Open Access

Antitumour activity of oleanolic acid: A systematic review and meta‑analysis

  • Authors:
    • Ying Zeng
    • Zhonglian Wang
    • Jing Zhang
    • Wei Jian
    • Qiaofen Fu
  • View Affiliations

  • Published online on: October 2, 2024     https://doi.org/10.3892/ol.2024.14715
  • Article Number: 582
  • Copyright: © Zeng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Oleanolic acid (OA), a compound known for its potent antitumour properties, has been the subject of investigations in both cell and animal models. Although OA has good biological activity, its low water solubility and bioavailability limit its therapeutic use, and therefore translating the potential of OA into the clinical oncology setting remains challenging. The present systematic review and meta‑analysis utilized evidence from animal model studies to gain insights into the antitumour mechanisms of OA to address the gap in understanding, and to provide guidance for future research directions and potential clinical applications. The guidelines outlined in the Preferred Reporting Items for Systematic Reviews and Meta‑Analyses were applied in the present study and a comprehensive search was conducted across the PubMed/MEDLINE, Web of Science, Cochrane Library and Embase databases, with a cut‑off date of June 30, 2023. The primary focus was on randomized controlled trials that used animal models to assess the antitumour effects of OA. The methodological quality appraisal was conducted using the Systematic Review Centre for Laboratory Animal Experimentation risk of bias tool, and tumour volume and weight served as the principal outcome measures. Data were analysed using the RevMan (version 5.3) and Stata SE11 software packages, with an assessment of heterogeneity conducted using the I² statistical test, sensitivity analysis conducted using the leave‑one‑out approach, and evaluation of publication bias performed using Egger's test and funnel plot analysis. The present study demonstrated a significant inhibitory effect of OA intervention on tumour growth and a decrease in tumour weight in animal models. Despite the broad spectrum of antitumour effects exhibited by OA, further investigations are warranted to optimize the dosage and administration routes of OA to maximize its efficacy in clinical cancer treatment.

Introduction

Cancer has been deemed a notable public health issue by the World Health Organization due to its prominent contributions to global morbidity and mortality (1,2). Moreover, the incidence and mortality rates of cancer are rapidly increasing worldwide. By 2040, the number of cancer cases worldwide is expected to reach 30.2 million, and the number of cancer-related deaths is expected to reach 16.3 million (3). Cancer is a heterogeneous disease and its development involves multiple biological processes and multiple factors, including environmental pollution or immune dysfunction (4,5).

At present, surgery, immunotherapy, and hormone, gene, radiation, laser and targeted therapies are the primary cancer treatment methods (68). Despite advances in cancer treatment, drug resistance remains a notable cause of relapse and poor survival in most tumor patients (9). Natural products can be used as important substrates to overcome drug resistance and improve the efficacy of cancer therapy (8). Since the late 1930s, natural products and their derivatives have been recognized as sources of antitumour drugs (10) due to properties such as preventing and delaying tumour growth (11,12). Oleanolic acid (OA; 3β-hydroxyolean-12-en-28-oic acid), as a representative natural product, has shown marked effects in this field (13).

OA is a pentacyclic triterpene compound that is widely found in the plant kingdom; it exists both as a free acid and as a triterpenoid saponin when joint with sugar chains (14). The richest source of OA is the leaves of the olive plant (Fig. 1A) (15). OA has a diverse range of biological and pharmacological activities, including liver protection (16,17), anti-inflammatory (18,19), antidiabetic (20,21), antiviral (22,23), bidirectional immunity (24) and antitumour effects (14,2527). The anticancer effects of OA have been demonstrated at the cellular level and in animal models in numerous types of cancer (Fig. 1B), including osteosarcoma, liver, lung and breast cancer (26,2844). The mechanisms underlying the antitumour activity of OA are multifaceted and include the inhibition of cellular proliferation, the promotion of apoptosis, the induction of autophagy, the modulation of cell cycle regulatory proteins, the inhibition of tumour cell migration and invasion, and the suppression of angiogenesis (4446). Furthermore, OA increases the sensitivity of tumour cells to radiation (47). OA, akin to numerous other triterpenes, exhibits a broad spectrum of pharmacological activities, coupled with low toxicity and favourable tolerance profiles (48). However, high doses or prolonged administration of OA are reported to induce hepatotoxicity (49). According to the Biopharmaceutical Classification System (BCS), OA is categorized as a BCS Class IV drug characterized by exceedingly low aqueous solubility and suboptimal intestinal permeability, which collectively constrain its absorption and bioavailability (50). With further research, the chemical synthesis of OA derivatives and novel dosage forms may markedly improve the water solubility and bioavailability, thus strengthening their antitumour effects and ensuring their biosafety (15,16).

At present, numerous studies have reported the antitumour effects of OA in cell and animal experiments (13,25,27,34,5153). However, research on the antitumour effects of OA in clinical applications is currently limited. The present systematic review aimed to evaluate the potential antitumour effects of OA by collecting and analysing data from animal model experiments to provide guidance for further clinical research on the application of OA in cancer.

Materials and methods

Search strategy

The present systematic review and meta-analysis were performed in accordance with the Preferred Reporting Project for Systematic Review and Meta-Analysis guidelines (54). The PubMed/MEDLINE, Web of Science, Cochrane Library and Embase databases were comprehensively searched up to June 30, 2023, using the following key words: ‘Oleanolic acid’, ‘cancer’, ‘tumor’ and ‘tumour’. A total of 12 studies with 190 animals were included in the present study. A specific search strategy was devised for each database based on the Population, Intervention, Condition and Outcome framework and Medical Subject Heading terms (Table SI) (55,56). The present review protocol was not registered.

Study selection
Inclusion criteria

The inclusion criteria were as follows: i) The study type was a randomized controlled trial using animals; ii) experimental subjects consisted of tumour model mice, without any specific limitations on disease models or modelling methods; iii) the interventions involved the use of OA alone; iv) the outcome indicators included tumour weight and/or tumour volume; v) the studies were published in English; and vi) any type of solid tumour was included in the present study.

Exclusion criteria

The exclusion criteria were as follows: i) The target disease was a non-malignant tumour or cancer; ii) no control group was included; iii) OA was used in combination with other drugs; iv) duplicate publications; v) observational or non-interventional studies, clinical studies, case reports, reviews, conference papers, systematic reviews, meta-analyses, editorial/letters or patent results; vi) unpublished dissertations; and vii) the full text could not be obtained or the data were incomplete.

Literature screening and data extraction

Based on the inclusion and exclusion criteria, two researchers independently screened the studies using EndNote (version X20; Clarivate plc). Data were from the included studies using Microsoft Excel (Microsoft Corporation) software and a subsequent cross-check was conducted to ensure accuracy. Any disagreements were resolved by consulting a third researcher. The following data were extracted: i) First author, year and country of journal publication; ii) animal species, weight, age, sample size and cancer type; iii) OA dosage, administration method and intervention time; and iv) tumour weight and tumour volume. In studies where the original publication presented the tumour sizes in only a graphical format, the Graph Grabber (version. 2.0.2; Quintessa Ltd.) program was used.

Risk of bias assessment

The Systematic Review Centre for Laboratory Animal Experimentation risk of bias tool (57) was used to evaluate the risk of bias of the included studies. The tool assessed 10 items across different domains, including selection bias, experimental bias, measurement bias, untracking bias and selective reporting bias (57). The analysis was independently performed by YZ and ZW. Any disagreements were resolved by consulting a third investigator. The tool offered three options: ‘Yes’ for a low bias risk, ‘No’ for a high bias risk and ‘Unclear’ when assigning bias was not possible.

Statistical methods

The data (mean ± standard deviation) from multiple intervention groups were merged in accordance with the Cochrane Handbook for Systematic Reviews of Interventions (58,59). The merged data were then analysed using RevMan (version 5.4; The Cochrane Collaboration) and Stata SE11 (StataCorp LP) software. Sensitivity and subgroup analyses were then performed to explore potential sources of heterogeneity and to assess the reliability of the results. A random-effects model was employed for the meta-analysis. P<0.05 was considered to indicate a statistically significant difference (60). The I2 statistical test was used to quantify the degree of heterogeneity among the studies. An I2 value of ≥50% was considered to indicate significant heterogeneity (58). Publication bias was assessed using Egger's test and funnel plots if ≥10 studies were included for an outcome.

Results

Literature retrieval results

The present search strategy produced a total of 3,654 articles (Fig. 2). Following the preliminary literature screen, 72 articles remained and the comprehensive review ultimately only included the results from 12 studies (27,46,47,5153,6166), with a total of 190 animals. The full texts of the 12 articles were read by two independent researchers who evaluated them according to the inclusion criteria.

Basic characteristics of the included studies

The present review encompassed 12 studies that addressed seven distinct types of cancer, with contributions from publications across six countries (Fig. 3A and B). Colorectal cancer emerged as the most prevalent form of malignancy within the present dataset, accounting for 34% of all cases. Notably, the journals from the United States and the Netherlands each contributed to 25% of the included studies.

The main characteristics of the included studies were summarised (Table I), including the following: i) First author and year; ii) species, strain, sex and age of the animals, of which BALB/c mice (n=10) were the most common, with 7 studies of female mice and 5 studies of male mice; iii) tumour types, which included colorectal (n=4), cervical (n=2), gastric (n=2), breast (n=1) and lung (n=1) cancer, hepatocellular carcinoma (n=1) and melanoma (n=1); iv) dosage of OA (range, 2–150 mg/kg); v) route of administration, including oral (n=2), intraperitoneal (n=6), intragastric (n=3) or subcutaneous (n=1) injections; vi) duration of drug intervention (range, 10–30 days); vii) type of control, with normal saline used in 50% of the included studies; and vii) country of journal publication which included the United States (n=3), Netherlands (n=3), England (n=2), Greece (n=2), United Arab Emirates (n=1) and Brazil (n=1).

Table I.

Basic characteristics of the included literature.

Table I.

Basic characteristics of the included literature.

Oleanolic acid
Animals

Type of cancerDosage groups, mg/kg (frequency)Medication time, daysRoute of administrationControl groupCountry of journal publication(Refs.)
First author, yearMouse speciesSexAge, weeksWeight, g
Li et al, 2015BALB/c athymic (nude)Male620-22Colorectal12.5 6 days (per week)16i.p.SalineGreece(51)
Tang et al, 2013KunmingFemale--Cervical carcinoma37.5 and 70 (once daily)10p.o.SalineEngland(61)
Liang et al, 2021BALB/cFemale4-Breast30 (once daily)14i.g.Corn oilUnited States(46)
Wang et al, 2019BALB/c nu/nuMale6-8-Hepatocellular carcinoma50 (once daily)14i.g.SalineUnited States(47)
Li et al, 2016BALB/c athymic (nude)Male620-22Colorectal12.5 (6 days a week)16i.p.SalineGreece(52)
Niu et al, 2018BALB/c nu/nuMale6-8-Colorectal5 (once daily)21i.p.DMSOUnited Arab Emirates(62)
Jiang et al, 2021BALB/c nudeMale520±2Cervical40 and 80 (once daily)16i.p.SalineUnited States(53)
Potočnjak et al, 2022Severe combined immune deficientFemale12-1524-28Colorectal2 and 10 (once daily)21i.g.-Netherlands(27)
Lee et al, 2021BALB/c nudeFemale418-22Gastric10 (once daily)14i.p.DMSONetherlands(63)
Gao et al, 2016BALB/c nude miceFemale-20±2Lung50 and 100 (every other day)14s.c.SalineBrazil(64)
Woo et al, 2021BALB/c nudeFemale4-Melanoma75 and 150 (5 times a week)13i.p.-Netherlands(65)
Nie et al, 2016BALB/c nudeFemale6-Gastric100 (once daily)30p.o.DMSOEngland(66)

[i] -, no relevant information was provided in the study. Dosage was calculated as dosage=dose/(time interval of medication × medication days). i.p., intraperitoneal injection; i.g. intragastric administration; p.o., oral administration; s.c., subcutaneous; DMSO, dimethyl sulfoxide.

This study evaluated the antitumor effects of oleanolic acid by tumor weight and tumor volume. But in addition to the information presented in Table I, further in vitro and in vivo tests using OA had been performed in the included studies of the present systematic review. A total of 5 studies had 2 intervention groups, in which two doses of OA were tested. Furthermore, 7 studies had only 1 intervention group. The most common in vitro tests were cytotoxicity tests (n=9) and flow cytometry (n=3). The most frequent in vivo assessments were tumour volume (n=12), tumour weight (n=8) and animal weight (n=8) measurements.

Results of the risk of bias assessment

All studies included in the present review were evaluated using the RoB guideline's assessment of the risk of bias (Table SII). Numerous parameters were described as ‘unclear’, which indicated that the information reported in the reviewed articles was incomplete or unclear. This lack of clarity was predominantly related to sequence generation, baseline features and covert grouping. Of note, in none of the included studies, the evaluators were blinded regarding the results; however, this deficiency did not affect the accuracy of the results due to the objectivity of the measured results. As a result, these studies were identified as low-risk in the bias risk assessment.

Meta-analysis

Tumour volume and tumour weight were extracted as outcome measures for subsequent analysis to evaluate the antitumour effects of OA in animal experiments. No additional studies were included in this assessment.

Tumour volume

Forest plot analysis demonstrated that 10 studies reported the tumour volumes from 159 animals (Fig. 3C). Statistically significant differences were observed between the OA groups and control groups (mean difference, −0.64; 95% CI, −0.89 to −0.39; P<0.00001). These results suggest that treatment with OA significantly inhibited tumour growth compared with the control treatment.

Tumour weight

In total, 9 articles assessed differences in tumour weight between the control and OA groups (Fig. 3D). The differences in tumour weight between the OA and control groups were statistically significant (mean difference, −0.43; 95% CI, −0.58 to −0.28; P<0.00001). By the end of the experiments evaluated in the present review, OA-treated animals had significantly lower tumour weights compared with control animals, regardless of the tumour type.

Analysis of sources of heterogeneity
Sensitivity analysis

Of the included studies, 10 analysed the tumour volume. After excluding one study at a time, the heterogeneity remained unchanged. The I2 of each pooled analysis remained between 98–99%, which indicated no significant change in heterogeneity. In the 9 studies that analysed tumour weight, similar results were observed after each study was excluded individually. The I2 of each pooled analysis remained between 98–99%, which suggested no significant change in heterogeneity (Table II; Fig. S1).

Table II.

Sensitivity analyses of studies that reported tumor volume and weight.

Table II.

Sensitivity analyses of studies that reported tumor volume and weight.

A, Tumor volume

First author of omitted study, yearI2, %Pooled MD (95% CI)P-value(Refs.)
Niu et al, 201899−0.57 (−0.81, −0.33)<0.00001(62)
Nie et al, 201698−0.45 (−0.63, −0.28)<0.00001(66)
Wang et al, 201999−0.65 (−0.91, −0.38)<0.00001(47)
Potočnjak et al, 202299−0.64 (−0.91, −0.38)<0.00001(27)
Lee et al, 202198−0.71 (−1.08, −0.34)<0.00001(63)
Jiang et al, 202199−0.69 (−1.11, −0.27)<0.00001(53)
Woo et al, 202199−0.69 (−0.95, −0.42)<0.00001(65)
Li et al, 201599−0.64 (−0.91, −0.38)<0.00001(51)
Li et al, 201699−0.64 (−0.91, −0.38)<0.00001(52)
Gao et al, 201699−0.67 (−0.95, −0.40)<0.00001(64)

B, Tumor weight

First author of omitted study, yearI2, %Pooled MD (95% CI)P-value(Refs.)

Niu et al, 201899−0.42 (−0.58, −0.26)<0.00001(62)
Potočnjak et al, 202299−0.36 (−0.51, −0.21)<0.00001(27)
Lee et al, 202198−0.51 (−0.71, −0.31)<0.00001(63)
Jiang et al, 202199−0.48 (−0.67, −0.30)<0.00001(53)
Woo et al, 202199−0.49 (−0.67, −0.31)<0.00001(65)
Li et al, 201599−0.43 (−0.59, −0.27)<0.00001(51)
Tang et al, 201398−0.35 (−0.48, −0.22)<0.00001(61)
Gao et al, 201699−0.49 (−0.68, −0.31)<0.00001(64)
Liang et al, 202198−0.36 (−0.50, −0.22)<0.00001(46)

[i] MD, mean difference; CI, confidence interval.

Subgroup analysis

According to the Cochrane Handbook for Systematic Reviews of Interventions (58), as certain interventions shared a control group, only the total number of participants were divided into subgroup analyses. The original mean and standard deviation were unchanged.

Subgroup analyses were conducted based on the following factors: i) Dose of OA, categorized as low (<50 mg/kg), medium (50 mg/kg) or high (>50 mg/kg); ii) route of administration, including oral gavage, intraperitoneal, intragastric and subcutaneous injections; iii) type of cancer, including hepatocellular carcinoma, melanoma, and colorectal, cervical, gastric, breast and lung cancer; and iv) strains of mice utilized, including Kunming, severe combined immunodeficiency and BALB/c nude or athymic mice.

The subgroup analysis of the effects on the tumour volume demonstrated statistically significant differences among the different treatment regimens, tumour types and mouse strains (Table III). It should be noted that oleanolic acid had no significant effect on tumor volume inhibition in melanoma and gastric cancer (P>0.05). The subgroup analysis of tumour weight indicated that the dose, administration mode, tumour type and mouse strain had significant effects on the outcome (Table IV).

Table III.

Subgroup analysis of tumor volume.

Table III.

Subgroup analysis of tumor volume.

Variable/subgroupNo. of studiesSMD (95% CI)I2, %P-value
Oleanolic acid dosage
  Low6−2.51 (−3.35, −1.67)44<0.00001
  Medium2−2.59 (−3.56, −1.62)0<0.00001
  High4−3.41 (−5.71, −1.11)860.00400
Route of administration
  Intraperitoneal6−2.22 (−3.22, −1.22)64<0.00001
  Oral1−10.62 (−14.98, −6.27)-<0.00001
  Intragastric2−2.75 (−3.75, −1.75)0<0.00001
  Subcutaneous1−2.29 (−3.73, −0.86)-0.00200
Type of cancer
  Colorectal4−2.62 (−3.70, −1.54)47<0.00001
  Cervical1−3.38 (−4.97, −1.79)-<0.0001
  Gastric2−5.78 (−14.80, 3.25)940.21000
  Hepatocellular carcinoma1−2.84 (−4.15, −1.52)-<0.0001
  Lung1−2.29 (−3.73, −0.86)-0.00200
  Melanoma1−0.87 (−2.01, 0.26)-0.13000
Mouse species
  BALB/c nude7−3.14 (−4.60, −1.68)80<0.0001
  BALB/c athymic (nude)2−2.23 (−3.05, −1.40)0<0.00001
  Severe combined immunodeficiency disease1−2.64 (−4.17, −1.10)-0.00080

[i] Low, dosages <50 mg/kg; Medium, dosages of 50 mg/kg; High, dosages >50 mg/kg; SMD, standardized mean difference; CI, confidence interval.

Table IV.

Subgroup analysis of tumor weight.

Table IV.

Subgroup analysis of tumor weight.

Variable/subgroupNo. of studiesSMD (95% CI)I2, %P-value
Oleanolic acid dosage
  Low7−3.77 (4.88, −2.66)54<0.00001
  Medium1−1.63 (2.90, −0.36)-0.01000
  High4−3.04 (4.81, −1.27)810.00080
Route of administration
  Intraperitoneal5−2.65 (3.35, −1.95)0<0.00001
  Oral1−7.06 (9.60, −4.52)-<0.00001
  Intragastric2−5.07 (9.14, −1.01)660.00100
  Subcutaneous1−1.63 (2.90, −0.36)-0.00100
Type of cancer
  Colorectal3−3.36 (4.37, −2.35)0<0.00001
  Cervical2−4.86 (8.90, −0.81)870.02000
  Breast1−7.74 (12.27, −3.21)-0.00080
  Gastric1−2.53 (4.43, −0.64)-0.00900
  Lung1−1.63 (2.90, −0.36)-0.01000
  Melanoma1−1.74 (3.03, −0.44)-0.00900
Mouse strain
  BALB/c1−7.74 (12.27, −3.21)-0.00080
  BALB/c nude5−2.18 (2.85, −1.51)0<0.00001
  BALB/c athymic (nude)1−3.61 (5.14, −2.09)-<0.00001
  Kunming1−7.06 (9.60, −4.52)-<0.00001
  Severe combined immunodeficiency disease1−3.46 (5.26, −1.66)-0.00020

[i] Low, dosages <50 mg/kg; Medium, dosages of 50 mg/kg; High, dosages >50 mg/kg; SMD, standardized mean difference; CI, confidence interval.

Risk of bias analysis

Due to the limited number of included studies that assessed tumour weight (<10 publications), the evaluation of publication bias was limited to studies that evaluated tumour volume as an outcome. Egger's test was performed to quantify bias in this parameter and a funnel plot was generated to visualize the results (y-intercept, −4.97; 95% CI, 7.22–2.73; P=0.001; Fig. 3E and F). Additionally, the funnel plot for tumour volume exhibited asymmetry and skewness, with 3 out of 10 selected studies falling outside the 95% CI. These findings suggested the presence of publication bias in studies that reported the tumour volume.

Discussion

OA is an important pentacyclic triterpene that is widely found in plants, foods and medicines; it was initially used to treat chronic hepatitis and liver injury (18,67). Žiberna et al (68) reported that OA has potential in clinical adjuvant anticancer treatment, supported by its direct anticancer activity, synergistic effect with chemotherapy drugs, inhibition of transporters, enhancement of radiotherapy efficacy, low toxicity and lack of adverse reactions. These attributes suggest the promising potential for OA in clinical cancer therapy.

However, the therapeutic efficacy of oleanolic acid is constrained by its limited water solubility, low bioavailability (69). Structural modification and dosage form optimization could notably increase the water solubility and bioavailability of OA (70). At present, OA derivatives are primarily modified on the C-3 hydroxyl group, C-12/C-13 double bond and C-28 carboxyl group (71). Novel OA dosage forms consist of nanoparticles, liposomes, solid dispersions and phospholipid complexes (7275).

Research has indicated that OA demonstrates antitumour activity across a number of in vitro and in vivo models, exerting its effects on multiple targets. These targets include, but are not limited to, aldoketo reductase family member 1B10, protein tyrosine phosphatase 1B and cell division cycle 25 phosphatase (7678). To the best of our knowledge, no systematic review has evaluated the antitumour effects of OA in animal models. Therefore, the present meta-analysis holds particular significance to address this limitation in the field and lays the groundwork for future clinical explorations of OA.

The present study systematically evaluated the antitumour effects of OA by analysing the tumour volume and weight in experimental mice. Studies have demonstrated that oleanolic acid can significantly reduce tumor weight and inhibit the volumetric growth of most tumors. However, its anti-tumor efficacy appears suboptimal for certain malignancies, such as gastric cancer and melanoma (P<0.05). This limitation may be attributed to the small sample size, which compromises the reliability of the findings. Future research should aim to increase the sample size and employ robust statistical methodologies to enhance the reliability and clinical applicability of the results. At the same time, further research on the anti-tumor mechanism of oleanolic acid and development of water-soluble and bioavailable oleanolic acid derivatives will help improve its clinical significance in tumor therapy. OA exerts its antitumour effects through the modulation of various signalling pathways. OA can induce HCT116 colon cancer cell death through the p38/FOXO3a/Sirt6 pathway (27), and induce apoptosis and autophagy in AGS human gastric cancer cells through the PI3K/AKT/mTOR pathway (63). Additionally, OA inhibits the migration and invasion of glioma cells by inactivating the MAPK/ERK signalling pathway (29); it also induces apoptosis in A375SM and A375P melanoma cells through the NF-κB pathway (65). Furthermore, OA suppresses the proliferation of human bladder cancer cells by targeting the Akt/mTOR/S6K and ERK1/2 signalling pathways (26).

The present meta-analysis revealed substantial heterogeneity, with I2 values of 99% for both the tumour volume and weight. A subgroup analysis of the included studies was performed to identify sources of heterogeneity in the data. This indicated that the mode of administration, tumour type and mouse strain were potential sources of heterogeneity. Despite a comprehensive search, the limited number of studies included in the present analysis may have introduced bias. Given the lack of standardization of animal experiments, meta-analyses of preclinical studies are more heterogeneous compared with clinical studies (57,79). Maganti et al (80) conducted a systematic review of preclinical animal studies on the efficacy of CRISPR/Cas9 gene-edited chimeric antigen receptor T cells against malignant tumours, demonstrating significant heterogeneity (I2=96%), which may be attributed to the range of cancer types studied. Similarly, a meta-analysis by Singh et al (81) on the anticancer effects of apigenin on animal models of cancer also demonstrated high heterogeneity. Subgroup analyses demonstrated that the dose of apigenin, tumour model, route of administration and duration of treatment were significant factors that influenced heterogeneity. Furthermore, a sensitivity analysis was performed on the included studies using the leave-one-out method in the present review. After excluding one study at a time, the heterogeneity did not change significantly and the I2-value of tumour growth inhibition and tumour weight remained between 98–99%, which indicated the stability of the results. The high heterogeneity in the present study may be attributed to variations in the animal models and experimental conditions used. Since the tumour weight index was included in <10 studies, publication bias was assessed only for the tumour volume, and Egger's test and funnel plot results both indicated publication bias.

The antitumour effect of OA is suggested to be closely related to the immune system. Programmed cell death protein 1/programmed death-ligand 1 (PD-L1) blockade therapy is a promising cancer treatment strategy that is considered to have revolutionized the treatment landscape for malignant tumours (82). Lu et al (41) reported that OA can restore the effect of T cells on killing gastric cancer cells, achieve DNA hypomethylation and downregulate PD-L1 by inhibiting the IL-1β/NF-κB/TET3 signalling pathway, thus serving an antitumour role. In addition, Luo et al (83) reported that OA nanomicelles showed significant anticancer potential in a tumour-bearing mouse model and stimulated immune cell infiltration. As the main component of the tumour immune microenvironment, immune-infiltrating cells can effectively control tumour suppression and immune escape (84). However, through an analysis of the available literature, it was found that the quantification of the antitumour effects of OA in animal experiments has focused mainly on the tumour volume and weight, and has almost never involved an evaluation of immunity. Further studies are needed to clarify the relationship between the antitumour effects of OA and immune regulation.

The present study had several limitations, including the small number of included references and potential publication bias. But it still demonstrates significant strengths and research value. Extensive searches of major medical databases were conducted, and standard search terms were used to retrieve the relevant literature as comprehensively as possible and to reduce bias in selective reporting. An assessment of the risk of bias in the included studies increased the reliability of the results. Through subgroup analyses, the antitumour effects of OA under different conditions were further investigated, which provided guidance for further research and clinical application. In addition, this study also explored the possibility of OA enhancing the anti-tumor effect by modulating the immune system, opening up a new perspective for understanding its mechanism of action. In summary, this study provides solid theoretical support for the anti-tumor potential of oleanolic acid and promotes its transformation from laboratory research to clinical application.

The present review highlighted the antitumour potential of OA in animal models. Future studies are necessary to expand the sample size, unify the experimental design and further explore how OA serves an antitumour role by regulating the immune system. Future research could include determining the optimal dose and route of administration of OA, and developing derivatives to improve their solubility and bioavailability, while evaluating their long-term safety and toxicity. In addition, combination therapy, the standardization of preclinical models, molecular mechanism analyses, personalized medicine strategies, drug delivery system innovation and clinical trial design are key directions for promoting the clinical application of OA. Ultimately, the discovery and validation of biomarkers that predict OA responses will improve the development of precision medicine and provide novel strategies for cancer treatment.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

This present study was supported by the National Natural Science Foundation of China (grant no. 82160529), the Applied Basic Science Research Foundation of Yunnan Province (grant no. 202201AT070294), the Yunnan Revitalization Talent Support Program (grant no. RLQB20220014) and the 535 Talent Project of the First Affiliated Hospital of Kunming Medical University (grant no. 2023535D17).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

YZ and ZW were responsible for screening the data and writing the draft paper. JZ and WJ completed the data extraction and analysis of the results. QF was responsible for a thorough review of the manuscript prior to submission. QF and YZ jointly planned the overall structural design of the paper. YZ and ZW confirm the authenticity of all the raw data. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Russo A, Cardile V, Graziano ACE, Avola R, Montenegro I, Cuellar M, Villena J and Madrid A: Antigrowth activity and induction of apoptosis in human melanoma cells by Drymis winteri forst extract and its active components. Chem Biol Interact. 305:79–85. 2019. View Article : Google Scholar : PubMed/NCBI

2 

Yuan R, Hou Y, Sun W, Yu J, Liu X, Niu Y, Lu JJ and Chen X: Natural products to prevent drug resistance in cancer chemotherapy: A review. Ann N Y Acad Sci. 1401:19–27. 2017. View Article : Google Scholar : PubMed/NCBI

3 

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. View Article : Google Scholar : PubMed/NCBI

4 

Hung KC, Huang TC, Cheng CH, Cheng YW, Lin DY, Fan JJ and Lee KH: The expression profile and prognostic significance of metallothionein genes in colorectal cancer. Int J Mol Sci. 20:38492019. View Article : Google Scholar : PubMed/NCBI

5 

Gao L, Yang T, Xue Z and Chan CKD: Hot spots and trends in the relationship between cancer and obesity: A systematic review and knowledge graph analysis. Life (Basel). 13:3372023.PubMed/NCBI

6 

Baskar R, Lee KA, Yeo R and Yeoh KW: Cancer and radiation therapy: Current advances and future directions. Int J Med Sci. 9:193–199. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Wang X, Zhang H and Chen X: Drug resistance and combating drug resistance in cancer. Cancer Drug Resist. 2:141–160. 2019.PubMed/NCBI

8 

Ramos A, Sadeghi S and Tabatabaeian H: Battling chemoresistance in cancer: Root causes and strategies to uproot them. Int J Mol Sci. 22:94512021. View Article : Google Scholar : PubMed/NCBI

9 

Vasseur S and Guillaumond F: Lipids in cancer: A global view of the contribution of lipid pathways to metastatic formation and treatment resistance. Oncogenesis. 11:462022. View Article : Google Scholar : PubMed/NCBI

10 

Newman DJ and Cragg GM: Natural products as sources of new drugs from 1981 to 2014. J Nat Prod. 79:629–661. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Bishayee A and Sethi G: Bioactive natural products in cancer prevention and therapy: Progress and promise. Semin Cancer Biol. 40–41. 1–3. 2016.PubMed/NCBI

12 

Akter R, Afrose A, Rahman MR, Chowdhury R, Nirzhor SSR, Khan RI and Kabir MT: A comprehensive analysis into the therapeutic application of natural products as SIRT6 modulators in alzheimer's disease, aging, cancer, inflammation, and diabetes. Int J Mol Sci. 22:41802021. View Article : Google Scholar : PubMed/NCBI

13 

Wei J, Liu H, Liu M, Wu N, Zhao J, Xiao L, Han L, Chu E and Lin X: Oleanolic acid potentiates the antitumor activity of 5-fluorouracil in pancreatic cancer cells. Oncol Rep. 28:1339–1345. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Sultana N and Ata A: Oleanolic acid and related derivatives as medicinally important compounds. J Enzyme Inhib Med Chem. 23:739–756. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Baer-Dubowska W, Narożna M and Krajka-Kuźniak V: Anti-cancer potential of synthetic oleanolic acid derivatives and their conjugates with NSAIDs. Molecules. 26:49572021. View Article : Google Scholar : PubMed/NCBI

16 

Liu J: Oleanolic acid and ursolic acid: Research perspectives. J Ethnopharmacol. 100:92–94. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Prasad S, Kalra N and Shukla Y: Hepatoprotective effects of lupeol and mango pulp extract of carcinogen induced alteration in Swiss albino mice. Mol Nutr Food Res. 51:352–359. 2007. View Article : Google Scholar : PubMed/NCBI

18 

Singh GB, Singh S, Bani S, Gupta BD and Banerjee SK: Anti-inflammatory activity of oleanolic acid in rats and mice. J Pharm Pharmacol. 44:456–458. 1992. View Article : Google Scholar : PubMed/NCBI

19 

Pádua TA, de Abreu BSSC, Costa TEMM, Nakamura MJ, Valente LMM, Henriques MDG, Siani AC and Rosas EC: Anti-inflammatory effects of methyl ursolate obtained from a chemically derived crude extract of apple peels: Potential use in rheumatoid arthritis. Arch Pharm Res. 37:1487–1495. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Alqahtani A, Hamid K, Kam A, Wong KH, Abdelhak Z, Razmovski-Naumovski V, Chan K, Li KM, Groundwater PW and Li GQ: The pentacyclic triterpenoids in herbal medicines and their pharmacological activities in diabetes and diabetic complications. Curr Med Chem. 20:908–931. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Teodoro T, Zhang L, Alexander T, Yue J, Vranic M and Volchuk A: Oleanolic acid enhances insulin secretion in pancreatic beta-cells. FEBS Lett. 582:1375–1380. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Yu F, Wang Q, Zhang Z, Peng Y, Qiu Y, Shi Y, Zheng Y, Xiao S, Wang H, Huang X, et al: Development of oleanane-type triterpenes as a new class of HCV entry inhibitors. J Med Chem. 56:4300–4319. 2013. View Article : Google Scholar : PubMed/NCBI

23 

Cichewicz RH and Kouzi SA: Chemistry, biological activity, and chemotherapeutic potential of betulinic acid for the prevention and treatment of cancer and HIV infection. Med Res Rev. 24:90–114. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Martín R, Carvalho-Tavares J, Hernández M, Arnés M, Ruiz-Gutiérrez V and Nieto ML: Beneficial actions of oleanolic acid in an experimental model of multiple sclerosis: A potential therapeutic role. Biochem Pharmacol. 79:198–208. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Zhao X, Liu M and Li D: Oleanolic acid suppresses the proliferation of lung carcinoma cells by miR-122/Cyclin G1/MEF2D axis. Mol Cell Biochem. 400:1–7. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Mu DW, Guo HQ, Zhou GB, Li JY and Su B: Oleanolic acid suppresses the proliferation of human bladder cancer by Akt/mTOR/S6K and ERK1/2 signaling. Int J Clin Exp Pathol. 8:13864–13870. 2015.PubMed/NCBI

27 

Potočnjak I, Šimić L, Vukelić I, Batičić L and Domitrović R: Oleanolic acid induces HCT116 colon cancer cell death through the p38/FOXO3a/Sirt6 pathway. Chem Biol Interact. 363:1100102022. View Article : Google Scholar : PubMed/NCBI

28 

Zhou R, Zhang Z, Zhao L, Jia C, Xu S, Mai Q, Lu M, Huang M, Wang L, Wang X, et al: Inhibition of mTOR signaling by oleanolic acid contributes to its anti-tumor activity in osteosarcoma cells. J Orthop Res. 29:846–852. 2011. View Article : Google Scholar : PubMed/NCBI

29 

Guo G, Yao W, Zhang Q and Bo Y: Oleanolic acid suppresses migration and invasion of malignant glioma cells by inactivating MAPK/ERK signaling pathway. PLoS One. 8:e720792013. View Article : Google Scholar : PubMed/NCBI

30 

Wang X, Bai H, Zhang X, Liu J, Cao P, Liao N, Zhang W, Wang Z and Hai C: Inhibitory effect of oleanolic acid on hepatocellular carcinoma via ERK-p53-mediated cell cycle arrest and mitochondrial-dependent apoptosis. Carcinogenesis. 34:1323–1330. 2013. View Article : Google Scholar : PubMed/NCBI

31 

Wei J, Liu M, Liu H, Wang H, Wang F, Zhang Y, Han L and Lin X: Oleanolic acid arrests cell cycle and induces apoptosis via ROS-mediated mitochondrial depolarization and lysosomal membrane permeabilization in human pancreatic cancer cells. J Appl Toxicol. 33:756–765. 2013. View Article : Google Scholar : PubMed/NCBI

32 

Liu J, Zheng L, Wu N, Ma L, Zhong J, Liu G, Ma L, Zhong J, Liu G and Lin X: Oleanolic acid induces metabolic adaptation in cancer cells by activating the AMP-activated protein kinase pathway. J Agric Food Chem. 62:5528–5537. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Liu J, Wu N, Ma LN, Zhong JT, Liu G, Zheng LH and Lin XK: p38 MAPK signaling mediates mitochondrial apoptosis in cancer cells induced by oleanolic acid. Asian Pac J Cancer Prev. 15:4519–4525. 2014. View Article : Google Scholar : PubMed/NCBI

34 

Li HF, Wang XA, Xiang SS, Hu YP, Jiang L, Shu YJ, Li ML, Wu XS, Zhang F, Ye YY, et al: Oleanolic acid induces mitochondrial-dependent apoptosis and G0/G1 phase arrest in gallbladder cancer cells. Drug Des Devel Ther. 9:3017–3030. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Li X, Song Y, Zhang P, Zhu H, Chen L, Xiao Y and Xing Y: Oleanolic acid inhibits cell survival and proliferation of prostate cancer cells in vitro and in vivo through the PI3K/Akt pathway. Tumour Biol. 37:7599–7613. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Xu Y, Shu B, Tian Y, Wang G, Wang Y, Wang J and Dong Y: Oleanolic acid induces osteosarcoma cell apoptosis by inhibition of Notch signaling. Mol Carcinog. 57:896–902. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Guo Y, Han B, Luo K, Ren Z, Cai L and Sun L: NOX2-ROS-HIF-1α signaling is critical for the inhibitory effect of oleanolic acid on rectal cancer cell proliferation. Biomed Pharmacother. 85:733–739. 2017. View Article : Google Scholar : PubMed/NCBI

38 

Li Y, Xu Q, Yang W, Wu T and Lu X: Oleanolic acid reduces aerobic glycolysis-associated proliferation by inhibiting yes-associated protein in gastric cancer cells. Gene. 712:1439562019. View Article : Google Scholar : PubMed/NCBI

39 

Duan L, Yang Z, Jiang X, Zhang J and Guo X: Oleanolic acid inhibits cell proliferation migration and invasion and induces SW579 thyroid cancer cell line apoptosis by targeting forkhead transcription factor A. Anticancer Drugs. 30:812–820. 2019. View Article : Google Scholar : PubMed/NCBI

40 

Kim GJ, Jo HJ, Lee KJ, Choi JW and An JH: Oleanolic acid induces p53-dependent apoptosis via the ERK/JNK/AKT pathway in cancer cell lines in prostatic cancer xenografts in mice. Oncotarget. 9:26370–26386. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Lu X, Li Y, Yang W, Tao M, Dai Y, Xu J and Xu Q: Inhibition of NF-κB is required for oleanolic acid to downregulate PD-L1 by promoting DNA demethylation in gastric cancer cells. J Biochem Mol Toxicol. 35:e226212021. View Article : Google Scholar : PubMed/NCBI

42 

Zeng Z, Yu J, Jiang Z and Zhao N: Oleanolic acid (OA) targeting UNC5B inhibits proliferation and EMT of ovarian cancer cell and increases chemotherapy sensitivity of niraparib. J Oncol. 2022:58876712022. View Article : Google Scholar : PubMed/NCBI

43 

Ren J, Yan J, Raza F, Zafar H, Wan H, Chen X, Cui Q, Li H and Wang X: A synergistic combination of oleanolic acid and apatinib to enhance antitumor effect on liver cancer cells and protect against hepatic injury. Recent Pat Anticancer Drug Discov. 19:199–208. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Hosny S, Sahyon H, Youssef M and Negm A: Oleanolic acid suppressed DMBA-induced liver carcinogenesis through induction of mitochondrial-mediated apoptosis and autophagy. Nutr Cancer. 73:968–982. 2021. View Article : Google Scholar : PubMed/NCBI

45 

Tang ZY, Li Y, Tang YT, Ma XD and Tang ZY: Anticancer activity of oleanolic acid and its derivatives: Recent advances in evidence, target profiling and mechanisms of action. Biomed Pharmacother. 145:1123972022. View Article : Google Scholar : PubMed/NCBI

46 

Liang Z, Pan R, Meng X, Su J, Guo Y, Wei G, Zhang Z and He K: Transcriptome study of oleanolic acid in the inhibition of breast tumor growth based on high-throughput sequencing. Aging (Albany NY). 13:22883–22897. 2021. View Article : Google Scholar : PubMed/NCBI

47 

Wang H, Zhong W, Zhao J, Zhang H, Zhang Q, Liang Y, Chen S, Liu H, Zong S, Tian Y, et al: Oleanolic acid inhibits epithelial-mesenchymal transition of hepatocellular carcinoma by promoting iNOS dimerization. Mol Cancer Ther. 18:62–74. 2019. View Article : Google Scholar : PubMed/NCBI

48 

Bednarczyk-Cwynar B, Wachowiak N, Szulc M, Kamińska E, Bogacz A, Bartkowiak-Wieczorek J, Zaprutko L and Mikolajczak PL: Strong and long-lasting antinociceptive and anti-inflammatory conjugate of naturally occurring oleanolic acid and aspirin. Front Pharmacol. 7:2022016. View Article : Google Scholar : PubMed/NCBI

49 

Feng H, Wu YQ, Xu YS, Wang KX, Qin XM and Lu YF: LC-MS-based metabolomic study of oleanolic acid-induced hepatotoxicity in mice. Front Pharmacol. 11:7472020. View Article : Google Scholar : PubMed/NCBI

50 

Liu Y, Luo X, Xu X, Gao N and Liu X: Preparation, characterization and in vivo pharmacokinetic study of PVP-modified oleanolic acid liposomes. Int J Pharm. 517:1–7. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Li L, Wei L, Shen A, Chu J, Lin J and Peng J: Oleanolic acid modulates multiple intracellular targets to inhibit colorectal cancer growth. Int J Oncol. 47:2247–2254. 2015. View Article : Google Scholar : PubMed/NCBI

52 

Li L, Lin J, Sun G, Wei L, Shen A, Zhang M and Peng J: Oleanolic acid inhibits colorectal cancer angiogenesis in vivo and in vitro via suppression of STAT3 and Hedgehog pathways. Mol Med Rep. 13:5276–5282. 2016. View Article : Google Scholar : PubMed/NCBI

53 

Jiang X, Shi M, Sui M, Yuan Y, Zhang S, Xia Q and Zhao K: Oleanolic acid inhibits cervical cancer Hela cell proliferation through modulation of the ACSL4 ferroptosis signaling pathway. Biochem Biophys Res Commun. 545:81–88. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Page MJ, McKenzie JE, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. BMJ. 372:n712021. View Article : Google Scholar : PubMed/NCBI

55 

Page MJ, Moher D, Bossuyt PM, Boutron I, Hoffmann TC, Mulrow CD, Shamseer L, Tetzlaff JM, Akl EA, Brennan SE, et al: PRISMA 2020 explanation and elaboration: Updated guidance and exemplars for reporting systematic reviews. BMJ. 372:n1602021. View Article : Google Scholar : PubMed/NCBI

56 

Richardson WS, Wilson MC, Nishikawa J and Hayward RS: The well-built clinical question: A key to evidence-based decisions. ACP J Club. 123:A12–A13. 1995. View Article : Google Scholar : PubMed/NCBI

57 

Hooijmans CR, Rovers MM, de Vries RBM, Leenaars M, Ritskes-Hoitinga M and Langendam MW: SYRCLE's risk of bias tool for animal studies. BMC Med Res Methodol. 14:432014. View Article : Google Scholar : PubMed/NCBI

58 

Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ and Welch VA: Cochrane handbook for systematic reviews of interventions version 6.2 [updated February 2021]. Cochrane. 2021.Available from:. https://training.cochrane.org/handbook

59 

Cao LM, Sun ZX, Makale EC, Du GK, Long WF and Huang HR: Antitumor activity of fucoidan: A systematic review and meta-analysis. Transl Cancer Res. 10:5390–5405. 2021. View Article : Google Scholar : PubMed/NCBI

60 

Vesterinen HM, Sena ES, Egan KJ, Hirst TC, Churolov L, Currie GL, Antonic A, Howells DW and Macleod MR: Meta-analysis of data from animal studies: a practical guide. J Neurosci Methods. 221:92–102. 2014. View Article : Google Scholar : PubMed/NCBI

61 

Tang S, Gao D, Zhao T, Zhou J and Zhao X: An evaluation of the anti-tumor efficacy of oleanolic acid-loaded PEGylated liposomes. Nanotechnology. 24:2351022013. View Article : Google Scholar : PubMed/NCBI

62 

Niu G, Sun L, Pei Y and Wang D: Oleanolic acid inhibits colorectal cancer angiogenesis by blocking the VEGFR2 signaling pathway. Anticancer Agents Med Chem. 18:583–590. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Lee JH, Yoo ES, Han SH, Jung GH, Han EJ, Jung SH, Kim BS, Cho SD, Nam JS, Choi C, et al: Oleanolic acid induces apoptosis and autophagy via the PI3K/AKT/mTOR pathway in AGS human gastric cancer cells. J Funct Foods. 87:1048542021. View Article : Google Scholar

64 

Gao YS, Yuan Y, Song G and Lin SQ: Inhibitory effect of ursolic acid and oleanolic acid from Eriobotrya fragrans on A549 cell viability in vivo. Genet Mol Res. 15:2016. View Article : Google Scholar

65 

Woo JS, Yoo ES, Kim SH, Lee JH, Han SH, Jung SH, Jung GH and Jung JY: Anticancer effects of oleanolic acid on human melanoma cells. Chem Biol Interact. 347:1096192021. View Article : Google Scholar : PubMed/NCBI

66 

Nie H, Wang Y, Qin Y and Gong XG: Oleanolic acid induces autophagic death in human gastric cancer cells in vitro and in vivo. Cell Biol Int. 40:770–778. 2016. View Article : Google Scholar : PubMed/NCBI

67 

Liu J, Liu Y, Madhu C and Klaassen CD: Protective effects of oleanolic acid on acetaminophen-induced hepatotoxicity in mice. J Pharmacol Exp Ther. 266:1607–1613. 1993.PubMed/NCBI

68 

Žiberna L, Šamec D, Mocan A, Nabavi SF, Bishayee A, Farooqi AA, Sureda A and Nabavi SM: Oleanolic acid alters multiple cell signaling pathways: implication in cancer prevention and therapy. Int J Mol Sci. 18:6432017. View Article : Google Scholar

69 

Zhong YY, Chen HS, Wu PP, Zhang BJ, Yang Y, Zhu QY, Zhang CG and Zhao SQ: Synthesis and biological evaluation of novel oleanolic acid analogues as potential α-glucosidase inhibitors. Eur J Med Chem. 164:706–716. 2019. View Article : Google Scholar : PubMed/NCBI

70 

Yang R, Huang X, Dou J, Zhai G and Su L: Self-microemulsifying drug delivery system for improved oral bioavailability of oleanolic acid: Design and evaluation. Int J Nanomedicine. 8:2917–2926. 2013.PubMed/NCBI

71 

Tang C, Chen Y, Bai S and Yang G: Advances in the study of structural modification and biological activities of oleanolic acid. Chin J Org Chem. 33:46–65. 2013. View Article : Google Scholar

72 

Jiang Q, Yang X, Du P, Zhang H and Zhang T: Dual strategies to improve oral bioavailability of oleanolic acid: Enhancing water-solubility, permeability and inhibiting cytochrome P450 isozymes. Eur J Pharm Biopharm. 99:65–72. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Gao N, Guo M, Fu Q and He Z: Application of hot melt extrusion to enhance the dissolution and oral bioavailability of oleanolic acid. Asian J Pharm Sci. 12:66–72. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Xia X, Liu H, Lv H, Zhang J, Zhou J and Zhao Z: Preparation, characterization, and in vitro/vivo studies of oleanolic acid-loaded lactoferrin nanoparticles. Drug Des Devel Ther. 11:1417–1427. 2017. View Article : Google Scholar : PubMed/NCBI

75 

Wei CT, Wang YW, Wu YC, Lin LW, Chen CC, Chen CY and Kuo SM: Reparative efficacy of liposome-encapsulated oleanolic acid against liver inflammation induced by fine ambient particulate matter and alcohol in mice. Pharmaceutics. 14:11082022. View Article : Google Scholar : PubMed/NCBI

76 

Lu Y, Zheng W, Lin S, Guo F, Zhu Y, Wei Y, Liu X, Jin S, Jin L and Li Y: Identification of an oleanane-type triterpene hedragonic acid as a novel farnesoid X receptor ligand with liver protective effects and anti-inflammatory activity. Mol Pharmacol. 93:63–72. 2018. View Article : Google Scholar : PubMed/NCBI

77 

Li Y, Yu Y, Jin K, Gao L, Luo T, Sheng L, Shao X and Li J: Synthesis and biological evaluation of novel thiadiazole amides as potent Cdc25B and PTP1B inhibitors. Bioorg Med Chem Lett. 24:4125–4128. 2014. View Article : Google Scholar : PubMed/NCBI

78 

Feng MT, Wang T, Liu AH, Li J, Yao LG, Wang B, Guo YW and Mao SC: PTP1B inhibitory and cytotoxic C-24 epimers of Δ28−24-hydroxy stigmastane-type steroids from the brown alga Dictyopteris undulata Holmes. Phytochemistry. 146:25–35. 2018. View Article : Google Scholar : PubMed/NCBI

79 

de Oliveira TV, Stein R, de Andrade DF and Beck RCR: Preclinical studies of the antitumor effect of curcumin-loaded polymeric nanocapsules: A systematic review and meta-analysis. Phytother Res. 36:3202–3214. 2022. View Article : Google Scholar : PubMed/NCBI

80 

Maganti HB, Kirkham AM, Bailey AJM, Shorr R, Kekre N, Pineault N and Allan DS: Use of CRISPR/Cas9 gene editing to improve chimeric antigen-receptor T cell therapy: A systematic review and meta-analysis of preclinical studies. Cytotherapy. 24:405–412. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Singh D, Gupta M, Sarwat M and Siddique HR: Apigenin in cancer prevention and therapy: A systematic review and meta-analysis of animal models. Crit Rev Oncol Hematol. 176:1037512022. View Article : Google Scholar : PubMed/NCBI

82 

Sun JY, Zhang D, Wu S, Xu M, Zhou X, Lu XJ and Ji J: Resistance to PD-1/PD-L1 blockade cancer immunotherapy: Mechanisms, predictive factors, and future perspectives. Biomark Res. 8:352020. View Article : Google Scholar : PubMed/NCBI

83 

Luo QW, Yao L, Li L, Yang Z, Zhao MM, Zheng YZ, Zhuo FF, Liu TT, Zhang XW, Liu D, et al: Inherent capability of self-assembling nanostructures in specific proteasome activation for cancer cell pyroptosis. Small. 19:e22055312023. View Article : Google Scholar : PubMed/NCBI

84 

Zhou Z, Song Q, Yang Y, Wang L and Wu Z: Comprehensive landscape of RRM2 with immune infiltration in pan-cancer. Cancers (Basel). 14:29382022. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 28 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zeng Y, Wang Z, Zhang J, Jian W and Fu Q: Antitumour activity of oleanolic acid: A systematic review and meta‑analysis. Oncol Lett 28: 582, 2024.
APA
Zeng, Y., Wang, Z., Zhang, J., Jian, W., & Fu, Q. (2024). Antitumour activity of oleanolic acid: A systematic review and meta‑analysis. Oncology Letters, 28, 582. https://doi.org/10.3892/ol.2024.14715
MLA
Zeng, Y., Wang, Z., Zhang, J., Jian, W., Fu, Q."Antitumour activity of oleanolic acid: A systematic review and meta‑analysis". Oncology Letters 28.6 (2024): 582.
Chicago
Zeng, Y., Wang, Z., Zhang, J., Jian, W., Fu, Q."Antitumour activity of oleanolic acid: A systematic review and meta‑analysis". Oncology Letters 28, no. 6 (2024): 582. https://doi.org/10.3892/ol.2024.14715