. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Yuan H, Li X, Zhang X, Kang R and Tang D: Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem Biophys Res Commun. 478:1338–1343. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Liang X, Long L, Guan F, Xu Z and Huang H: Research status and potential applications of circRNAs affecting colorectal cancer by regulating ferroptosis. Life Sci. 352:1228702024. View Article : Google Scholar : PubMed/NCBI | |
|
Yang Y, Luo M, Zhang K, Zhang J, Gao T, Connell DO, Yao F, Mu C, Cai B, Shang Y and Chen W: Nedd4 ubiquitylates VDAC2/3 to suppress erastin-induced ferroptosis in melanoma. Nat Commun. 11:4332020. View Article : Google Scholar : PubMed/NCBI | |
|
Sun Y, Deng R and Zhang C: Erastin induces apoptotic and ferroptotic cell death by inducing ROS accumulation by causing mitochondrial dysfunction in gastric cancer cell HGC-27. Mol Med Rep. 22:2826–2832. 2020.PubMed/NCBI | |
|
Li Y, Zeng X, Lu D, Yin M, Shan M and Gao Y: Erastin induces ferroptosis via ferroportin-mediated iron accumulation in endometriosis. Hum Reprod. 36:951–964. 2021. View Article : Google Scholar | |
|
Huang C, Yang M, Deng J, Li P, Su W and Jiang R: Upregulation and activation of p53 by erastin-induced reactive oxygen species contribute to cytotoxic and cytostatic effects in A549 lung cancer cells. Oncol Rep. 40:2363–2370. 2018.PubMed/NCBI | |
|
Badgley MA, Kremer DM, Maurer HC, DelGiorno KE, Lee HJ, Purohit V, Sagalovskiy IR, Ma A, Kapilian J, Firl CEM, et al: Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science. 368:85–89. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang Y and Sun M: SLC7A11: The Achilles heel of tumor? Front Immunol. 15:14388072024. View Article : Google Scholar : PubMed/NCBI | |
|
Sun X, Ou Z, Chen R, Niu X, Chen D, Kang R and Tang D: Activation of the p62-Keap1-NRF2 pathway protects against ferroptosis in hepatocellular carcinoma cells. Hepatology. 63:173–184. 2016. View Article : Google Scholar | |
|
Zhao X, Wang T, Shang F, Yan J, Jiang M, Zou X, Li G, Song Z and Huang J: Coumarin-Quinazolinone based photosensitizers: Mitochondria and endoplasmic reticulum targeting for enhanced phototherapy via different cell death pathways. Eur J Med Chem. 280:1169902024. View Article : Google Scholar : PubMed/NCBI | |
|
Xing Z, Yan J, Miao Y, Ruan Y, Yao H, Zhou Y, Tang Y, Li G, Song Z, Peng Y and Huang J: Endoplasmic reticulum-targeting quinazolinone-based lipophilic probe for specific photoinduced ferroptosis and its induced lipid dynamic regulation. J Med Chem. 67:1900–1913. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Huang Y, Xiong K, Wang A, Wang Z, Cui Q, Xie H, Yang T, Fan X, Jiang W, Tan X and Huang Q: Cold stress causes liver damage by inducing ferroptosis through the p38 MAPK/Drp1 pathway. Cryobiology. 113:1045632023. View Article : Google Scholar : PubMed/NCBI | |
|
Li X, Ran Q, He X, Peng D, Xiong A, Jiang M, Zhang L, Wang J, Bai L, Liu S, et al: HO-1 upregulation promotes mitophagy-dependent ferroptosis in PM2.5-exposed hippocampal neurons. Ecotoxicol Environ Saf. 277:1163142024. View Article : Google Scholar : PubMed/NCBI | |
|
Nakamura T, Hipp C, Dias Mourão A, Borggräfe J, Aldrovandi M, Henkelmann B, Wanninger J, Mishima E, Lytton E, Emler D, et al: Phase separation of FSP1 promotes ferroptosis. Nature. 619:371–377. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Wu Z, Zhu Y, Liu W, Balasubramanian B, Xu X, Yao J and Lei X: Ferroptosis in liver disease: Natural active compounds and therapeutic implications. Antioxidants (Basel). 13:3522024. View Article : Google Scholar : PubMed/NCBI | |
|
Balushi KA, Hadhrami AA, Balushi HA, Lawati AA and Das S: Tebentafusp as a promising drug for the treatment of uveal melanoma. Curr Drug Targets. 25:149–157. 2024. View Article : Google Scholar | |
|
Hu S, Sechi M, Singh PK, Dai L, McCann S, Sun D, Ljungman M and Neamati N: A novel redox modulator induces a GPX4-mediated cell death that is dependent on iron and reactive oxygen species. Eur J Med Chem. 63:9838–9855. 2020. View Article : Google Scholar | |
|
Cheng Z: The FoxO-autophagy axis in health and disease. Trends Endocrinol Metab. 30:658–671. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Galluzzi L, Vitale I, Aaronson SA, Abrams JM, Adam D, Agostinis P, Alnemri ES, Altucci L, Amelio I, Andrews DW, et al: Molecular mechanisms of cell death: Recommendations of the nomenclature committee on cell death 2018. Cell Death Differ. 25:486–541. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Buzun K, Gornowicz A, Lesyk R, Bielawski K and Bielawska A: Autophagy modulators in cancer therapy. Int J Mol Sci. 22:58042021. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang B and Liu L: Autophagy is a double-edged sword in the therapy of colorectal cancer. Oncol Lett. 21:3782021. View Article : Google Scholar : PubMed/NCBI | |
|
Gump JM and Thorburn A: Autophagy and apoptosis: What is the connection? Trends Cell Biol. 21:387–392. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Kumar S, Guru SK, Pathania AS, Mupparapu N, Kumar A, Malik F, Bharate SB, Ahmed QN, Vishwakarma RA and Bhushan S: A novel quinazolinone derivative induces cytochrome c interdependent apoptosis and autophagy in human leukemia MOLT-4 cells. Toxicol Rep. 1:1013–1025. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma R, Chatterjee E, Mathew J, Sharma S, Rao NV, Pan CH, Lee SB, Dhingra A, Grewal AS, Liou JP, et al: Accommodation of ring C expanded deoxyvasicinone in the HDAC inhibitory pharmacophore culminates into a tractable anti-lung cancer agent and pH-responsive nanocarrier. Eur J Med Chem. 240:1146022022. View Article : Google Scholar : PubMed/NCBI | |
|
Xia X, Wang L, Zhang X, Wang S, Lei L, Cheng L, Xu Y, Sun Y, Hang B, Zhang G, et al: Halofuginone-induced autophagy suppresses the migration and invasion of MCF-7 cells via regulation of STMN1 and p53. J Cell Biochem. 119:4009–4020. 2018. View Article : Google Scholar | |
|
Ha HA, Chiang JH, Tsai FJ, Bau DT, Juan YN, Lo YH, Hour MJ and Yang JS: Novel quinazolinone MJ-33 induces AKT/mTOR-mediated autophagy-associated apoptosis in 5FU-resistant colorectal cancer cells. Oncol Rep. 45:680–692. 2020. View Article : Google Scholar | |
|
ElZahabi HSA, Nafie MS, Osman D, Elghazawy NH, Soliman DH, El-Helby AAH and Arafa RK: Design, synthesis and evaluation of new quinazolin-4-one derivatives as apoptotic enhancers and autophagy inhibitors with potent antitumor activity. Eur J Med Chem. 222:1136092021. View Article : Google Scholar : PubMed/NCBI | |
|
Roger L, Tomas F and Gire V: Mechanisms and regulation of cellular senescence. Int J Mol Sci. 22:131732021. View Article : Google Scholar : PubMed/NCBI | |
|
Huang W, Hickson LJ, Eirin A, Kirkland JL and Lerman LO: Cellular senescence: The good, the bad and the unknown. Nat Rev Nephrol. 18:611–627. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
de Magalhães JP: Cellular senescence in normal physiology. Science. 384:1300–1301. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Schmitt CA, Wang B and Demaria M: Senescence and cancer-role and therapeutic opportunities. Nat Rev Clin Oncol. 19:619–636. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang L, Lankhorst L and Bernards R: Exploiting senescence for the treatment of cancer. Nat Rev Cancer. 22:340–355. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Kamal A, Sultana F, Ramaiah MJ, Srikanth YVV, Viswanath A, Bharathi EV, Nayak R, Pushpavalli SNCVL, Srinivas C and Pal-Bhadra M: 3-Diarylethyne quinazolinones: A new class of senescence inducers. Med Chem Comm. 4:575–581. 2013. View Article : Google Scholar | |
|
Venkatesh R, Ramaiah MJ, Gaikwad HK, Janardhan S, Bantu R, Nagarapu L, Sastry GN, Ganesh AR and Bhadra M: Luotonin-A based quinazolinones cause apoptosis and senescence via HDAC inhibition and activation of tumor suppressor proteins in HeLa cells. Eur J Med Chem. 94:87–101. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Proskuryakov SY and Gabai VL: Mechanisms of tumor cell necrosis. Curr Pharm Des. 16:56–58. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Szabó C: Mechanisms of cell necrosis. Crit Care Med. 33:530–534. 2005. View Article : Google Scholar | |
|
Loveless R, Bloomquist R and Teng Y: Pyroptosis at the forefront of anticancer immunity. J Exp Clin Cancer Res. 40:2642021. View Article : Google Scholar : PubMed/NCBI | |
|
Wei X, Xie F, Zhou X, Wu Y, Yan H, Liu T, Huang J, Wang F, Zhou F and Zhang L: Role of pyroptosis in inflammation and cancer. Cell Mol Immunol. 19:971–992. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Shams LM, Minaei-Tehrani D, Gholipour H and Nohehkhan M: Effects of quinazolinones on Balb/C mice embryonic livers. Indian J Exp Biol. 49:183–190. 2011.PubMed/NCBI | |
|
Piamsiri C, Maneechote C, Jinawong K, Arunsak B, Chunchai T, Nawara W, Kerdphoo S, Chattipakorn SC and Chattipakorn N: Chronic mitochondrial dynamic-targeted therapy alleviates left ventricular dysfunction by reducing multiple programmed cell death in post-myocardial infarction rats. Eur J Pharmacol. 977:1767362024. View Article : Google Scholar : PubMed/NCBI | |
|
Li L, Mu Z, Liu P, Wang Y, Yang F and Han X: Mdivi-1 alleviates atopic dermatitis through the inhibition of NLRP3 inflammasome. Exp Dermatol. 30:1734–1744. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Tanriover C, Copur S, Ucku D, Cakir AB, Hasbal NB, Soler MJ and Kanbay M: The mitochondrion: A promising target for kidney disease. Pharmaceutics. 15:5702023. View Article : Google Scholar : PubMed/NCBI | |
|
Liu R, Wang SC, Li M, Ma XH, Jia XN, Bu Y, Sun L and Yu KJ: An inhibitor of DRP1 (Mdivi-1) alleviates LPS-induced septic AKI by inhibiting NLRP3 inflammasome activation. Biomed Res Int. 2020:1–11. 2020. | |
|
Qian W, Wang J, Roginskaya V, McDermott LA, Edwards RP, Stolz DB, Llambi F, Green DR and Van Houten B: Novel combination of mitochondrial division inhibitor 1 (mdivi-1) and platinum agents produces synergistic pro-apoptotic effect in drug resistant tumor cells. Oncotarget. 5:4180–4194. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Tusskorn O, Khunluck T, Prawan A, Senggunprai L and Kukongviriyapan V: Mitochondrial division inhibitor-1 potentiates cisplatin-induced apoptosis via the mitochondrial death pathway in cholangiocarcinoma cells. Biomed Pharmacother. 111:109–118. 2019. View Article : Google Scholar | |
|
Vargo JW, Walker SN, Gopal SR, Deshmukh AR, McDermott BM Jr, Alagramam KN and Stepanyan R: Inhibition of mitochondrial division attenuates cisplatin-induced toxicity in the neuromast hair cells. Front Cell Neurosci. 11:3932017. View Article : Google Scholar | |
|
Lai KC, Chia YT, Yih LH, Lu YL, Chang ST, Hong ZX, Chen TL and Hour MJ: Antitumor effects of the novel quinazolinone Holu-12: Induction of mitotic arrest and apoptosis in human oral squamous cell carcinoma CAL27 cells. Anticancer Res. 41:259–268. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Pathania D, Kuang Y, Sechi M and Neamati N: Mechanisms underlying the cytotoxicity of a novel quinazolinedione-based redox modulator, QD232, in pancreatic cancer cells. Br J Pharmacol. 172:50–63. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou J, Ji M, Yao H, Cao R, Zhao H, Wang X, Chen X and Xu B: Discovery of quinazoline-2,4(1H,3H)-dione derivatives as novel PARP-1/2 inhibitors: Design, synthesis and their antitumor activity. Org Biomol Chem. 16:3189–3202. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Zhou Q, Ji M, Zhou J, Jin J, Xue N, Chen J, Xu B and Chen X: Poly (ADP-ribose) polymerases inhibitor, Zj6413, as a potential therapeutic agent against breast cancer. Biochem Pharmacol. 107:29–40. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Giannini G, Battistuzzi G, Vesci L, Milazzo FM, De Paolis F, Barbarino M, Guglielmi MB, Carollo V, Gallo G, Artali R and Dallavalle S: Novel PARP-1 inhibitors based on a 2-propanoyl-3H-quinazolin-4-one scaffold. Bioorg Med Chem Lett. 24:462–466. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ma Y, Yang J, Wei X, Pei Y, Ye J, Li X, Si G, Tian J, Dong Y and Liu G: Nonpeptidic quinazolinone derivatives as dual nucleotide-binding oligomerization domain-like receptor 1/2 antagonists for adjuvant cancer chemotherapy. Eur J Med Chem. 207:1127232020. View Article : Google Scholar : PubMed/NCBI | |
|
Smolewski P and Rydygier D: Efficacy and safety of idelalisib for the treatment of indolent B-cell malignancies. Expert Opin Pharmacother. 21:1915–1926. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Miller BW, Przepiorka D, de Claro RA, Lee K, Nie L, Simpson N, Gudi R, Saber H, Shord S, Bullock J, et al: FDA approval: Idelalisib monotherapy for the treatment of patients with follicular lymphoma and small lymphocytic Lymphoma. Clin Cancer Res. 21:1525–1529. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Merli M, Passamonti F and Arcaini L: The double significance of idelalisib immune-related toxicity. Leuk Lymphoma. 62:2815–2817. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Liu K, Li D, Zheng W, Shi M, Chen Y, Tang M, Yang T, Zhao M, Deng D, Zhang C, et al: Discovery, optimization, and evaluation of quinazolinone derivatives with novel linkers as orally efficacious phosphoinositide-3-kinase delta inhibitors for treatment of inflammatory diseases. J Med Chem. 64:8951–8970. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yu M, Chen J, Xu Z, Yang B, He Q, Luo P, Yan H and Yang X: Development and safety of PI3K inhibitors in cancer. Arch Toxicol. 97:635–650. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ramanathan S, Jin F, Sharma S and Kearney BP: Clinical pharmacokinetic and pharmacodynamic profile of idelalisib. Clin Pharmacokinet. 55:33–45. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Wiese W, Barczuk J, Racinska O, Siwecka N, Rozpedek-Kaminska W, Slupianek A, Sierpinski R and Majsterek I: PI3K/Akt/mTOR signaling pathway in blood malignancies-new therapeutic possibilities. Cancers (Basel). 15:52972023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu J, Wang P, Shehu AI, Lu J, Bi H and Ma X: Identification of novel pathways in idelalisib metabolism and bioactivation. Chem Res Toxicol. 31:548–555. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Graf SA and Gopal AK: Idelalisib for the treatment of non-Hodgkin lymphoma. Expert Opin Pharmacother. 17:265–274. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Lin X, Zhang Y, Huang H, Zhuang W and Wu L: Post-marketing safety concern of PI3K inhibitors in the cancer therapies: An 8-year disproportionality analysis from the FDA adverse event reporting system. Expert Opin Drug Saf. 24:1–12. 2024. | |
|
Hus I, Puła B and Robak T: PI3K inhibitors for the treatment of chronic lymphocytic leukemia: Current status and future perspectives. Cancers (Basel). 14:15712022. View Article : Google Scholar : PubMed/NCBI | |
|
Zirlik K and Veelken H: Idelalisib. Recent Results Cancer Res. 212:243–264. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Scheffold A, Jebaraj BMC, Tausch E, Bloehdorn J, Ghia P, Yahiaoui A, Dolnik A, Blätte TJ, Bullinger L, Dheenadayalan RP, et al: IGF1R as druggable target mediating PI3K-δ inhibitor resistance in a murine model of chronic lymphocytic leukemia. Blood. 134:534–547. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Park GB, Chung YH, Jeong JY and Kim D: A p110δ-specific inhibitor combined with bortezomib blocks drug resistance properties of EBV-related B cell origin cancer cells via regulation of NF-κB. Int J Oncol. 50:1711–1720. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Alossaimi MA, Riadi Y, Alnuwaybit GN, Md S, Alkreathy HM, Elekhnawy E, Geesi MH, Alqahtani SM and Afzal O: Design, synthesis, molecular docking, and in vitro studies of 2-mercaptoquinazolin-4(3H)-ones as potential anti-breast cancer agents. Saudi Pharm J. 32:1019712024. View Article : Google Scholar : PubMed/NCBI | |
|
Škubník J, Jurášek M, Ruml T and Rimpelová S: Mitotic poisons in research and medicine. Molecules. 25:46322020. View Article : Google Scholar : PubMed/NCBI | |
|
Shahin R and Aljamal S: Kinesin spindle protein inhibitors in cancer: From high throughput screening to novel therapeutic strategies. Future Sci OA. 8:FSO7782022. View Article : Google Scholar : PubMed/NCBI | |
|
Lee CW, Bélanger K, Rao SC, Petrella TM, Tozer RG, Wood L, Savage KJ, Eisenhauer EA, Synold TW, Wainman N and Seymour L: A phase II study of ispinesib (SB-715992) in patients with metastatic or recurrent malignant melanoma: A national cancer institute of Canada clinical trials group trial. Invest New Drugs. 26:249–255. 2008. View Article : Google Scholar | |
|
Purcell JW, Davis J, Reddy M, Martin S, Samayoa K, Vo H, Thomsen K, Bean P, Kuo WL, Ziyad S, et al: Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992) in models of breast cancer. Clin Cancer Res. 16:566–576. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Lee RT, Beekman KE, Hussain M, Davis NB, Clark JI, Thomas SP, Nichols KF and Stadler WM: A University of Chicago consortium phase II trial of SB-715992 in advanced renal cell cancer. Clin Genitourin Cancer. 6:21–24. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Murase Y, Ono H, Ogawa K, Yoshioka R, Ishikawa Y, Ueda H, Akahoshi K, Ban D, Kudo A, Tanaka S and Tanabe M: Inhibitor library screening identifies ispinesib as a new potential chemotherapeutic agent for pancreatic cancers. Cancer Sci. 112:4641–4654. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Gomez HL, Philco M, Pimentel P, Kiyan M, Monsalvo ML, Conlan MG, Saikali KG, Chen MM, Seroogy JJ, Wolff AA and Escandon RD: Phase I dose-escalation and pharmacokinetic study of ispinesib, a kinesin spindle protein inhibitor, administered on days 1 and 15 of a 28-day schedule in patients with no prior treatment for advanced breast cancer. Anticancer Drugs. 23:335–341. 2012. View Article : Google Scholar | |
|
Kenchappa RS, Dovas A, Argenziano MG, Meyer CT, Stopfer LE, Banu MA, Pereira B, Griffith J, Mohammad A, Talele S, et al: Activation of STAT3 through combined SRC and EGFR signaling drives resistance to a mitotic kinesin inhibitor in glioblastoma. Cell Rep. 39:1109912022. View Article : Google Scholar : PubMed/NCBI | |
|
Ansbro MR, Shukla S, Ambudkar SV, Yuspa SH and Li L: Screening compounds with a novel high-throughput ABCB1-mediated efflux assay identifies drugs with known therapeutic targets at risk for multidrug resistance interference. PLoS One. 8:e603342013. View Article : Google Scholar : PubMed/NCBI | |
|
Purcell JW, Davis J, Reddy M, Martin S, Samayoa K, Vo H, Thomsen K, Bean P, Kuo WL, Ziyad S, et al: Activity of the kinesin spindle protein inhibitor ispinesib (SB-715992) in models of breast cancer. Clin Cancer Res. 16:566–576. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Gish RG, Porta C, Lazar L, Ruff P, Feld R, Croitoru A, Feun L, Jeziorski K, Leighton J, Gallo J and Kennealey GT: Phase III randomized controlled trial comparing the survival of patients with unresectable hepatocellular carcinoma treated with nolatrexed or doxorubicin. J Clin Oncol. 25:3069–3075. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Lord R, Suddle A and Ross PJ: Emerging strategies in the treatment of advanced hepatocellular carcinoma: The role of targeted therapies. Int J Clin Pract. 65:182–188. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Gish RG, Porta C, Lazar L, Ruff P, Feld R, Croitoru A, Feun L, Jeziorski K, Leighton J, Knox J and Kennealey GT: Phase III randomized controlled trial comparing the survival of patients with unresectable hepatocellular carcinoma treated with nolatrexed or doxorubicin. J Clin Oncol. 25:3069–3075. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Giovannetti E, Backus HH, Wouters D, Ferreira CG, van Houten VM, Brakenhoff RH, Poupon MF, Azzarello A, Pinedo HM and Peters GJ: Changes in the status of p53 affect drug sensitivity to thymidylate synthase (TS) inhibitors by altering TS levels. Br J Cancer. 96:769–775. 2007. View Article : Google Scholar : PubMed/NCBI | |
|
Van Triest B, Pinedo HM, Giaccone G and Peters GJ: Downstream molecular determinants of response to 5-fluorouracil and antifolate thymidylate synthase inhibitors. Ann Oncol. 11:385–391. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Hasan RU, Mian SS, Arfi S, Begum B, Verma S, Ahmad R, Hussain I and Asif M: Study of pharmacologically active drugs containing quinazoline pharmacophore: A brief overview. J Adv Zool. 45:1166–1184. 2024. | |
|
Han Y, Liu S, Zhu J, Liu P, Meng Z, Li Y, Li S, Fan F, Zhang M and Liu H: Experimental study on the inhibitory effect of Halofuginone on NSCLC. European Journal of Pharmacology. 2024. | |
|
Chen Y, Liu W and Wang P, Hou H, Liu N, Gong L, Wang Y, Ji K, Zhao L and Wang P: Halofuginone inhibits radiotherapy-induced epithelial mesenchymal transition in lung cancer. Oncotarget. 7:71341–71352. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Zuo R, Guo X, Song X, Gao X, Zhang J, Jiang S, Adam V, Kuca K, Wu W and Guo D: New uses of halofuginone to treat cancer. J Pharm Anal. 15:1010802024. View Article : Google Scholar | |
|
Mi L, Liu J, Zhang Y, Su A, Tang M, Xing Z, He T, Wei T, Li Z and Wu W: The EPRS-ATF4-COLI pathway axis is a potential target for anaplastic thyroid carcinoma therapy. Phytomedicine. 129:1556702024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhao H, Li R, Chen Y, Yang X and Shang Z: Stromal nicotinamide N-methyltransferase orchestrates the crosstalk between fibroblasts and tumour cells in oral squamous cell carcinoma: Evidence from patient-derived assembled organoids. Oncogene. 42:1166–1180. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Panda H, Suzuki M, Naito M, Saito R, Wen H, Baird L, Uruno A, Miyata K and Yamamoto M: Halofuginone micelle nanoparticles eradicate Nrf2-activated lung adenocarcinoma without systemic toxicity. Free Radic Biol Med. 187:92–104. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Li Y, Liu P, Liu S, Zhu J, Han Y, Jiang Z, Tang D, Meng Z, Li S, Zhang M, et al: Halofuginone targets Serine/Glycine synthesis to reverse epidermal growth factor receptor tyrosine Kinase inhibitor resistance in lung adenocarcinoma. Phytomedicine. 143:1567882025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang L, Yue G, Lu Y and Tang J: BRCA1 as a target for attenuating paclitaxel resistance by Halofuginone treatment in basal-like breast cancer. J Funct Foods. 118:1062452024. View Article : Google Scholar | |
|
Wang C, Zhu JB, Yan YY, Zhang W, Gong XJ, Wang X and Wang XL: Halofuginone inhibits tumorigenic progression of 5-FU-resistant human colorectal cancer HCT-15/FU cells by targeting miR-132-3p in vitro. Oncol Lett. 20:3852020. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu S, Wang J, Chandrashekar G, Smith E, Liu X and Zhang Y: Synthesis and evaluation of 4-quinazolinone compounds as potential antimalarial agents. Eur J Med Chem. 45:3864–3869. 2010. View Article : Google Scholar : PubMed/NCBI | |
|
Zuo R, Zhang J, Song X, Hu S, Gao X, Wang J, Ji H, Ji C, Peng L, Si H, et al: Encapsulating halofuginone hydrobromide in TPGS polymeric micelles enhances efficacy against triple-negative breast cancer cells. Int J Nanomedicine. 16:1587–1600. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen J, Fan S, Guo J, Yang J, Pan L and Xia Y: Discovery of anticancer function of Febrifugine: Inhibition of cell proliferation, induction of apoptosis and suppression steroid synthesis in bladder cancer cells. Toxicol Appl Pharmacol. 484:1168782024. View Article : Google Scholar : PubMed/NCBI | |
|
Lin F, Wang R, Du J, Wen C, Wang X, Jin Y and Shao L: Pharmacokinetics and bioavailability of febrifugine in rat plasma determined by UPLC-MS/MS. Acta Chromatogr. 37: View Article : Google Scholar : 2024. | |
|
Chen J, Fan S, Guo J, Yang J, Pan L and Xia Y: Discovery of anticancer function of Febrifugine: Inhibition of cell proliferation, induction of apoptosis and suppression steroid synthesis in bladder cancer cells. Toxicol Appl Pharmacol. 484:1168782024. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang J, Xu HX, Zhu JQ, Dou YX, Xian YF and Lin ZX: Natural Nrf2 inhibitors: A review of their potential for cancer treatment. Int J Biol Sci. 19:3029–3041. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Zhu J, Liu S, Liu P, Zhai K and Liu H: Traditional medicine meets modern science: Halofuginone's role in combating autoimmune diseases. J Nat Med. 79:1017–1029. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Hasan RU, Mian SS, Arfi S, Begum B, Verma S, Ahmad R, Hussain I and Asif M: Study of pharmacologically active drugs containing quinazoline pharmacophore: A brief overview. J Adv Zool. 45:1166–1184. 2024. | |
|
Haider K, Das S, Joseph A and Yar MS: An appraisal of anticancer activity with structure-activity relationship of quinazoline and quinazolinone analogues through EGFR and VEGFR inhibition: A review. Drug Dev Res. 83:859–890. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang X, Yu J, Zhou Z, Kongsted J, Song Y, Pannecouque C, De Clercq E, Kang D, Poongavanam V, Liu X and Zhan P: Molecular design opportunities presented by solvent-exposed regions of target proteins. Med Res Rev. 39:2194–2238. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
El-Sayed NNE, Al-Otaibi TM, Barakat A, Almarhoon ZM, Hassan MZ, Al-Zaben MI, Krayem N, Masand VH and Bacha AB: Synthesis and biological evaluation of some New 3-Aryl-2-thioxo-2,3-dihydroquinazolin-4(1H)-ones and 3-Aryl-2-(benzylthio)quinazolin-4(3H)-ones as antioxidants; COX-2, LDHA, α-Glucosidase and α-amylase inhibitors; and anti-colon carcinoma and apoptosis-inducing agents. Pharmaceuticals (Basel). 16:13922023. View Article : Google Scholar | |
|
Kurogi Y, Inoue Y, Tsutsumi K, Nakamura S, Nagao K, Yoshitsugu H and Tsuda Y: Synthesis and hypolipidemic activities of novel 2-[4-[diethoxyphosphoryl)methyl]phenyl] quinazolines and 4(3H)-quinazolinones. J Med Chem. 39:1433–1437. 1996. View Article : Google Scholar : PubMed/NCBI | |
|
Huestis MP, Dela Cruz D, DiPasquale AG, Durk MR, Eigenbrot C, Gibbons P, Gobbi A, Hunsaker TL, La H, Leung DH, et al: Targeting KRAS mutant cancers via combination treatment: Discovery of a 5-Fluoro-4-(3H)-quinazolinone Aryl urea pan-RAF kinase inhibitor. J Med Chem. 64:3940–3955. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Rezaeinasab R, Jafari E and Khodarahmi G: Quinazolinone-based hybrids with diverse biological activities: A mini-review. J Res Med Sci. 27:682022. View Article : Google Scholar : PubMed/NCBI | |
|
Lv L, Maimaitiming M, Xia S, Yang J, Zhang T, Wang Y, Li X, Pinchuk I, Wang P, Wang CY and Liu Z: MR2938 relieves DSS-induced colitis in mice through inhibiting NF-κB signaling and improving epithelial barrier. Mar Life Sci Technol. 18: View Article : Google Scholar : 2025. | |
|
Upadhyay R, Tandel P and Patel AB: Halogen-based quinazolin-4(3H)-one derivatives as MCF-7 breast cancer inhibitors: Current developments and structure-activity relationship. Arch Pharm (Weinheim). 358:e24007402024. View Article : Google Scholar : PubMed/NCBI | |
|
Wahan SK, Sharma B and Chawla PA: Medicinal perspective of quinazolinone derivatives: Recent developments and structure-activity relationship studies. Curr Top Med Chem. 59:239–257. 2021. | |
|
Kumar P, Tomar V, Joshi RK and Nemiwal M: Nanocatalyzed synthetic approach for quinazoline and quinazolinone derivatives: A review (2015-present). Synth Commun. 52:795–826. 2022. View Article : Google Scholar | |
|
Chen Y, Sun SN, Chen XH, Chen ML, Lin JM, Niu Q, Li SL, Liu J and Lan YQ: Predesign of covalent-organic frameworks for efficient photocatalytic dehydrogenative cross-coupling reaction. Adv Mater. 37:e24136382025. View Article : Google Scholar | |
|
Huang FP, Qin WJ, Pan XY, Yang K, Wang K and Teng QH: Visible-Light-Induced chemodivergent synthesis of tetracyclic quinazolinones and 3-iminoisoindoliones via the substrate control strategy. J Org Chem. 89:4395–4405. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Kavitha K, Srinivasan N and Harbabu Y: Review of quinazolinone scaffold as anticancer agents. World J Pharm Res. 7:434–454. 2018. | |
|
Wang Q, Pan Y, Luo H, Zhang Y, Gao F, Wang J and Zheng J: Novel approaches for the solid-phase synthesis of dihydroquinazoline-2(1H)-one derivatives and biological evaluation as potential anticancer agents. Molecules. 27:85772022. View Article : Google Scholar : PubMed/NCBI | |
|
Aljohani AKB, Almadani SA, Alsulaimany M, Aljuhani N, Samman WA, Al-Shareef AH, Alghamdi R, Tayeb SM, Alharbi HY, Aljohani MS, et al: Nano-carrier, design, synthesis, in silico ADMET, anti-proliferative assessments and docking of [1,2,4]triazolo[4,3-a]quinoxalines as Topo-II inhibitors and DNA intercalators. Naunyn Schmiedebergs Arch Pharmacol. 11: View Article : Google Scholar : 2025. | |
|
Udayasree N, Haridasyam RB, Palabindela R, Krishna TM and Narsimha S: One-pot synthesis, anticancer, EGFR and caspases assays of novel fused [1,2,3]triazolo-pyrrolo[2,1-b]quinazolinones. J Mol Struct. 1320:1395702025. View Article : Google Scholar | |
|
Zhu MS, Zhang G, Xu YJ, Sun R and Ge JF: Conjugated structures based on quinazolinones and their application in fluorescent labeling. Org Biomol Chem. 21:1992–2000. 2023. View Article : Google Scholar : PubMed/NCBI |