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.
Lung cancer (LC) is among the most prevalent and lethal malignancies worldwide, ranking as the leading cause of cancer-related mortality (1). Early-stage LC is often asymptomatic and difficult to detect, resulting in delayed diagnosis. For patients diagnosed at advanced stages, the five-year survival rate remains dismally low at ~6% (2). Globally, ~1.6 million new LC cases are reported annually (3). In men, LC constitutes the primary cause of both cancer incidence and mortality, whereas among women, its incidence ranks second only to breast and colorectal cancers (4). LC poses a significant public health burden, with major risk factors including exogenous environmental exposures such as tobacco exposure, occupational and environmental carcinogens, and air pollution, as well as endogenous biological factors such as genetic predisposition, chronic lung disease, and hormonal and metabolic factors (5,6). Despite substantial advancements in diagnostic and therapeutic modalities in recent years, the overall prognosis of LC remains poor, particularly for patients with advanced disease (7,8).
Chemoprevention, defined as the use of natural or synthetic agents to prevent, inhibit, delay, or reverse carcinogenesis, has attracted considerable attention as a strategy to combat LC (9). Although various candidate chemo-preventive agents such as vitamin E, isotretinoin and aspirin have been evaluated, none have demonstrated definitive clinical efficacy (10). Natural bioactive compounds, characterized by diverse pharmacological activities and relatively low toxicity, have emerged as promising candidates in cancer prevention and treatment, thereby becoming a central focus of chemoprevention research (11). Ursolic acid (UA; PubChem Compound ID: 64945) is a pentacyclic triterpenoid compound (C30H48O3) widely distributed in various plants, including calendula, lavender, oregano, Melaleuca, sage, apple, rosemary and pear. It has garnered significant scientific interest due to its diverse biological activities (12,13). Previous studies have revealed that UA possesses multifaceted biological activities, including anti-inflammatory (14), antioxidant (15), and anti-obesity effects (16). Additionally, UA exhibits therapeutic potential in a variety of conditions, including diabetes (17), liver diseases (18), cardiovascular diseases (19), gastrointestinal disorders (20) and neurodegenerative diseases (21). Of particular note is its remarkable anticancer potential (22). UA has been demonstrated to inhibit tumor cell proliferation, induce apoptosis, and suppress tumor invasion and metastasis across multiple cancer types, including breast (23), pancreatic (24), prostate (25), colorectal (26), cervical (27) and renal cancers (28).
In LC chemoprevention, UA exerts its effects through multiple molecular mechanisms. Although extensive preclinical evidence supports the potential role of UA in LC prevention, the precise molecular pathways remain to be fully elucidated. Moreover, challenges related to the bioavailability, safety, and clinical efficacy of UA must be addressed to facilitate its translational application. The present review aims to comprehensively summarize the chemo-preventive effects and molecular mechanisms of UA in LC, thereby providing new insights and a theoretical framework for its future application in LC prevention and therapy.
UA is a pentacyclic triterpenoid compound that naturally occurs either as triterpene saponins or in its free acid form (29,30). It is also referred to as urson, malol, prunol, or 3β-3-hydroxy-urs-12-en-28-oic acid (31). UA has the molecular formula C30H48O3, a molecular weight of 456.68 g/mol, and a melting point ranging from 283–285°C (12,32). Its chemical structure comprises five rings-four six-membered and one five-membered-along with multiple hydroxyl and carboxyl functional groups. This distinctive molecular framework confers a broad spectrum of biological activities, including anti-inflammatory, antioxidant, antimicrobial and anticancer effects (33–35). Numerous studies have demonstrated that UA exerts potent anticancer effects across various malignancies (36,37). Its mechanisms of action extend beyond conventional pathways such as apoptosis induction and proliferation inhibition, encompassing unique molecular targets and biological processes. Importantly, UA selectively targets cancer stem cells (CSCs), impairing their self-renewal capacity, which is critical for tumor initiation and recurrence (38). Moreover, UA exhibits context-dependent modulation of autophagy, a process highly relevant to cancer therapy. At low concentrations, UA activates the AMP-activated protein kinase/mammalian target of rapamycin pathway, inducing protective autophagy that facilitates the clearance of damaged cellular components and enhances chemoresistance. Conversely, at high concentrations, UA inhibits autophagy by blocking autophagosome-lysosome fusion, thereby promoting autophagic cell death and augmenting its anticancer efficacy. Beyond autophagy regulation, UA exerts precise anticancer effects by modulating glycolytic metabolism (39), inducing structural modifications (40), and remodeling the tumor microenvironment (41). These multifaceted actions underscore UA as a promising candidate for cancer therapy.
To improve the pharmacological activity and therapeutic potential of UA, various derivatives have been synthesized through chemical modifications targeting its hydroxyl, carboxyl and cyclic backbone moieties. Most reported UA derivatives fall into two principal categories: modifications at C-3/C-28, C-11, C-17 and C-28 positions, and structural alterations involving C-2/C-3 and the A-ring (13,42). Representative examples include UA-piperazine-dithio-carbamate ruthenium (II) polypyridyl complexes, which induce antitumor activity via necrosis (43); corosolic acid (CA), a natural UA derivative that inhibits cancer cell proliferation through β-catenin downregulation (44); UA232, which promotes apoptosis by inducing cell cycle arrest and endoplasmic reticulum (ER) stress (45); and compound 17, a UA-derived small molecule that triggers cancer cell death through macropinocytosis (46). Collectively, these findings suggest that UA derivatives exert anticancer effects through multi-target and multi-pathway mechanisms, enhancing the pharmacological potency and drug-like properties of UA. This not only reinforces its therapeutic efficacy but also offers novel insights for the development of innovative anticancer agents.
Apoptosis plays a pivotal role in tumor development, progression and metastasis, yet cancer cells often evade apoptotic mechanisms to sustain survival (47). Induction of apoptosis facilitates the elimination of potentially harmful or precancerous cells, whereas inhibition of apoptotic pathways contributes to uncontrolled cell proliferation and malignant transformation (48). UA primarily induces apoptosis in LC cells through mitochondria-dependent (MD) pathways, ER stress pathways, and modulation of associated signaling cascades (Fig. 1).
Multiple studies have demonstrated that UA inhibits the proliferation of non-small cell LC (NSCLC) cells in a dose- and time-dependent manner while promoting apoptosis. UA activates caspase-3 and caspase-9, downregulates the anti-apoptotic protein Bcl-2, and upregulates the pro-apoptotic protein Bax, collectively inducing apoptotic cell death in LC cells (49). Matrix metalloproteinases (MMPs), a family of zinc-dependent endopeptidases, mediate extracellular matrix (ECM) remodeling, which is implicated in various pathological processes (50). Notably, upregulation of MMP gene expression has been associated with apoptosis induction in NSCLC cells. Lai et al (51) reported that treatment of H460 cells with UA for 24 h led to increased expression of MMP-1, −2, −3, −9, and −10, concomitant with activation of caspase-3, nuclear morphological changes, and DNA fragmentation, thereby promoting apoptosis. Additionally, Gou et al (45) demonstrated that the UA derivative UA232 induces apoptosis in A549 and H460 cells by causing G0/G1 phase cell cycle arrest and upregulating C/EBP-homologous protein, which triggers apoptosis via the ER stress pathway. Consistently, UA has been shown to exert anticancer effects in both A549 and H460 cells by inducing G0/G1 arrest and apoptosis, thereby suppressing tumor growth and progression (52).
Mitochondria are essential organelles involved in regulating cellular functions such as autophagy and apoptosis under stress conditions (53). Chen et al (54) observed that UA increases the expression of apoptosis-inducing factor and endonuclease G in NCI-H292 cells, promoting their release via the MD pathway and triggering apoptosis. To address UA's limited solubility and lack of tumor specificity, Ma et al (55) developed a hyaluronic acid-liposome UA delivery system (HA-Lipo/UA). Their results showed that HA-Lipo/UA upregulated apoptosis-related protein in the TGF-β signaling pathway and p53 expression in A549 cells, activated caspase-3, and enhanced mitochondrial apoptosis, suggesting a promising strategy for targeted LC therapy.
Mitophagy, a selective autophagic process that degrades damaged mitochondria via lysosomal pathways, is another mechanism by which UA exerts anticancer effects. Castrejon-Jiménez et al (56) reported that UA induces mitophagy in A549 cells through a Parkin-independent mechanism, leading to p62 overexpression and subsequent apoptosis. Furthermore, Song et al (57) demonstrated that UA significantly enhances radiosensitivity in NSCLC cells, especially in radiation-resistant cells overexpressing hypoxia-inducible factor 1 alpha (HIF-1α). By reducing the glutathione ratio and downregulating HIF-1α in H1299/M-HIF-1α cells, UA increases radiosensitivity and promotes radiation-induced cell death.
DNA damage repair (DDR) is critical for maintaining genomic stability (58). A strong correlation exists between DNA damage and the initiation of cell death pathways (59). When DNA repair mechanisms are compromised, apoptosis acts as a safeguard to eliminate damaged cells (60). Vaccinia-related kinase 1 (VRK1), a mitotic kinase frequently overexpressed in lung adenocarcinoma, belongs to the nuclear serine-threonine chromatin kinase family (61). UA has been shown to bind the catalytic domain of VRK1, inhibiting its kinase activity. This interference impairs VRK1-mediated 53BP1 foci formation, thereby disrupting DDR and contributing to the therapeutic efficacy of UA against LC (62).
Tumor metastasis is a major contributor to the high morbidity and mortality associated with cancer (63). Dysregulation of cancer cell migration critically influences the ability of tumor cells to detach from the primary site and invade surrounding tissues, thereby facilitating metastasis (63).
Proteases play a vital role in cancer progression not only by mediating ECM remodeling but also by regulating the bioavailability of growth factors, pro-angiogenic factors, and cytokines, which collectively promote tumor development through both direct and indirect mechanisms (64). However, the clinical application of UA is limited by its poor water solubility and insufficient tumor-targeting capability (65). To address these challenges, Xu et al (66) developed a novel multifunctional nanoparticle formulation [HA-modified UA and astragaloside IV (AS-IV)-loaded polydopamine nanoparticles (NPs; UA/(AS-IV)@PDA-HA)]. This delivery system significantly enhanced the cytotoxicity of UA and improved its anti-metastatic efficacy in NSCLC cells. Furthermore, UA has been shown to inhibit LC cell invasion in a concentration-dependent manner, with notable suppression of cell migration observed at concentrations between 4 and 16 µmol/l (67).
Epithelial-mesenchymal transition (EMT) is a fundamental cellular program involved in embryonic development, tissue repair and stem cell plasticity, but it also contributes pathologically to fibrosis and cancer metastasis by enhancing cellular motility and invasiveness (68). Experimental studies demonstrated that UA reduces EMT induction in H1975 LC cells triggered by transforming growth factor-β1, thereby inhibiting metastatic potential (69). Additionally, investigations in human NSCLC A549 cells revealed that UA suppresses EMT by inhibiting the nuclear factor Kappa-light-chain-enhancer of activated B Cells (NF-κB) signaling pathway, downregulating astrocyte-elevated gene-1 (AEG-1), and modulating key EMT markers-upregulating epithelial marker E-cadherin while downregulating mesenchymal markers N-cadherin and vimentin. This mechanism effectively curtails tumor cell migration and invasion, underscoring the promise of UA as an anti-metastatic agent in LC treatment (Fig. 2) (70).
Cell proliferation is fundamental to the normal development and homeostasis of multicellular organisms. Dysregulated cell proliferation is a hallmark of tumorigenesis and can also contribute to congenital malformations (71).
Cancer/Testis Antigen Family 45 Member A2 (CT45A2) is implicated as an oncogene in NSCLC and is associated with Epidermal Growth Factor Receptor (EGFR) T790 mutations. The antitumor effects of UA in NSCLC predominantly depend on CT45A2 expression in H1975 cells. UA inhibits CT45A2 transcription by targeting the β-catenin/transcription factor 4 (TCF4) signaling pathway. Specifically, UA negatively regulates the β-catenin/TCF4/CT45A2 axis, thereby suppressing the proliferation of H1975 cells (72). Autophagy-related gene 5 (ATG5) plays a pivotal role in autophagy by participating in the formation of autophagic complexes and autophagosomes, thus amplifying the autophagic response and offering potential therapeutic avenues (73,74). Mutations in ATG5 have been linked to various pathologies. Notably, inhibition of ATG5 via chloroquine (CQ) or siRNA enhances UA's anti-proliferative effects by suppressing autophagy, suggesting that autophagy may serve as a protective mechanism in cancer cells under UA treatment (75).
Cell proliferation involves the biosynthesis of macromolecules such as proteins, lipids and nucleic acids, as well as an increase in cell size and mass. Aberrant regulation of cell proliferation and division contribute to tumor development. Wu et al (76) demonstrated that UA suppresses NSCLC cell proliferation by inducing phosphorylation of stress-activated protein kinase/c-Jun N-terminal kinase and subsequently downregulating Sp1 expression. Furthermore, Way et al (49) reported that UA inhibits LC cell proliferation through modulation of key signaling proteins, including mTOR and AMPK. Their findings indicate that UA activates AMPK in a dose- and time-dependent manner, which in turn suppresses mTOR activity-a central regulator of protein synthesis and cellular proliferation (Fig. 3).
Chemotherapy remains a cornerstone in the treatment of LC (77,78). However, the emergence of drug resistance constitutes a significant barrier to successful chemotherapy outcomes (79). Cisplatin resistance, in particular, is a major cause of therapeutic failure in patients with LC (80). Recently, UA has attracted increasing attention for its potential to overcome cisplatin resistance in LC (Fig. 4).
Fan et al (81) utilized the A549 human LC cell line as a model for cisplatin-resistant cells and demonstrated that UA treatment inhibited the activation of the Janus kinase 2/signal transducer and activator of transcription 3 (Jak2/Stat3) signaling pathway. This inhibition led to downregulation of pluripotency-associated transcription factors and suppression of CSC enrichment, thereby attenuating chemoresistance.
To improve UA delivery and targeting, Li et al (82) developed a hydrogel-based drug delivery system co-loaded with UA and cisplatin. This platform enhances the targeting of UA to LC cells by facilitating its binding to the LC-specific membrane protein TMEM16A, effectively inhibiting lung adenocarcinoma progression. The hydrogel system thus provides a promising strategy to potentiate UA's therapeutic efficacy in resistant LC.
Furthermore, Chen et al (83) identified the miRNA-149-5p/MyD88 signaling axis as a critical mediator of chemoresistance in NSCLC cells. Their study revealed that UA suppresses this signaling pathway, thereby reducing cancer stemness, overcoming chemotherapy resistance, and ultimately enhancing treatment efficacy in LC.
Currently, most anticancer drugs have the drawback of poor targeting, which can cause irreversible damage to the body during treatment. New drug delivery systems aimed at ensuring drug safety and efficient therapy may become key to enhancing the efficacy of anticancer drugs (84). Research indicates that nanoparticle-based drug delivery systems (Nano-DDS) could potentially resolve the challenges associated with drug administration in cancer therapy (85).
In an experiment by Wu et al (79), a biomimetic red blood cell membrane (RBCM) nano-carrier, UA-loaded NPs coated with RBCM, was developed. This novel biomimetic drug delivery platform enhanced the tumor targeting, stability and biocompatibility of the UA NPs. It addressed the clinical limitations of UA and improved its bioavailability. This innovative biomimetic delivery system significantly improved the anticancer efficacy in advanced NSCLC model, offering a promising strategy for enhancing UA-based treatments. In addition, Xu et al (66) developed a HA-modified UA/AS-IV-loaded polydopamine (PDA) nanomedicine (UA/(AS-IV)@PDA-HA). This nanomedicine effectively enhanced the water solubility and targeting ability of UA. It allows UA to specifically bind to the overexpressed cluster of differentiation 44 on the surface of NSCLC cells. Moreover, the modified nanomedicine significantly improved the cytotoxicity mediated by UA and enhanced its ability to inhibit NSCLC cell proliferation and metastasis. These findings suggest that UA has great potential as an effective targeted anticancer drug for LC therapy. Novel drug delivery systems have greatly enhanced the ability of UA to target LC, enabling precise control that reduces systemic toxicity and improves anticancer efficacy. However, the integration and interaction between targeted therapies and UA in addiction-related oncogenes of NSCLC remain unclear and require further investigation.
According to the Biopharmaceutics Classification System (BCS), UA is classified as a Class IV drug, characterized by low solubility and low permeability. Although UA demonstrates unique advantages in the treatment of cancers such as LC, its pharmacological efficacy is limited. Due to its lipophilic nature, UA exhibits poor oral bioavailability and has difficulty penetrating biological membranes, thereby reducing its therapeutic effectiveness against tumors (86). To overcome these challenges, Antonio et al (87) developed chitosan (CS)-modified polylactic acid NPs encapsulating UA. Their study showed that this nanoparticle system reduced the average particle size and promoted the amorphous transformation of UA, enabling sustained release. This significantly reduced cytotoxicity in tumor cells and enhanced UA's oral absorption, clearance rate and metabolic efficiency in rat models. Furthermore, Yang et al (88) demonstrated that amorphous UA NPs prepared using the supercritical antisolvent technique effectively improved UA's supersaturation, solubility and absorption. In another study, Yu et al (89) formulated a supramolecular co-amorphous system combining UA and piperine. Through differential scanning calorimetry and scanning electron microscopy, it was confirmed that the co-amorphous system significantly enhanced UA's oral bioavailability and solubility in physiological media compared to its crystalline form.
Regarding drug absorption, high-performance liquid chromatography-mass spectrometry was used to analyze the tissue distribution of UA in rats 1 h after oral administration. The study revealed that UA concentrations were highest in the lungs, followed by the spleen, liver, brain, heart and kidneys, in descending order (90). Additionally, Wang et al (91) found that a novel CS-coated UA liposome achieved significantly higher accumulation at tumor sites in mice compared with conventional UA liposomes and free UA. These findings offer new insights and strategies for enhancing UA's therapeutic efficacy against LC.
In terms of drug metabolism and excretion, research on UA remains relatively limited. Available studies indicate that UA is primarily metabolized in the human body by the hepatic cytochrome P450 (CYP450) enzyme system, along with phase II conjugation reactions. In Caco-2 cells, CYP3A4 and CYP2C9 play key roles in UA metabolism, and the PXR-RXRα pathway has been shown to significantly upregulate CYP2C9 expression, thereby promoting UA biotransformation (92).
In summary, although UA shows promising pharmacokinetic behavior, further studies are needed to fully elucidate its absorption, metabolism and elimination profiles, which will be essential for advancing its clinical application.
LC, a malignant tumor originating from lung tissue, is one of the most common cancers worldwide and a leading cause of cancer-related mortality (93,94). UA plays a significant role in the treatment of LC, with mechanisms that include inhibiting cell proliferation, invasion and migration, and reducing drug resistance. These effects involve signaling pathways such as β-catenin, β-catenin/TCF4/CT45A2, NF-κB, Jak2/Stat3, and the regulation of related genes such as ATG5, CT45A2, MMP and AEG-1, as well as the expression of proteins such as Bcl-2, Bax, upregulation of E-cadherin, downregulation of N-cadherin, vimentin, mTOR and sp1, alongside the involvement of proteases and kinases such as caspase-3, caspase-9, urokinase, cathepsin B, AMPK and VRK1 (Table I).
Due to the limited water solubility, poor targeting ability, and drug resistance of UA (95), the development of novel DDS, including Nano-DDS and hydrogel-based delivery systems, has greatly addressed these issues. These innovations significantly enhance the delivery efficiency, targeting ability and bioavailability of UA, improving the effectiveness of monotherapy and boosting the antitumor effect through synergistic strategies. Furthermore, Ram Kumar Pandian et al (96) found that polyhydroxybutyrate NPs could deliver UA, enhancing its stability and bioactivity, thus improving the treatment of tumors. In another study by Sharma et al (97), HA-UA conjugates were used to prepare PTX-loaded HA-UA NPs, which significantly inhibited tumor cell proliferation in experimental models, offering new ideas and methods for combined chemotherapy delivery. If these two delivery systems are applied to the treatment of LC, they might yield unforeseen effects and become a new direction for future research. In addition, smoking as an important factor affecting LC is detrimental to the whole process of carcinogenesis. It has been identified that polycyclic aromatic hydrocarbons in cigarettes can affect the metabolism of drugs by key drug-metabolizing enzymes of cytochrome P450 and isoforms of the glucuronosyltransferase family, shortening the duration of drug action by accelerating the metabolism of related drugs and thus affecting therapeutic efficacy (98). In addition, Zevin et al (99) found that smoking significantly upregulated CYP2E1 activity, increasing the rate of metabolizing drugs while activating carcinogens and increasing the risk of disease. Whether smoking affects the metabolism and utilization of natural active ingredients, such as UA, has not been confirmed by relevant experiments, and the authors will further explore this issue in future studies.
Although UA shows remarkable therapeutic potential in LC treatment, there are still some limitations: First, low bioavailability: Its poor water solubility results in low absorption and bioavailability in the body. Second, lack of clinical trial data: Most studies on UA in LC remain at the laboratory stage, lacking large-scale clinical trials to support its safety and efficacy. In particular, clinical evidence for UA combination chemotherapy in non-small cell carcinoma remains low and a distant dream. Third, Dose and administration challenges: In clinical application, determining the appropriate dose is crucial. A very low dose may result in ineffective treatment, while a very high dose could cause toxicity. Further experimental research is needed to optimize dosing and administration methods. Fourth, drug interactions: UA may interact with other drugs, influencing their metabolism and therapeutic effects. Identifying interactions with other drugs is one of the future research directions. Fifth, side effects and toxicity: While UA is considered a relatively safe natural compound, high doses or prolonged use may still produce toxicity and side effects, which need further investigation. Sixth, individual differences: Patients may have different responses to UA based on factors such as genetic background, tumor type, stage and overall health. Hence, it is essential to avoid generalizing treatment approaches.
In conclusion, while UA has made significant progress in non-clinical research, some issues and limitations remain. In the future, the authors will focus research on the cultivation of real animal cells, to deepen the analysis of their multi-target regulatory networks and develop efficient delivery systems, to promote the clinical translation of UA and its derivatives, and ultimately to realize the leap from cellular evidence to human disease intervention (Fig. 5).
Not applicable.
The present study was supported by the Youth Program of the Shandong Provincial Natural Science Foundation (grant no. ZR2023QH079).
Not applicable.
ZL wrote the manuscript and drew the pictures. QC and ZC collected and organized literature. TP and JB proofread the manuscript. FM are fully responsible for the study designing, research fields, drafting, and finalizing the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.
Not applicable.
Not applicable.
The authors declare that they have no competing interests.
|
Frydrychowicz M, Kuszel Ł, Dworacki G and Budna-Tukan J: MicroRNA in lung cancer-a novel potential way for early diagnosis and therapy. J Appl Genet. 64:459–477. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Juang YR, Ang L and Seow WJ: Predictive performance of risk prediction models for lung cancer incidence in Western and Asian countries: A systematic review and meta-analysis. Sci Rep. 15:42592025. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang M and Ma C: LSR promotes cell proliferation and invasion in lung cancer. Comput Math Methods Med. 2021:66519072021.PubMed/NCBI | |
|
Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI | |
|
Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021.PubMed/NCBI | |
|
Pallis AG and Syrigos KN: Lung cancer in never smokers: Disease characteristics and risk factors. Crit Rev Oncol Hematol. 88:494–503. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Jones GS and Baldwin DR: Recent advances in the management of lung cancer. Clin Med (Lond). 18 (Suppl 2):S41–S46. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Lim RB: End-of-life care in patients with advanced lung cancer. Ther Adv Respir Dis. 10:455–467. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Koh YC, Ho CT and Pan MH: Recent advances in cancer chemoprevention with phytochemicals. J Food Drug Anal. 28:14–37. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Bade BC and Dela Cruz CS: Lung cancer 2020: Epidemiology, etiology, and prevention. Clin Chest Med. 41:1–24. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Similie D, Minda D, Bora L, Kroškins V, Lugiņina J, Turks M, Dehelean CA and Danciu C: An update on pentacyclic triterpenoids ursolic and oleanolic acids and related derivatives as anticancer candidates. Antioxidants (Basel). 13:9522024. View Article : Google Scholar : PubMed/NCBI | |
|
Pironi AM, de Araújo PR, Fernandes MA, Salgado HRN and Chorilli M: Characteristics, biological properties and analytical methods of ursolic acid: A review. Crit Rev Anal Chem. 48:86–93. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Mlala S, Oyedeji AO, Gondwe M and Oyedeji OO: Ursolic acid and its derivatives as bioactive agents. Molecules. 24:27512019. View Article : Google Scholar : PubMed/NCBI | |
|
Kashyap D, Sharma A, Tuli HS, Punia S and Sharma AK: Ursolic acid and oleanolic acid: Pentacyclic terpenoids with promising Anti-inflammatory activities. Recent Pat Inflamm Allergy Drug Discov. 10:21–33. 2016. View Article : Google Scholar : PubMed/NCBI | |
|
Liobikas J, Majiene D, Trumbeckaite S, Kursvietiene L, Masteikova R, Kopustinskiene DM, Savickas A and Bernatoniene J: Uncoupling and antioxidant effects of ursolic acid in isolated rat heart mitochondria. J Nat Prod. 74:1640–1644. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Jayaprakasam B, Olson LK, Schutzki RE, Tai MH and Nair MG: Amelioration of obesity and glucose intolerance in high-fat-fed C57BL/6 mice by anthocyanins and ursolic acid in Cornelian cherry (Cornus mas). J Agric Food Chem. 54:243–248. 2006. View Article : Google Scholar : PubMed/NCBI | |
|
Yu SG, Zhang CJ, Xu XE, Sun JH, Zhang L and Yu PF: Ursolic acid derivative ameliorates Streptozotocin-induced diabestic bone deleterious effects in mice. Int J Clin Exp Pathol. 8:3681–3690. 2015.PubMed/NCBI | |
|
Sundaresan A, Radhiga T and Pugalendi KV: Effect of ursolic acid and Rosiglitazone combination on hepatic lipid accumulation in high fat diet-fed C57BL/6J mice. Eur J Pharmacol. 741:297–303. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Erdmann J, Kujaciński M and Wiciński M: Beneficial effects of ursolic acid and its derivatives-focus on potential biochemical mechanisms in cardiovascular conditions. Nutrients. 13:39002021. View Article : Google Scholar : PubMed/NCBI | |
|
Shi Y, Leng Y, Liu D, Liu X, Ren Y, Zhang J and Chen F: Research advances in protective effects of ursolic acid and oleanolic acid against gastrointestinal diseases. Am J Chin Med. 49:413–435. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Chen C, Ai Q, Shi A, Wang N, Wang L and Wei Y: Oleanolic acid and ursolic acid: Therapeutic potential in neurodegenerative diseases, neuropsychiatric diseases and other brain disorders. Nutr Neurosci. 26:414–428. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Sandhu SS, Rouz SK, Kumar S, Swamy N, Deshmukh L, Hussain A, Haque S and Tuli HS: Ursolic acid: A pentacyclic triterpenoid that exhibits anticancer therapeutic potential by modulating multiple oncogenic targets. Biotechnol Genet Eng Rev. 39:729–759. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Iqbal J, Abbasi BA, Ahmad R, Mahmood T, Kanwal S, Ali B, Khalil AT, Shah SA, Alam MM and Badshahet H: Ursolic acid a promising candidate in the therapeutics of breast cancer: Current status and future implications. Biomed Pharmacother. 108:752–756. 2018. View Article : Google Scholar : PubMed/NCBI | |
|
Lin JH, Xu JJ, Liu XF, Liu WH, Wang YF, Li ZS, Wang LW and Wang W: Ursolic acid promotes apoptosis, autophagy, and chemosensitivity in gemcitabine-resistant human pancreatic cancer cells. Phytother Res. 34:2053–2066. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Meng Y, Lin ZM, Ge N, Zhang DL, Huang J and Kong F: Ursolic acid induces apoptosis of prostate cancer cells via the PI3K/Akt/mTOR pathway. Am J Chin Med. 43:1471–1486. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang H, Zhang X, Ma X and Wang X: Ursolic acid in colorectal cancer: Mechanisms, current status, challenges, and future research directions. Pharmacol Rep. 77:72–86. 2025. View Article : Google Scholar : PubMed/NCBI | |
|
Guo JL, Han T, Bao L, Li XM, Ma JQ and Tang LP: Ursolic acid promotes the apoptosis of cervical cancer cells by regulating endoplasmic reticulum stress. J Obstet Gynaecol Res. 45:877–881. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Cai C, Zhi Y, Xie C, Geng S, Sun F, Ji Z, Zhang P, Wang H and Tang J: Ursolic acid-downregulated long noncoding RNA ASMTL-AS1 inhibits renal cell carcinoma growth via binding to HuR and reducing vascular endothelial growth factor expression. J Biochem Mol Toxicol. 37:e233892023. View Article : Google Scholar : PubMed/NCBI | |
|
Namdeo P, Gidwani B, Tiwari S, Jain V, Joshi V, Shukla SS, Pandey RK and Vyas A: Therapeutic potential and novel formulations of ursolic acid and its derivatives: An updated review. J Sci Food Agric. 103:4275–4292. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Ma X, Bai Y, Liu K, Han Y, Zhang J, Liu Y, Hou X, Hao E, Hou Y and Bai G: Ursolic acid inhibits the cholesterol biosynthesis and alleviates high fat Diet-induced hypercholesterolemia via irreversible inhibition of HMGCS1 in vivo. Phytomedicine. 103:1542332022. View Article : Google Scholar : PubMed/NCBI | |
|
Fernández-Hernández A, Martinez A, Rivas F, García-Mesa JA and Parra A: Effect of the solvent and the sample preparation on the determination of triterpene compounds in two-phase olive-mill-waste samples. J Agric Food Chem. 63:4269–4275. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J: Pharmacology of oleanolic acid and ursolic acid. J Ethnopharmacol. 49:57–68. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Ikeda Y, Murakami A and Ohigashi H: Ursolic acid: An anti- and pro-inflammatory triterpenoid. Mol Nutr Food Res. 52:26–42. 2008. View Article : Google Scholar : PubMed/NCBI | |
|
Wang X, Xiong W, Wang X, Qin L, Zhong M, Liu Y, Xiong Y, Yi X, Wang X and Zhang H: Ursolic acid attenuates cholestasis through NRF2-mediated regulation of UGT2B7 and BSEP/MRP2. Naunyn Schmiedebergs Arch Pharmacol. 397:2257–2267. 2024. View Article : Google Scholar : PubMed/NCBI | |
|
Luan M, Wang H, Wang J, Zhang X, Zhao F, Liu Z and Meng Q: Advances in Anti-inflammatory activity, mechanism and therapeutic application of ursolic acid. Mini Rev Med Chem. 22:422–436. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Shanmugam MK, Dai X, Kumar AP, Tan BK, Sethi G and Bishayee A: Ursolic acid in cancer prevention and treatment: Molecular targets, pharmacokinetics and clinical studies. Biochem Pharmacol. 85:1579–1587. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Khwaza V, Oyedeji OO and Aderibigbe BA: Ursolic Acid-based derivatives as potential anti-cancer agents: An update. Int J Mol Sci. 21:59202020. View Article : Google Scholar : PubMed/NCBI | |
|
Kang DY, Sp N, Jang KJ, Jo ES, Bae SW and Yang YM: Antitumor effects of natural bioactive ursolic acid in embryonic cancer stem cells. J Oncol. 2022:67372482022. View Article : Google Scholar : PubMed/NCBI | |
|
Wang S, Chang X, Zhang J, Li J, Wang N, Yang B, Pan B, Zheng Y, Wang X, Ou H and Wang Z: Ursolic acid inhibits breast cancer metastasis by suppressing glycolytic metabolism via activating SP1/Caveolin-1 signaling. Front Oncol. 11:7455842021. View Article : Google Scholar : PubMed/NCBI | |
|
Panda SS, Thangaraju M and Lokeshwar BL: Ursolic acid analogs as potential therapeutics for cancer. Molecules. 27:89812022. View Article : Google Scholar : PubMed/NCBI | |
|
Zhang N, Liu S, Shi S, Chen Y, Xu F, Wei X and Xu Y: Solubilization and delivery of Ursolic-acid for modulating tumor microenvironment and regulatory T cell activities in cancer immunotherapy. J Control Release. 320:168–178. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen H, Gao Y, Wang A, Zhou X, Zheng Y and Zhou J: Evolution in medicinal chemistry of ursolic acid derivatives as anticancer agents. Eur J Med Chem. 92:648–655. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Jiang H, Wei JH, Lin CY, Liang GB, He RJ, Huang RZ, Ma XL, Huang GB and Zhang Y: Ursolic acid-piperazine-dithiocarbamate ruthenium(II) polypyridyl complexes induced necroptosis in MGC-803 cells. Metallomics. 14:mfac0722022. View Article : Google Scholar : PubMed/NCBI | |
|
Kim JH, Kim YH, Song GY, Kim DE, Jeong YJ, Liu KH, Chung YH and Oh S: Ursolic acid and its natural derivative corosolic acid suppress the proliferation of APC-mutated colon cancer cells through promotion of beta-catenin degradation. Food Chem Toxicol. 67:87–95. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Gou W, Luo N, Wei H, Wu H, Yu X, Duan Y, Bi C, Ning H, Hou W and Li Y: Ursolic acid derivative UA232 evokes apoptosis of lung cancer cells induced by endoplasmic reticulum stress. Pharm Biol. 58:707–715. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Sun L, Li B, Su X, Chen G, Li Y, Yu L, Li L and Wei W: An ursolic acid derived small molecule triggers cancer cell death through hyperstimulation of macropinocytosis. J Med Chem. 60:6638–6648. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Liu X, Qi M, Li X, Wang J and Wang M: Curcumin: A natural organic component that plays a Multi-faceted role in ovarian cancer. J Ovarian Res. 16:472023. View Article : Google Scholar : PubMed/NCBI | |
|
Nedopekina DA, Gubaidullin RR, Odinokov VN, Maximchik PV, Zhivotovsky B, Bel'skii YP, Khazanov VA, Manuylova AV, Gogvadze V and Spivak AY: Mitochondria-targeted betulinic and ursolic acid derivatives: Synthesis and anticancer activity. Medchemcomm. 8:1934–1945. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Way TD, Tsai SJ, Wang CM, Ho CT and Chou CH: Chemical constituents of Rhododendron formosanum show pronounced growth inhibitory effect on non-small-cell lung carcinoma cells. J Agric Food Chem. 62:875–884. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Abdel-Hamid NM and Abass SA: Matrix metalloproteinase contribution in management of cancer proliferation, metastasis and drug targeting. Mol Biol Rep. 48:6525–6538. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Lai MY, Leung HW, Yang WH, Chen WH and Lee HZ: Up-regulation of matrix metalloproteinase family gene involvement in ursolic acid-induced human lung non-small carcinoma cell apoptosis. Anticancer Res. 27:145–153. 2007.PubMed/NCBI | |
|
Kang DY, Sp N, Lee JM and Jang KJ: Antitumor effects of ursolic acid through mediating the inhibition of STAT3/PD-L1 signaling in Non-small cell lung cancer cells. Biomedicines. 9:2972021. View Article : Google Scholar : PubMed/NCBI | |
|
Nunnari J and Suomalainen A: Mitochondria: In sickness and in health. Cell. 148:1145–1159. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Chen CJ, Shih YL, Yeh MY, Liao NC, Chung HY, Liu KL, Lee MH, Chou PY, Hou HY, Chou JS and Chung JG: Ursolic acid induces apoptotic cell death through AIF and endo G release through a Mitochondria-dependent pathway in NCI-H292 human lung cancer cells in vitro. In Vivo. 33:383–391. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Ma T, Zhou J, Li J and Chen Q: Hyaluronic Acid-modified liposomes for ursolic Acid-targeted delivery treat lung cancer based on p53/ARTS-mediated mitochondrial apoptosis. Iran J Pharm Res. 22:e1317582023. View Article : Google Scholar : PubMed/NCBI | |
|
Castrejón-Jiménez NS, Leyva-Paredes K, Baltierra-Uribe SL, Castillo-Cruz J, Campillo-Navarro M, Hernández-Pérez AD, Luna-Angulo AB, Chacón-Salinas R, Coral-Vázquez RM, Estrada-García I, et al: Ursolic and oleanolic acids induce mitophagy in A549 human lung cancer cells. Molecules. 24:34441019. View Article : Google Scholar | |
|
Song B, Zhang Q, Yu M, Qi X, Wang G, Xiao L, Yi Q and Jin W: Ursolic acid sensitizes radioresistant NSCLC cells expressing HIF-1α through reducing endogenous GSH and inhibiting HIF-1α. Oncol Lett. 13:754–762. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Smith G, Alholm Z, Coleman RL and Monk BJ: DNA damage repair Inhibitors-combination therapies. Cancer J. 27:501–505. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Wang JY: DNA damage and apoptosis. Cell Death Differ. 8:1047–1048. 2001. View Article : Google Scholar : PubMed/NCBI | |
|
Kaina B: DNA damage-triggered apoptosis: Critical role of DNA repair, double-strand breaks, cell proliferation and signaling. Biochem Pharmacol. 66:1547–1554. 2003. View Article : Google Scholar : PubMed/NCBI | |
|
Campillo-Marcos I, García-González R, Navarro-Carrasco E and Lazo PA: The human VRK1 chromatin kinase in cancer biology. Cancer Lett. 503:117–128. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Kim SH, Ryu HG, Lee J, Shin J, Harikishore A, Jung HY, Kim YS, Lyu HN, Oh E, Baek NI, et al: Ursolic acid exerts anti-cancer activity by suppressing Vaccinia-related kinase 1-mediated damage repair in lung cancer cells. Sci Rep. 5:145702015. View Article : Google Scholar : PubMed/NCBI | |
|
Han T, Kang D, Ji D, Wang X, Zhan W, Fu M, Xin HB and Wang JB: How does cancer cell metabolism affect tumor migration and invasion? Cell Adh Migr. 7:395–403. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Affara NI, Andreu P and Coussens LM: Delineating protease functions during cancer development. Methods Mol Biol. 539:1–32. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Liu J: Oleanolic acid and ursolic acid: Research perspectives. J Ethnopharmacol. 100:92–94. 2005. View Article : Google Scholar : PubMed/NCBI | |
|
Xu F, Li M, Que Z, Su M, Yao W, Zhang Y, Luo B, Li Y, Zhang Z and Tian J: Combined chemo-immuno-photothermal therapy based on ursolic acid/astragaloside IV-loaded hyaluronic acid-modified polydopamine nanomedicine inhibiting the growth and metastasis of non-small cell lung cancer. J Mater Chem B. 11:3453–3472. 2023. View Article : Google Scholar : PubMed/NCBI | |
|
Huang CY, Lin CY, Tsai CW and Yin MC: Inhibition of cell proliferation, invasion and migration by ursolic acid in human lung cancer cell lines. Toxicol In Vitro. 25:1274–1280. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Lamouille S, Xu J and Derynck R: Molecular mechanisms of epithelial-mesenchymal transition. Nat Rev Mol Cell Biol. 15:178–196. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Ruan JS, Zhou H, Yang L, Wang L, Jiang ZS, Sun H and Wang SM: Ursolic acid attenuates TGF-β1-induced epithelial-mesenchymal transition in NSCLC by targeting integrin αVβ5/MMPs signaling. Oncol Res. 27:593–600. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Liu K, Guo L, Miao L, Bao W, Yang J, Li X, Xi T and Zhao W: Ursolic acid inhibits epithelial-mesenchymal transition by suppressing the expression of astrocyte-elevated gene-1 in human nonsmall cell lung cancer A549 cells. Anticancer Drugs. 24:494–503. 2013. View Article : Google Scholar : PubMed/NCBI | |
|
Jesudason EC, Connell MG, Fernig DG, Lloyd DA and Losty PD: Cell proliferation and apoptosis in experimental lung hypoplasia. J Pediatr Surg. 35:129–133. 2000. View Article : Google Scholar : PubMed/NCBI | |
|
Yang K, Chen Y, Zhou J, Ma L, Shan Y, Cheng X, Wang Y, Zhang Z, Ji X, Chen L, et al: Ursolic acid promotes apoptosis and mediates transcriptional suppression of CT45A2 gene expression in non-small-cell lung carcinoma harbouring EGFR T790M mutations. Br J Pharmacol. 176:4609–4624. 2019. View Article : Google Scholar : PubMed/NCBI | |
|
Changotra H, Kaur S, Yadav SS, Gupta GL, Parkash J and Duseja A: ATG5: A central autophagy regulator implicated in various human diseases. Cell Biochem Funct. 40:650–667. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Baeva ME and Camara-Lemarroy C: The role of autophagy protein Atg5 in multiple sclerosis. Mult Scler Relat Disord. 79:1050292023. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M, Yu H, Wu R, Chen ZY, Hu Q, Zhang YF, Gao SH and Zhou GB: Autophagy inhibition enhances the inhibitory effects of ursolic acid on lung cancer cells. Int J Mol Med. 46:1816–1826. 2020.PubMed/NCBI | |
|
Wu J, Zhao S, Tang Q, Zheng F, Chen Y, Yang L, Yang X, Li L, Wu W and Hann SS: Activation of SAPK/JNK mediated the inhibition and reciprocal interaction of DNA methyltransferase 1 and EZH2 by ursolic acid in human lung cancer cells. J Exp Clin Cancer Res. 34:992015. View Article : Google Scholar : PubMed/NCBI | |
|
Higgins MJ and Ettinger DS: Chemotherapy for lung cancer: The state of the art in 2009. Expert Rev Anticancer Ther. 9:1365–1378. 2009. View Article : Google Scholar : PubMed/NCBI | |
|
Souhami RL: Defining the role of chemotherapy in non-small-cell lung cancer. Ann Oncol. 6:317–318. 1995. View Article : Google Scholar : PubMed/NCBI | |
|
Wu T, Yan D, Hou W, Jiang H, Wu M, Wang Y, Chen G, Tang C, Wang Y and Xu H: Biomimetic red blood cell membrane-Mediated nanodrugs loading ursolic acid for targeting NSCLC therapy. Cancers (Basel). 14:45202022. View Article : Google Scholar : PubMed/NCBI | |
|
Shen M, Xu Z, Xu W, Jiang K, Zhang F, Ding Q, Xu Z and Chen Y: Inhibition of ATM reverses EMT and decreases metastatic potential of cisplatin-resistant lung cancer cells through JAK/STAT3/PD-L1 pathway. J Exp Clin Cancer Res. 38:1492019. View Article : Google Scholar : PubMed/NCBI | |
|
Fan L, Wang X, Cheng C, Wang S, Li X, Cui J, Zhang B and Shi L: Inhibitory effect and mechanism of ursolic acid on Cisplatin-induced resistance and stemness in human lung cancer A549 cells. Evid Based Complement Alternat Med. 2023:13073232023. View Article : Google Scholar : PubMed/NCBI | |
|
Li S, Guo X, Liu H, Chen Y, Wan H, Kang X, Qin J and Guo S: Ursolic acid, an inhibitor of TMEM16A, co-loaded with cisplatin in hydrogel drug delivery system for multi-targeted therapy of lung cancer. Int J Biol Macromol. 277:1345872024. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Q, Luo J, Wu C, Lu H, Cai S, Bao C, Liu D and Kong J: The miRNA-149-5p/MyD88 axis is responsible for ursolic Acid-mediated attenuation of the stemness and chemoresistance of non-small cell lung cancer cells. Environ Toxicol. 35:561–569. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Pérez-Herrero E and Fernández-Medarde A: Advanced targeted therapies in cancer: Drug nanocarriers, the future of chemotherapy. Eur J Pharm Biopharm. 93:52–79. 2015. View Article : Google Scholar : PubMed/NCBI | |
|
Fu S, Li G, Zang W, Zhou X, Shi K and Zhai Y: Pure drug Nano-assemblies: A facile carrier-free nanoplatform for efficient cancer therapy. Acta Pharm Sin B. 12:92–106. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Jinhua W: Ursolic acid: Pharmacokinetics process in vitro and in vivo, a mini review. Arch Pharm (Weinheim). 352:e18002222019. View Article : Google Scholar : PubMed/NCBI | |
|
Antonio E, Dos Reis Antunes Junior O, Marcano RGDJV, Diedrich C, da Silva Santos J, Machado CS, Khalil NM and Mainardes RM: Chitosan modified poly (lactic acid) nanoparticles increased the ursolic acid oral bioavailability. Int J Biol Macromol. 172:133–142. 2021. View Article : Google Scholar : PubMed/NCBI | |
|
Yang L, Sun Z, Zu Y, Zhao C, Sun X, Zhang Z and Zhang L: Physicochemical properties and oral bioavailability of ursolic acid nanoparticles using supercritical anti-solvent (SAS) process. Food Chem. 132:319–325. 2012. View Article : Google Scholar : PubMed/NCBI | |
|
Yu D, Kan Z, Shan F, Zang J and Zhou J: Triple strategies to improve oral bioavailability by fabricating coamorphous forms of ursolic acid with piperine: Enhancing Water-solubility, permeability, and inhibiting cytochrome P450 isozymes. Mol Pharm. 17:4443–4462. 2020. View Article : Google Scholar : PubMed/NCBI | |
|
Chen Q, Luo S, Zhang Y and Chen Z: Development of a liquid chromatography-mass spectrometry method for the determination of ursolic acid in rat plasma and tissue: Application to the pharmacokinetic and tissue distribution study. Anal Bioanal Chem. 399:2877–2884. 2011. View Article : Google Scholar : PubMed/NCBI | |
|
Wang M, Zhao T, Liu Y, Wang Q, Xing S, Li L, Wang L, Liu L and Gao D: Ursolic acid liposomes with chitosan modification: Promising antitumor drug delivery and efficacy. Mater Sci Eng C Mater Biol Appl. 71:1231–1240. 2017. View Article : Google Scholar : PubMed/NCBI | |
|
Lei P, Li Z, Hua Q, Song P, Gao L, Zhou L and Cai Q: Ursolic acid alleviates neuroinflammation after intracerebral hemorrhage by mediating microglial pyroptosis via the NF-κB/NLRP3/GSDMD pathway. Int J Mol Sci. 24:147712023. View Article : Google Scholar : PubMed/NCBI | |
|
Sun K, Wu H, Zhu Q, Gu K, Wei H, Wang S, Li L, Wu C, Chen R, Pang Y, et al: Global landscape and trends in lifetime risks of haematologic malignancies in 185 countries: Population-based estimates from GLOBOCAN 2022. EClinicalMedicine. 83:1031932025. View Article : Google Scholar : PubMed/NCBI | |
|
Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI | |
|
Wu BY and Parks LM: Chemical studies on ursolic acid. J Am Pharm Assoc Am Pharm Assoc. 42:603–606. 1953. View Article : Google Scholar : PubMed/NCBI | |
|
Ram Kumar Pandian S, Kunjiappan S, Pavadai P, Sundarapandian V, Chandramohan V and Sundar K: Delivery of ursolic acid by polyhydroxybutyrate nanoparticles for cancer therapy: In silico and in vitro studies. Drug Res (Stuttg). 72:72–81. 2022. View Article : Google Scholar : PubMed/NCBI | |
|
Sharma R, Yadav V, Jha S, Dighe S and Jain S: Unveiling the potential of ursolic acid modified hyaluronate nanoparticles for combination drug therapy in triple negative breast cancer. Carbohydr Polym. 338:1221962024. View Article : Google Scholar : PubMed/NCBI | |
|
O'Malley M, King AN, Conte M, Ellingrod VL and Ramnath N: Effects of cigarette smoking on metabolism and effectiveness of systemic therapy for lung cancer. J Thorac Oncol. 9:917–926. 2014. View Article : Google Scholar : PubMed/NCBI | |
|
Zevin S and Benowitz NL: Drug interactions with tobacco smoking. An update. Clin Pharmacokinet. 36:425–438. 1999. View Article : Google Scholar : PubMed/NCBI |