Next Article in Journal
Magnesium Depletion Score as an Indicator of Health Risk and Nutritional Status—A Scoping Review
Previous Article in Journal
New Anthropometry-Based Formulae to Predict 24 h Sodium Excretion from Spot Urine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Potential of Traditional Chinese Medicine Brucea javanica in Cancer Treatment: A Review of Chemical Constituents, Pharmacology, and Clinical Applications

1
National Institutes for Food and Drug Control, Beijing 100050, China
2
Public Experimental Center, Changchun University of Chinese Medicine, Changchun 130117, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2025, 17(20), 3285; https://doi.org/10.3390/nu17203285
Submission received: 17 September 2025 / Revised: 7 October 2025 / Accepted: 16 October 2025 / Published: 20 October 2025

Abstract

Brucea javanica (BJ), a key representative of traditional Chinese herbal medicine, is derived from the dried mature fruit of Brucea javanica (L.) Merr., a plant in the Simaroubaceae family. Its pharmacological activity is largely attributed to diverse chemical constituents. To date, approximately 200 distinct chemical constituents have been isolated and identified, mainly comprising quassinoids, triterpenes, alkaloids, steroids, phenylpropanoids, and flavonoids. Contemporary pharmacological studies have demonstrated the significant activities of BJ in various areas, including anti-tumor, anti-inflammatory, and anti-parasitic effects. Notably, its oil form (Brucea javanica oil) has been extensively utilized in treating various cancer types. This review aims to systematically summarize the antitumor components, mechanisms of action, and clinical applications in cancer therapy, with the goal of providing theoretical support for further antitumor research and the development of new BJ-based drugs, highlighting its potential as an antitumor agent.

1. Introduction

Cancer, a highly destructive and heterogeneous disease, is primarily characterized by uncontrolled cell proliferation and the invasive behavior of abnormal cells [1]. Although approximately 5% to 10% of cancer cases are hereditary, the majority are closely linked to exogenous factors, such as environmental stimuli, dietary habits, lifestyle choices, and environmental pollution [2]. Cancer mortality rates have steadily increased, making it one of the leading global health threats [3]. Despite extensive research over the past decades into the treatment of malignant diseases, a definitive cure remains elusive. Current therapeutic approaches—including surgery, radiotherapy, chemotherapy, hormone therapy, immunotherapy, and targeted therapy—are limited by challenges such as systemic toxicity, severe side effects, drug resistance, high treatment costs, and low patient tolerance [4,5,6]. These factors significantly limit the acceptability and clinical applicability of these treatments. Therefore, the search for more efficient, selective, and novel therapeutic approaches to address the needs of various malignancies remains an urgent task.
Since antiquity, natural products and traditional Chinese herbal medicines have been extensively applied in cancer treatment and have played a crucial role in the discovery and development of anticancer agents [7,8]. Numerous studies have revealed that a vast array of natural products constitute a critical resource for anticancer agents, with over half of clinically used anticancer drugs derived from these substances [9,10,11]. For example, paclitaxel [12,13] and camptothecin [14], extracted from Taxus wallichiana var. chinensis (Pilger) Florin and Camptotheca acuminata Decne., respectively, have become pivotal drugs in anticancer therapy. Active components in Chinese herbal medicines—such as terpenoids, alkaloids, and flavonoids—exert anticancer effects through multiple mechanisms, including the modulation of signaling pathways, induction of apoptosis, and inhibition of angiogenesis. Clinical studies have demonstrated that the combined use of specific Chinese herbal medicines and chemotherapeutic agents can enhance therapeutic efficacy while mitigating adverse effects [15,16]. Furthermore, the structural diversity of natural products holds significant potential in cancer chemoprevention, providing novel insights for innovative drug design.
Brucea javanica (L.) Merr. (Simaroubaceae) is widely distributed in Southeast Asia and northern Australia. In China, it is primarily located in Fujian, Taiwan, Guangdong, Guangxi, Hainan, and Yunnan provinces [17]. Its dried mature fruits are commonly used as medicinal materials and hold significant importance in traditional Chinese medicine, exhibiting properties such as heat-clearing, dampness-drying, and detoxification. BJ demonstrates notable efficacy in treating dysentery, expelling parasites, and combating malaria, while also exhibiting antitumor activity against various cancers, including lung, rectal, esophageal, and colorectal cancers [17,18]. Additionally, it can be applied topically to treat skin lesions, such as warts and corns. Modern pharmacological studies have confirmed that BJ exerts significant inhibitory effects on multiple malignant tumors, particularly showing promising therapeutic outcomes in respiratory [19] and digestive system cancers [20,21]. Consequently, research on the active components of BJ and its anticancer properties has garnered increasing academic attention, with numerous researchers dedicated to elucidating its underlying molecular mechanisms. Since the 1970s, Chinese researchers have successfully developed Brucea javanica oil emulsion (BJOE) for intravenous administration as an anticancer agent. To date, Brucea javanica oil injection has been widely used in clinical practice, demonstrating significant therapeutic efficacy both as a standalone treatment [22] and in combination with radiotherapy [23] or chemotherapy agents [24]. Brucea javanica oil (BJO, also known as BJOE injection), a fatty oil extracted from the dried ripe fruits of Brucea javanica (L.) Merr., has become an important adjunctive therapy for patients with solid tumors during chemotherapy. This preparation enhances chemotherapy efficacy by increasing overall response rates, reducing postoperative adverse effects, and improving patients’ quality of life [25,26]. It is well established that chemotherapy agents often cause substantial harm to patients, with common adverse effects including leukopenia, hepatorenal impairment, and vomiting [27]. However, anticancer traditional Chinese medicine injections primarily serve as adjuvant therapies for tumor radiotherapy and chemotherapy, exhibiting functions such as toxicity reduction, symptom alleviation, and efficacy enhancement [28]. Furthermore, BJ-containing formulations, including capsules and oral preparations, have gained widespread clinical application as anticancer agents.
With the ongoing expansion of research in natural products, plant-derived bioactive compounds have become a focal point of investigation. Concurrently, the structural diversity of natural products has shown significant potential in cancer chemoprevention, providing novel insights for innovative drug design. In recent years, BJ has emerged as a research hotspot due to its remarkable antitumor bioactivity. Numerous studies have confirmed that BJ exhibits significant inhibitory effects against various malignant tumors, underscoring its high research value. Given the strong interest among readers for comprehensive information about BJ, this review systematically summarizes the antitumor-active chemical constituents isolated from BJ and their potential mechanisms of action. Furthermore, it elucidates the therapeutic roles of these compounds and provides an in-depth discussion of relevant clinical applications, aiming to offer theoretical support and references for further research, development, and clinical utilization of Brucea javanica.

2. Anticancer Chemical Ingredients

In recent years, the number of researchers dedicated to Brucea javanica (BJ) studies has increased significantly, with research primarily focused on bioactive molecules and their antitumor properties. With continuous advancements in analytical techniques, researchers have isolated over 200 natural compounds from BJ. These compounds can be broadly categorized into six major groups: quassinoids, alkaloids, triterpenes, steroids, phenylpropanoids, flavonoids, and others [21,29,30,31,32]. Notable examples include bruceine [33], brusatol [34], bruceantin [35], dehydrobrusatol [36], dehydrobruceantinol, dihydrobruceine, bruceantinol, bruceaketolic acid, yadanzioside [36,37], bruceoside, and bruceolide. Among these, quassinoids—the primary constituents of BJ—are widely recognized as the key bioactive components responsible for its anticancer activity. Notably, the most prominent quassinoids are classified as C20-quassinoids [38]. Their core structure consists of three hexagonal rings and a lactone ring. This class includes 110 compounds, with brusatol and bruceantin serving as representative examples exhibiting diverse pharmacological effects, including anti-inflammatory and tumor growth-inhibiting activities [39]. Research indicates that brusatol is primarily extracted from the seeds and fruits of Brucea javanica, where it constitutes approximately 0.3% of the fruit content [39,40]. It exhibits high concentrations and demonstrates significant antitumor activity against various cancers. The molecular structure of brusatol consists of an α, β-unsaturated cyclohexanone ring, two cyclohexane rings, a six-membered lactone ring, and a tetrahydrofuran ring [41]. Furthermore, studies on the pharmacophore moieties of the compound reveal that its activity primarily originates from the keto oxygen at the C-2 position, the enol oxygen at the C-11 position, and the epoxy methane bridge spanning C-8 to C-13 or C-8 to C-11 [42,43,44]. Figure 1 presents the structural schematic of anticancer active compounds in BJ.

3. Anticancer Effects

3.1. Anti-Lung Cancer

Lung cancer is the most frequently diagnosed malignancy in many countries and remains the foremost cause of cancer-related mortality worldwide [45,46]. Despite continuous progress in diagnostic and therapeutic strategies, patient survival rates remain poor. The discovery of multiple compounds in BJ that exhibit significant cytotoxic effects on lung cancer cells suggests that these components hold substantial potential for further investigation in lung cancer research. In the A549 lung cancer cell line, bruceanol series compounds demonstrated significant antitumor activity. Specifically, bruceanol C, E, D, and F had half-maximal inhibitory concentrations as low as 0.0064 μmol/L [47,48,49]. Additionally, bruceoside C, D, E, and F exhibit significant cytotoxicity in non-small cell lung cancer (NSCLC) cell lines [50,51]. Multiple studies have shown that bruceine B, 24-epipiscidinol A, yadanziolide B, quassilactone A and B, and bruceine D significantly inhibit the survival rate of H460 and A549 cells [29,49,52,53,54]. Cytotoxicity assays revealed that ethanol, petroleum ether, ethyl acetate, and n-butanol extracts of BJ exerted strong cytotoxic effects against the A549 cell line, with IC50 values spanning 0.02–17.47 μg/mL [55]. The water extract of BJ can induce apoptosis in non-small cell lung cancer (NSCLC) A549 cells, with an IC50 of 50 μg/mL [56]. Additionally, researchers observed that this extract exhibits specific inhibitory activity against the NSCLC cell line H1975, which carries the L858R/T790M epidermal growth factor receptor (EGFR) mutation [57]. Makong et al. [58] revealed that the methanol extract, dichloromethane, and ethyl acetate-soluble fractions of BJ root and bark, as well as compounds isolated from them (bruceacanthinones A, B et al.), exhibited significant cytotoxic effects on the A549 cell line. The IC50 values ranged from 50.0 ± 5.2 to 80.5 ± 1.8 μg/mL, with the cytotoxic effects of individual compounds being less potent compared to the crude extract. In A549 xenograft models, combined treatment with brusatol and cisplatin enhanced apoptosis, suppressed cell proliferation, and more effectively inhibited tumor growth than cisplatin alone [59].

3.2. Anti-Digestive System Cancer

Pancreatic cancer is a deadly disease characterized by aggressive tumor biology, with a higher prevalence in men [60]. Unfortunately, this disease is often asymptomatic, leading to diagnoses at advanced stages [61]. In vitro experiments on pancreatic cancer cells demonstrated that brusatol had IC50 values of 0.36 mmol/L for PANC-1 and 0.10 mmol/L for SW1990 cell lines [21], respectively. It also inhibited the proliferation of the PATU-8988 cell line and induced apoptosis [62]. The IC50 value of bruceine D for Hs68 cells exceeds 30 μmol/L [63], and it exhibits significant cytotoxic effects on Panc-1, SW1990, and Capan-1 cells, with antiproliferative effects comparable to those of the positive control drugs camptothecin and gemcitabine [64]. Bruceine A exhibited strong cytotoxic effects on human pancreatic cancer cell lines MIA PaCa-2, SW1990, PANC-1, and AsPC-1, inhibiting cancer cell proliferation in a time- and dose-dependent manner [65]. Furthermore, bruceine A caused a dose-dependent reduction in tumor growth in human pancreatic tumor-bearing mice, with its inhibitory effect at a dose of 0.5 mg/kg comparable to that of the positive control drug gemcitabine [65]. Moreover, Zhao et al. [21] demonstrated that bruceantinoside A, brusatol, yadanzioside A, yadanzioside C, bruceine D, bruceine H, and javanicoside G possess in vitro anti-pancreatic cancer activity, with brusatol showing the greatest potency.
Colorectal cancer is the third most commonly diagnosed malignancy worldwide and the second leading cause of cancer-related mortality, representing a major burden on global healthcare systems [66]. Tumor heterogeneity and clonal evolution during treatment contribute to frequent drug resistance issues [67]. However, natural products have garnered considerable attention due to their potent anticancer properties and relatively low adverse reaction rates. In 1993, Imamura et al. [47] demonstrated that bruceanol D, E, and F exhibited cytotoxicity against HCT-8 ileocecal cancer cells, with median effective dose (ED50) values ranging from 0.16 to 0.67 μg/mL. Subsequently, yadanziolide B, T, and bruceine B, D, E, and H exhibited cytotoxicity against HCT-8 cells, with IC50 values ranging from 1.3 to 6.7 μmol/L [29,68]. Numerous experimental studies in colorectal cancer treatment research have confirmed the sensitivity of the CT26 [69], HCT116 [70], PKO, SW480, and COLO205 cell lines to brusatol [71]. Notably, brusatol shows a marked cytotoxic effect on HCT116 cells when its concentration exceeds 15 nmol/L [70]. In CT26 cells, the IC50 value of this compound reaches 373 nmol/L [69]. Further studies indicate that cytotoxicity assessments of the SW480 cell line revealed IC50 values for brusatol, bruceine B, D, and yadanziolide A ranging from 0.1 to 28.5 μmol/L [43]. Based on these data, it can be concluded that brusatol exhibits the most potent cytotoxic effect on colorectal cancer cells among these monomeric compounds. Moreover, the IC50 values of the ethanol extract of BJ against HCT-116 and HT29 cells were 8.9 ± 1.32 μg/mL [72] and 48 ± 2.5 μg/mL [20], respectively. In in vivo experiments, brusatol administered at a dose of 2 mg/kg effectively inhibited the growth of xenograft and in situ tumors [69,71]. Notably, cisplatin and irinotecan, long-term treatment drugs for colorectal cancer, significantly enhance treatment efficacy when used in combination with brusatol or cisplatin [71,73].
Liver cancer is the sixth most frequently diagnosed malignancy worldwide and the fourth major cause of cancer-related mortality, with a five-year survival rate of only 18% [74]. Its high incidence and mortality rates contribute to its status as one of the most socially challenging global health issues [75]. Research on liver cancer has shown that brusatol, bruceine D, and B all exhibit significant cytotoxic effects. These studies are primarily conducted in vitro, with brusatol exhibiting the most significant cytotoxic effect in the SMMC7721 cell line, where the IC50 value is below 0.064 μmol/L. In contrast, bruceine B exhibits an IC50 value of 0.15 μmol/L for SMMC7721 hepatocellular carcinoma cells [49]. Furthermore, Ye et al. [76] reported that brusatol suppressed the proliferation of hepatocellular carcinoma cells in a dose-dependent fashion, with IC50 values of 0.69 μmol/L (Hep3B), 0.34 μmol/L (Huh7), 12.49 μmol/L (LM3), and 18.04 nmol/L (Bel-7404). Additionally, javanicolide H and E, bruceine B, E, H, and dehydrobrusatol exhibited cytotoxicity against HepG2 cells, with IC50 values ranging between 0.81 and 3.3 μmol/L [68]. Similarly, Yadanziolide T, B [29], together with bruceantinol [35], demonstrated marked growth-inhibitory activity in Bel-7402 and Bel-7404 cells, with IC50 values of 3.5–4.5 μmol/L and 10 μmol/L, respectively. In 2005, Lau et al. [56] reported that the aqueous extract of BJ triggered apoptosis in Hep3B cells, with an IC50 of approximately 50 μg/mL. A decade later, Chen et al. [77] further validated the cytotoxic activity of the BJ aqueous extract against the same cell line, observing an IC50 of 4 mg/mL. Notably, differences in dose–response effects of the BJ water extract may be attributed to inconsistencies in extraction methods.
Gastric cancer represents one of the most common malignancies worldwide, ranking as the fifth most frequently diagnosed cancer [78]. Because the majority of cases are identified at advanced stages, mortality remains substantial, positioning gastric cancer as the third primary cause of cancer-associated deaths. In the gastric cancer cell line BGC-823, yadanziolide T, yadanziolide B, javanicolide H, bruceine B, bruceine D, bruceine E, bruceine H, dehydrobrusatol, javanicolide E, and 24-epipiscidinol A all demonstrated significant cytotoxic effects. Among these compounds, javanicolide H showed the strongest cytotoxic effect, with an IC50 value of 0.52 μmol/L [29,52,68].

3.3. Anti-Reproductive System Cancer

Ovarian cancer is a highly aggressive malignancy, often diagnosed at advanced stages [79]. Despite initial surgery and chemotherapy, recurrence occurs in the majority of patients, underscoring the urgent need for novel therapeutic strategies. Globally, ovarian cancer—including malignancies of the ovary, fallopian tube, and peritoneum—accounts for an estimated 313,959 new cases and 207,252 deaths each year [80,81]. Studies exploring the application of BJ in reproductive system tumors have largely concentrated on breast, ovarian, and cervical cancers. In this field, preliminary investigations have examined the potential activity of compounds such as brusatol, bruceantinol, bruceine A, bruceine B, and brujavanol E against breast cancer. Furthermore, bruceine B, D, and H have been shown to exert notable cytotoxicity toward ovarian cancer cells. In addition, Bruceosides D, E, and F displayed marked and selective cytotoxic activity in ovarian cancer cell lines [51]. Subsequently, the cytotoxic effects of javanicolide E, bruceine B, D, E, H, and dehydrobrusatol were confirmed in SKOV3 cells, with half-maximal inhibitory concentration values ranging from 0.12 to 2.5 μmol/L [68].
Breast cancer is the most prevalent cancer among women globally. Characterized by distinct epidemiological features and marked heterogeneity, it continues to be a major cause of cancer-related mortality and poses a serious threat to women’s health [82]. Recent investigations have shown that compounds including brusatol, bruceantin, bruceines A, B, D, and E, brujavanol E, yadanziolide A, and yadanziosides G and B exert notable inhibitory effects on the MCF-7 human breast cancer cell line [35,37,49,83,84]. In the MDA-MB-231 cell line, brusatol, bruceantinol, bruceine A, and bruceantarin exhibited significant antitumor activity, with IC50 values ranging from 0.081 to 0.238 μmol/L [37]. Additionally, BJ hot water extracts induced apoptosis in the MDA-MB231 breast cancer cell line, with an IC50 around 50 μg/mL [56]. BJ ethanol extracts also demonstrated selective cytotoxicity and induced apoptosis at a concentration of 90 μg/mL [85]. A comprehensive analysis indicates that, compared to ethanol extracts, BJ water extracts demonstrate stronger inhibitory effects on the MDA-MB231 breast cancer cell line. In studies on cervical cancer, quassilactones A and B exhibited cytotoxic effects against HeLa cells, showing IC50 values of 78.95 ± 0.11 and 92.57 ± 0.13 μmol/L, respectively [86].

3.4. Anti-Leukemia

Leukemia represents a heterogeneous category of malignant blood disorders that arise from hematopoietic progenitor cells at different stages of hematopoietic maturation [87]. The etiology of leukemia involves genetic factors, environmental factors, and viral infections, typically triggered by monoclonal genetic or epigenetic abnormalities, but manifesting as a polyclonal disease. Bruceantin was first reported to exhibit anti-leukemia effects as early as 1973 [88]. Subsequently, the anti-leukemia effects of bruceantino, bruceantinoside A and B, brusatol, bruceanol A and B, and yadanzioside P were also documented [89,90,91,92,93]. In vitro experiments demonstrated that bruceoside C [50], yadanzioside G, N [94], bruceantinoside C [94], bruceanic acid D [95], and bruceanol C, D, E, and F [47,48] all exhibit potent cytotoxicity against P-388 lymphocytic leukemia cells, with ED50 values ranging from 0.16 to 7.49 μmol/L. Among these compounds, bruceanol D exhibited the highest safety profile. Additionally, reports indicate that javanicosides B, I, J, K, and L possess marked cytotoxic effects against P-388 murine leukemia cells, with IC50 values between 0.68 and 0.77 μmol/L [36,96]. Studies have shown that compounds including 17–18, (20R)-O-(3)-β-D-glucopyranosyl-(1→2)-α-L-arabinopyranosyl-pregn-5-en-3β, 20-diol, as well as brusatol, bruceines B, D, and E, yadanziolides A and C, yadanzigan, bruceoside A, and javanicolides B and S, display notable inhibitory effects on human promyelocytic leukemia (HL-60) cells [44,49,53]. Additionally, Liu [49] and colleagues demonstrated through in vitro experiments on the HL-60 cell line that brusatol and bruceine B exhibit significant inhibitory effects, with IC50 values of 0.06 and 0.27 μmol/L, respectively. Further studies indicate that brusatol and bruceantin have the potential to induce differentiation, inhibit proliferation, and exert differential cytotoxic effects across 11 leukemia cell lines. Experimental results indicate that brusatol and bruceantin exhibit lower cytotoxic responses in HL-60, K562, Kasumi-1, and Reh cell lines, but demonstrate extreme sensitivity in NB4, U937, BV173, SUPB13, RS4;11, Daudi, and DHL-6 cells, with significant cytotoxic effects [97]. In in vivo studies of P388 lymphocytic leukemia in mice, yadanzioside A, B, C, D, E, and G demonstrated significant antileukemic activity at a dose of 10 mg/kg, with an increased survival rate (ILS) ranging from 2.0% to 9.2% in experimental mice [98]. Yadanzioside P showed antileukemic effects at 5 and 10 mg/kg/day, elevating the ILS values of mice to 15.5% and 28.9%, respectively [93]. Similarly, Yadanzioside O exhibited activity at 2 and 4 mg/kg/day, with ILS increases of 37.1% and 47.2%, respectively [98]. As a monomer isolated from BJ, yadanzioside O provided the greatest survival benefit in animal studies.

3.5. Other Cancers

Beyond their pronounced antitumor effects in the tumor types discussed above, BJ compounds have been further investigated in the context of several other malignancies, such as oral cancer, nasopharyngeal carcinoma, glioma, renal cancer, medulloblastoma, and melanoma (Figure 2). Notably, as early as 1991, bruceantin, bruceolide, and bruceanic acid A were first identified by researchers [99]. Later investigations revealed that bruceoside C, bruceanol (D–G), and brujavanol (A, B, and E) displayed marked cytotoxic activity against human oral carcinoma cells, with IC50 values between 0.55 and 6.45 μmol/L [47,50,84,100]. Additionally, studies have demonstrated that BJ extract exhibits antiproliferative activity against KB and ORL-48 cells, with IC50 values of 24.37 ± 1.75 and 6.67 ± 1.15 μg/mL, respectively [101]. Based on the above findings, bruceanol G exhibits excellent cytotoxic activity in oral cancer. Bruceanol D, E, and F exhibited significant cytotoxic effects in TE-671 medulloblastoma cells, with ED50 values ranging from 0.14 to 0.22 μmol/L [47]. Further studies revealed that bruceanol demonstrates inhibitory activity against glioblastoma U87, U251, and MGG152 cells harboring gene mutations, particularly against IDH1-mutated U251 cells, with an IC50 value of approximately 20 nmol/L [102,103]. In investigations on head and neck squamous cell carcinoma (HNSCC) models, bruceanol markedly decreased cell viability, yielding IC50 values between 6 and 38 nmol/L across TU167, UMSCC47, UDSCC2, YD-10B, JMAR, FaDu, HN-9, and LN686 cell lines [104]. Bruceoside C, bruceanol D, E, and F [51] exhibited significant cytotoxic effects in melanoma cells (RPMI-7951), with ED50 below 0.15 μmol/L [47,50]. Additionally, bruceoside D, E, and F exhibit selective cytotoxicity against renal cancer cells, with log GI50 values ranging from −4.43 to −4.97 [51]. Among these, bruceanol D shows the lowest ED50, indicating a higher safety profile. Table 1 summarizes the IC50 values of various BJ compounds and extracts across different cancer cell lines, providing a clear comparison of their cytotoxic potencies.

4. Anticancer Mechanisms

4.1. Inducing Apoptosis

Apoptosis is a fundamental process in cancer pathophysiology and has a profound impact on the effectiveness of anticancer therapies. It can be triggered through two primary pathways: the intrinsic (mitochondrial) and extrinsic (death receptor) routes, both converging on the activation of the caspase family of cysteine proteases. Effector caspases, including caspase-3, -6, and -7, are responsible for executing the apoptotic program [106]. Multiple studies suggest that brusatol induces apoptosis primarily by decreasing mitochondrial membrane potential, increasing pro-apoptotic proteins such as Bax and Bak, and concurrently suppressing anti-apoptotic proteins including Bcl-2 and Bcl-xL, thereby facilitating cytochrome C release from mitochondria and activating the caspase cascade. This mitochondrial pathway induces apoptosis in various cell types, including pancreatic cancer (PANC-1, PATU-8988, Capan-2) [62,107], hepatocellular carcinoma (Bel-7404) [76], non-small cell lung cancer (PC9) [105], pituitary adenoma (GH3, MMQ) [108], nasopharyngeal carcinoma (CNE-1) [109], and head and neck squamous cell carcinoma (HNSCC, UD SCC2). In this pathway, caspase-9 activity and its downstream effector molecules (caspase-3, caspase-7, and poly (ADP-ribose) polymerase (PARP)) are activated. Concurrently, the expression levels of caspase-3 precursor (procaspase-3) and caspase-9 precursor (procaspase-9) decrease, while the levels of active caspase-3 (cleaved-caspase-3), caspase-8 (cleaved-caspase-8), and PARP (cleaved-PARP) significantly increase [73,76,109]. According to research, brusatol and bruceine D activate the JNK [110]/p38 MAPK [62] signaling pathways while suppressing the activation of Stat3/NF-κB, PI3K/Akt/mTOR [76], and PI3K/Akt/NF-κB [111] pathways, thereby inducing apoptosis in various tumor cells (Figure 3). These compounds reduce the protective effects of cancer cells by decreasing the activity of phosphorylated extracellular signal-regulated kinase (p-ERK), while simultaneously increasing the levels of phosphorylated p38 (p-P38) and phosphorylated c-Jun N-terminal kinase (p-JNK), thereby promoting the expression of pro-apoptotic proteins and inducing apoptosis [112]. In addition to regulating the aforementioned pathway proteins to induce apoptosis, bruceine D promotes apoptosis by suppressing the expression of microRNA-95 (miR-95), thereby upregulating the pro-apoptotic gene CUG triplet repeat RNA-binding protein 2 (CUGBP2) [113]. Furthermore, this compound downregulates P62 protein expression and upregulates microtubule-associated protein 1 light chain 3 (LC3) expression, thereby promoting apoptosis [76].

4.2. Inhibition of Cell Proliferation and Inducing Cell Cycle Arrest

In cancer treatment, inhibiting cell proliferation is one of the key strategies for anticancer therapy. Numerous anticancer drugs and treatments, including chemotherapy, targeted therapy, and immunotherapy, aim to control tumor growth and metastasis by suppressing tumor cell proliferation. However, the cell cycle, as the central process governing cell proliferation and division, is often dysregulated and is closely linked to cancer initiation, progression, and treatment [114]. The normal cell cycle consists of the G1, S, G2, and M phases, with regulation primarily depending on the precise control of cyclin proteins. Dysregulation of these regulatory mechanisms leads to uncontrolled cell cycle progression, resulting in unrestrained cell proliferation and ultimately tumor formation. As a result, cell cycle arrest has become a primary strategy for inhibiting tumor cell proliferation. Many anticancer drugs inhibit tumor growth by inducing cell cycle arrest. Research indicates that brusatol inhibits the Nrf2-Notch1 signaling pathway by downregulating Nrf2 and Notch1 protein expression, as well as their downstream target Hes1, thereby delaying cell proliferation [115]. The Cheng [116] research team demonstrated that bruceine D inhibits cell proliferation through the following five mechanisms: (1) promoting β-catenin protease cleavage, (2) inhibiting β-catenin protease and transcription factor-4 activity, (3) attenuating Wnt signaling pathway activity, (4) reducing jagged1 protein expression, and (5) inhibiting the Notch signaling pathway. Further research confirmed that bruceine D significantly reduces PI3K/Akt signaling pathway activity, inhibiting aerobic glycolysis [117]. This suppression targets hexokinase, phosphofructokinase, pyruvate kinase, and lactate dehydrogenase activity, thereby inhibiting cell proliferation. Crude BJEE and BJAE extracts induce cell cycle arrest by inhibiting the PI3K/Akt/mTOR signaling pathway [118] and elevating reactive oxygen species (ROS) levels in cancer cells [20], demonstrating their anticancer activity. Furthermore, BJAE induces EGFR expression to overcome drug resistance and promote cell cycle arrest at the sub-G1 phase [57]. Multiple experimental studies demonstrate that BJ extract significantly reduces the proportion of cells in the S phase while increasing the proportion of cells in the G0/G1 or sub-G1 phases. Reports indicate that brusatol, when combined with gemcitabine or 5-fluorouracil, induces G2/M phase cell cycle arrest in PANC-1 cells and enhances apoptotic effects [107]. Studies show that brusatol induces G1 phase arrest in melanoma cells [119]. In leukemia cell lines U937 and RS4:11, it induces G1 arrest and significantly increases the proportion of S phase cells, a phenomenon potentially associated with downregulation of c-Myc expression [97]. Brusatol induces breast cancer cells to arrest in sub-G0/G1 and G2/M phases by inhibiting Nrf2 [120]. In nasopharyngeal carcinoma studies, it induces G2/M phase cell cycle arrest by downregulating CyclinD1, Cdc2, and Cdc25c expression [109] (Figure 4).

4.3. Inhibition of Migration/Invasion

One of the hallmark features of many malignant tumors is their ability to migrate and invade [121]. Cancer cells detach from the primary tumor, invade the surrounding tissue, enter the circulatory system, and establish secondary tumor colonies in distant organs. The invasion–metastasis cascade is a multifaceted biological process, comprising several key stages: (1) local invasion of the basement membrane and cellular migration, (2) intravasation into blood vessels and/or the lymphatic system, (3) survival in the circulation, (4) arrest and extravasation at distant organs, and (5) colonization at the metastatic site [122,123]. Therefore, blocking cancer cell migration and invasion is considered a key strategy for cancer prevention and therapy. Studies indicate that BJ inhibits tumor cell invasion and metastasis through diverse mechanisms. First, BJ regulates epithelial–mesenchymal transition (EMT) in cancer cells, thereby diminishing their migratory and invasive abilities [124]. Specifically, Brucator inhibited the invasion and migration of liver tumors both in vivo and in vitro by modulating EMT, characterized by decreased N-cadherin and vimentin expression and increased E-cadherin expression [76]. Furthermore, Brucetin suppresses colorectal cancer cell metastasis by targeting the RhoA/ROCK1 signaling pathway and reversing EMT, as reflected in reduced expression of vimentin, N-cadherin, MMP2, and MMP9 proteins. In hepatocellular carcinoma, Brusatol reduced EMT markers such as fibronectin, vimentin, N-cadherin, Twist, and Snail, thereby suppressing STAT3-driven metastasis [125,126]. Second, angiogenesis is essential for cancer cell migration and metastasis. Vascular endothelial growth factor (VEGF), a downstream target of hypoxia-inducible factor 1 (HIF-1), is recognized as one of the most potent drivers of angiogenesis [127]. Brusatol induces the degradation of HIF-1α protein in HCT116 cells under hypoxic conditions, downregulates the expression of its downstream target VEGF, and consequently inhibits angiogenesis [70]. In gastric cancer studies, Brusatol reduces VEGF expression by inhibiting the Nrf2/HO-1 axis, thereby decreasing angiogenesis in cancer cells [128]. Third, multiple studies have reported that proteins such as transcription factor IIB-related factor 2 (BrF2) [129], insulin-like growth factor-binding protein 2 (IGFBP-2) [130], and CD151 [131] play regulatory roles in cancer cell growth, proliferation, migration, and invasion. The expression of proteins linked to metastasis is significantly diminished by brusatol in A549 cells, resulting in impaired migratory and invasive functions of cancer cells [132].

5. BJ Antitumor Clinical Preparations Research

Traditional Chinese medicine (TCM) has been employed in cancer therapy for centuries and has shown notable therapeutic outcomes [133,134]. The Chinese market has currently approved multiple BJO preparations for sale, including BJOE for injection, BJO soft capsules, BJO oral emulsion, and BJ pills, all of which have a wide range of clinical applications. These TCM formulations are primarily used as adjuvant therapies in cancer treatment, with their market sales showing a consistent upward trend. Considering factors such as market size, clinical application scope, and sales volume, the market performance of various formulations follows this hierarchy: BJOE injection > BJO soft capsules > BJO oral emulsion > other formulations. Current evidence on the safety and efficacy of BJOE and BJO in clinical settings is derived mainly from observational studies, underscoring the necessity for stronger evidence-based validation (Figure 5). Adverse reactions reported during clinical use include nausea, vomiting, liver damage, other digestive system injuries, pruritus, rash, skin lesions, and neurological impairments such as dizziness and headache [135,136]. These factors collectively limit the global promotion of BJO preparations. An analysis of the causes of adverse reactions associated with BJO preparations suggests potential toxicity from water-soluble saponins [137], as well as interactions between oleic acid in BJOE and solvents, which can lead to reduced formulation stability. Additionally, excipients like soy lecithin and glycerol can compromise the purity and stability of BJO and BJOE throughout manufacturing, storage, and utilization, thereby increasing the risk of adverse reactions. An additional concern is the excessive use of excipients, which complicates the detection of adulteration in formulations. Using electrospray ionization, researchers established an ultra-high-performance liquid chromatography–mass spectrometry (UHPLC-MS) method that enabled the identification of 69 components in BJO [138]. This technique not only effectively detects adulteration but also provides a solid theoretical foundation for establishing quality control standards for BJO.
TCM offers several advantages in multi-targeted therapy, including low drug resistance, verifiable efficacy, minimal side effects, and lower costs. Moreover, TCM effectively mitigates the toxic side effects of chemotherapy drugs. Current studies on BJO and BJOE have confirmed their cytotoxic activity against various tumor cell lines [139,140,141,142] (Figure 5). Researchers have conducted extensive investigations into the antitumor mechanisms of BJO and BJOE, which include the induction of apoptosis, disruption of the cell cycle, interference with cellular energy metabolism, inhibition of VEGF expression, suppression of tumor proliferation, reversal of drug resistance, enhancement of chemotherapy efficacy, reduction in drug toxicity and side effects, and delay of tumor progression [143,144,145,146]. Consequently, this drug not only inhibits and kills cancer cells but also enhances both cellular and humoral immunity without affecting normal cells [147].
Research indicates that BJOE significantly affects the survival rate, migration, and invasive capacity of esophageal cancer cells, while inhibiting their proliferation via the cyclin D1-CDK4/6 signaling pathway [148]. Additionally, BJOE exerts its antitumor effects by regulating the key factor MiR-8485, thereby modulating the LAMTOR3/mTOR/ATG13 signaling pathway to promote autophagy and apoptosis in ovarian cancer cells while inhibiting cell proliferation [146]. In investigating tumor autophagy, BJOE research has elucidated its mechanism of action in colorectal cancer cells. The study revealed that BJOE decreases LC3 protein expression, encompassing both LC3-I and LC3-II isoforms. Notably, in HCT116 colon cancer cells characterized by high basal autophagy, BJOE exerted dual actions by suppressing autophagy while simultaneously inducing apoptosis [141]. Another investigation demonstrated that BJOE reduced oncogene E6 expression in human cervical carcinoma SiHa cells in a dose-dependent fashion, thereby inducing apoptosis. In addition, BJOE markedly suppressed the growth of SiHa xenograft tumors, an effect likely mediated through modulation of the ERK/MAPK and NF-κB signaling cascades [149]. The study findings indicate that BJO significantly prolongs the survival of H22 ascites tumor-bearing mice. This effect is closely linked to the activation of miRNA-29b and p53-related apoptotic mechanisms, which involve downregulating Bcl-2 protein expression while upregulating Bax, Bad, and other protein levels, as well as the mitochondrial pathway, which reduces cytochrome C protein expression [150]. Su et al. [151]. identified a novel mechanism underlying BJO’s efficacy and safety profile. By modulating gut microbiota metabolism to alter the host’s amino acid composition, BJO activates mTOR and exhibits microbiota-dependent inhibition of MDA-MB-231 xenograft tumor growth in mice, with no toxicity observed in non-target organs. Researchers revealed the molecular mechanism by which BJO induces apoptosis in T24 bladder cancer cells through in vitro experiments [152]. The proposed mechanism appears to involve enhanced expression of caspase-3 and caspase-9 proteins, coupled with concurrent suppression of NF-κB and COX-2, ultimately leading to activation of the caspase-dependent apoptotic pathway. Additionally, modulation of p53 and cyclin D1 expression by BJO induces G0/G1 phase arrest in A549 and H446 cells, consequently suppressing the proliferation of these lung cancer cell lines. Subsequent investigations indicated that BJO may promote apoptosis in these two lung cancer cell lines by elevating ROS generation and initiating the mitochondrial caspase signaling cascade [142]. In leukemia, the mechanisms underlying the actions of BJO and BJOE have been partially clarified through experimental studies. These mechanisms include suppression of PI3K/Akt pathway activation and upregulation of downstream effectors such as p53, thereby triggering apoptotic processes [153]. Additionally, they downregulate c-FLIP (L/S), myeloid leukemia-1, Bcl-2, and XIAP expression, inducing apoptosis through mitochondrial and death receptor pathways [139].
However, evidence from multiple randomized controlled trials indicates that BJO is a promising adjunctive therapy for various cancers, particularly gastrointestinal tumors [154,155,156]. Meta-analyses further support this notion: BJOEI combined with conventional chemotherapy significantly increases the clinical total effective rate of gastric cancer patients, improves their physical condition, and effectively alleviates chemotherapy-related adverse reactions, such as gastrointestinal reactions, bone marrow suppression, and liver function impairment [26,157]. This suggests that BJOEI not only enhances the efficacy of chemotherapy but also improves patient tolerance and quality of life, thereby increasing overall treatment adherence. Moreover, other studies have evaluated the efficacy and safety of BJO as an adjuvant therapy for hepatocellular carcinoma. The results demonstrated that the addition of BJO to standard anticancer regimens may reduce the incidence and severity of chemotherapy- or surgery-related adverse events and improve patients’ clinical conditions and physiological parameters [158]. This evidence further highlights the favorable safety profile and therapeutic potential of BJO, offering clinicians a promising complementary option for the integrated management of liver cancer.
Overall, BJO appears to exert synergistic antitumor effects and toxicity-attenuating properties in the treatment of digestive system malignancies. Its mechanisms of action may involve the modulation of tumor cell apoptosis, inhibition of inflammatory responses, and enhancement of host immune function. Nevertheless, large-scale, multicenter, randomized controlled trials are still needed to further confirm its clinical efficacy and safety, as well as to elucidate its pharmacological mechanisms and pharmacokinetic characteristics. These efforts will provide a robust scientific foundation for the clinical translation and precise application of BJO and its derivatives in cancer therapy.

6. Conclusions and Outlook

Numerous studies indicate that traditional Chinese medicine has become a significant trend in the development of novel anticancer drugs. Brucea javanica, known for its heat-clearing and toxin-eliminating properties, has been utilized in traditional Chinese medicine for centuries. This herb not only demonstrates remarkable therapeutic effects against various tumors but also exhibits synergistic effects when combined with chemotherapy or radiotherapy. Consequently, it shows great promise for pharmaceutical development and clinical utilization, establishing itself as one of the most extensively applied herbal resources in traditional Chinese medicine. The present work seeks to deliver an extensive overview of the bioactive compounds, therapeutic potential, anticancer mechanisms, and clinical performance of BJ in tumor therapy. Through an in-depth analysis of existing literature and research, this study examines the bioactive components present in BJ, including alkaloids, berberidins, triterpenoids, steroids, phenylpropanoids, and flavonoids. These compounds demonstrate notable anticancer activity in experimental models, including both in vitro and in vivo systems. Moreover, this review investigates the possible anticancer mechanisms of BJ, covering diverse aspects such as induction of tumor cell apoptosis, modulation of the cell cycle, inhibition of DNA synthesis, suppression of proliferation, obstruction of angiogenesis, restriction of tumor growth, enhancement of immune responses, and reversal of multidrug resistance. BJ demonstrates significant antitumor activity by mediating multiple signaling pathways and regulating the expression of various proteins. Specifically, it inhibits proliferation, adhesion, invasion, and metastasis in various tumor cells by modulating signaling pathways such as JNK, HIF-1α, PI3K/Akt, Nrf2, Wnt, JAK2-STAT3, and EGFR. Moreover, through signaling pathways such as Nrf2, RhoA/ROCK, EGFR/PI3K/Akt, Bax, and PI3K/Akt/mTOR, BJ also suppresses tumor proliferation and metastasis. Simultaneously, it induces tumor cell apoptosis and inhibits proliferation through pathways such as PI3K/YAP1/TAZ, PI3K/Akt, EGFR, and NF-κB, demonstrating high safety. As a result, the clinical application of BJ is gradually expanding.
To date, the BJ preparations used in clinical practice remain predominantly BJOE and BJO, with no specific single effective component identified for clinical application. In clinical practice, BJ preparations are frequently administered alongside chemotherapeutic agents such as gemcitabine, 5-fluorouracil, carboplatin, gefitinib, and cisplatin. The diverse chemical composition of BJ preparations enables multifaceted approaches to combat tumor cells, making them applicable in the treatment of various cancers. Moreover, BJ preparations are increasingly used as adjuncts to radiotherapy and chemotherapy to reduce drug dosages, mitigate toxic side effects, and alleviate adverse reactions induced by these treatments. Future research should focus on the following areas: First, advanced methodologies—including artificial intelligence, organoid models, high-throughput screening, multi-omics approaches, and bioinformatics—should be employed to elucidate the anticancer pharmacological mechanisms of BJ, thereby constructing a comprehensive gene-signaling pathway framework to underpin clinical translation. Second, optimizing oral formulations by incorporating safer excipients with superior solubility can improve the physicochemical stability of both conventional and enteric-coated preparations while reducing the likelihood of adverse reactions. In clinical practice, there is an increasing demand for innovative dosage forms and diversified administration routes that can enhance patient compliance and therapeutic flexibility. Accordingly, future research should focus on developing advanced formulation strategies to improve the aqueous solubility and systemic bioavailability of BJ derivatives, which are currently constrained by poor solubility and extensive first-pass metabolism. Approaches such as nanoemulsion systems, solid lipid nanoparticles, and self-emulsifying drug delivery systems merit particular attention, as they can enhance intestinal absorption, prolong systemic circulation, and enable controlled or targeted drug release. Collectively, these formulation advancements are expected to improve in vivo efficacy, overcome existing pharmacokinetic limitations, and ultimately optimize the therapeutic potential of BJ-based anticancer agents. Third, BJ should undergo comprehensive preclinical and clinical studies in the future to confirm its anticancer mechanism, therapeutic effects, and safety profile. Particular emphasis should be placed on elucidating its pharmacokinetic and pharmacodynamic characteristics, as well as assessing its potential toxicity toward normal non-target cells and tissues to define a clear therapeutic window. Mechanistic studies are also required to clarify the pharmacological basis and structure–activity relationships of its active quassinoids and related compounds, thereby enabling rational optimization to enhance selectivity and minimize adverse effects. Furthermore, promising bioactive molecules should be isolated and subjected to systematic preclinical evaluation for efficacy and safety. In parallel, the development of advanced drug delivery systems—such as nanoparticle, liposomal, or targeted formulations—will be crucial for improving bioavailability, tumor specificity, and overall therapeutic performance. Fourth, current clinical studies primarily focus on short-term efficacy assessments, with limited long-term follow-up data. Future research should strengthen the establishment of long-term follow-up mechanisms to thoroughly assess patients’ long-term quality of life and tumor recurrence rates. Additionally, emphasis should be placed on developing new formulations of BJ and advancing their clinical translation processes to enhance their application value in cancer treatment. Current evidence suggests that BJ possesses considerable anticancer potential, highlighting its promising prospects for future applications in oncology. This review seeks to furnish researchers with a thorough reference for investigating the anticancer properties of BJ, while offering insights and theoretical guidance for advancing research and promoting the integration of traditional Chinese medicine into oncological practice. This will contribute to the development and integration of traditional Chinese medicine into the modern medical system.

Author Contributions

Conceptualization, W.X. and F.W.; methodology, J.L.; software, Y.Z.; validation, Y.Z., R.G. and W.X.; formal analysis, Y.Z.; investigation, R.G.; resources, H.Y.; data curation, R.G.; writing—original draft preparation, W.X.; writing—review and editing, W.X.; visualization, J.L.; supervision, Y.L.; project administration, Y.L.; funding acquisition, F.W. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Scientific Research Project of Traditional Chinese Medicine Supervision, China Academy of Chinese Medical Sciences (no. ZYJGKX202411).

Acknowledgments

We would like to thank the participants for their invaluable contributions.

Conflicts of Interest

The authors declare that they have no competing interest.

References

  1. Schwartz, S.M. Epidemiology of cancer. Clin. Chem. 2024, 70, 140–149. [Google Scholar] [CrossRef] [PubMed]
  2. Wu, S.; Zhu, W.; Thompson, P.; Hannun, Y.A. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 2018, 9, 3490. [Google Scholar] [CrossRef]
  3. Kalachaveedu, M.; Senthil, R.; Azhagiyamanavalan, S.; Ravi, R.; Meenakshisundaram, H.; Dharmarajan, A. Traditional medicine herbs as natural product matrices in cancer chemoprevention: A trans pharmacological perspective (scoping review). Phytother. Res. 2023, 37, 1539–1573. [Google Scholar] [CrossRef]
  4. Fu, B.; Wang, N.; Tan, H.Y.; Li, S.; Cheung, F.; Feng, Y. Multi-component herbal products in the prevention and treatment of chemotherapy-associated toxicity and side effects: A review on experimental and clinical evidences. Front. Pharmacol. 2018, 9, 1394. [Google Scholar] [CrossRef]
  5. Block, K.I.; Gyllenhaal, C.; Lowe, L.; Amedei, A.; Amin, A.; Amin, A.; Aquilano, K.; Arbiser, J.; Arreola, A.; Arzumanyan, A.; et al. Designing a broad-spectrum integrative approach for cancer prevention and treatment. Semin. Cancer Biol. 2015, 35 (Suppl.), S276–S304. [Google Scholar] [CrossRef]
  6. Hu, X.; Huang, W.; Fan, M. Emerging therapies for breast cancer. J. Hematol. Oncol. 2017, 10, 98. [Google Scholar] [CrossRef]
  7. Ren, Y.; Kinghorn, A.D. Development of potential antitumor agents from the scaffolds of plant-derived terpenoid lactones. J. Med. Chem. 2020, 63, 15410–15448. [Google Scholar] [CrossRef] [PubMed]
  8. Yang, L.; Chen, Y.; Liu, K.; Chen, Y.; Zhang, Y.; Zhang, Z.; Li, H. Investigating the immune mechanism of natural products in the treatment of lung cancer. Front. Pharmacol. 2024, 15, 1289957. [Google Scholar] [CrossRef] [PubMed]
  9. Atanasov, A.G.; Zotchev, S.B.; Dirsch, V.M.; Supuran, C.T. Natural products in drug discovery: Advances and opportunities. Nat. Rev. Drug Discov. 2021, 20, 200–216. [Google Scholar] [CrossRef]
  10. Chen, H.; Li, R.; Zhang, F.; Yao, Q.; Guo, Y. A scientometric visualization analysis for natural products on cancer research from 2008 to 2020. Front. Pharmacol. 2021, 12, 650141. [Google Scholar] [CrossRef]
  11. Huang, M.; Lu, J.J.; Ding, J. Natural products in cancer therapy: Past, present and future. Nat. Prod. Bioprospect. 2021, 11, 5–13. [Google Scholar] [CrossRef]
  12. Zhao, S.; Tang, Y.; Wang, R.; Najafi, M. Mechanisms of cancer cell death induction by paclitaxel: An updated review. Apoptosis 2022, 27, 647–667. [Google Scholar] [CrossRef]
  13. Ying, N.; Liu, S.; Zhang, M.; Cheng, J.; Luo, L.; Jiang, J.; Shi, G.; Wu, S.; Ji, J.; Su, H.; et al. Nano delivery system for paclitaxel: Recent advances in cancer theranostics. Colloids Surf. B Biointerfaces 2023, 228, 113419. [Google Scholar] [CrossRef] [PubMed]
  14. Chu, B.; Deng, H.; Niu, T.; Qu, Y.; Qian, Z. Stimulus-responsive nano-prodrug strategies for cancer therapy: A focus on camptothecin delivery. Small Methods 2024, 8, e2301271. [Google Scholar] [CrossRef]
  15. Wong, A.S.; Che, C.M.; Leung, K.W. Recent advances in ginseng as cancer therapeutics: A functional and mechanistic overview. Nat. Prod. Rep. 2015, 32, 256–272. [Google Scholar] [CrossRef] [PubMed]
  16. Hu, Z.; Yang, X.; Ho, P.C.; Chan, S.Y.; Heng, P.W.; Chan, E.; Duan, W.; Koh, H.L.; Zhou, S. Herb-drug interactions: A literature review. Drugs 2005, 65, 1239–1282. [Google Scholar] [CrossRef] [PubMed]
  17. Yu, X.Q.; Shang, X.Y.; Huang, X.X.; Yao, G.D.; Song, S.J. Brusatol: A potential anti-tumor quassinoid from Brucea javanica. Chin. Herb. Med. 2020, 12, 359–366. [Google Scholar] [CrossRef]
  18. Chen, J.; Yu, D.; Li, X.; Deng, Q.; Yang, H.; Chen, L.; Bai, L. A review of Brucea javanica: Metabolites, pharmacology and clinical application. Front. Pharmacol. 2023, 14, 1317620. [Google Scholar] [CrossRef]
  19. Tao, S.; Rojo de la Vega, M.; Chapman, E.; Ooi, A.; Zhang, D.D. The effects of NRF2 modulation on the initiation and progression of chemically and genetically induced lung cancer. Mol. Carcinog. 2018, 57, 182–192. [Google Scholar] [CrossRef]
  20. Bagheri, E.; Hajiaghaalipour, F.; Nyamathulla, S.; Salehen, N. The apoptotic effects of Brucea javanica fruit extract against HT29 cells associated with p53 upregulation and inhibition of NF-κB translocation. Drug Des. Dev. Ther. 2018, 12, 657–671. [Google Scholar] [CrossRef]
  21. Zhao, M.; Lau, S.T.; Leung, P.S.; Che, C.T.; Lin, Z.X. Seven quassinoids from Fructus Bruceae with cytotoxic effects on pancreatic adenocarcinoma cell lines. Phytother. Res. 2011, 25, 1796–1800. [Google Scholar] [CrossRef] [PubMed]
  22. Nan, X.; He, D.; Dang, J.; Al, E. Treatment of prostatic carcinoma (stage C to D) with fructus bruceae emulsion. J. Mod. Urol. 1998, 68–71. [Google Scholar]
  23. Shan, G.Y.; Zhang, S.; Li, G.W.; Chen, Y.S.; Liu, X.A.; Wang, J.K. Clinical evaluation of oral Fructus bruceae oil combined with radiotherapy for the treatment of esophageal cancer. Chin. J. Integr. Med. 2011, 17, 933–936. [Google Scholar] [CrossRef] [PubMed]
  24. Ji, Z.Q.; Huang, X.E.; Wu, X.Y.; Liu, J.; Wang, L.; Tang, J.H. Safety of Brucea javanica and cantharidin combined with chemotherapy for treatment of NSCLC patients. Asian Pac. J. Cancer Prev. 2014, 15, 8603–8605. [Google Scholar] [CrossRef]
  25. Xu, W.; Jiang, X.; Xu, Z.; Ye, T.; Shi, Q. The Efficacy of Brucea javanica oil emulsion injection as adjunctive therapy for advanced non-small-cell lung cancer: A meta-analysis. Evid. Based Complement. Altern. Med. 2016, 2016, 5928562. [Google Scholar] [CrossRef]
  26. Wang, X.; Wang, H.; Cao, L.; Wu, J.; Lu, T.; Li, S.; Li, J. Efficacy and safety of Brucea javanica oil emulsion injection in the treatment of gastric cancer: A systematic review and meta-analysis. Front. Nutr. 2021, 8, 784164. [Google Scholar] [CrossRef]
  27. Rohilla, S.; Dureja, H.; Chawla, V. Cytoprotective agents to avoid chemotherapy induced sideeffects on normal cells: A review. Curr. Cancer Drug Targets 2019, 19, 765–781. [Google Scholar] [CrossRef]
  28. Zhao, H.Y.; Zhou, H.Y.; Wang, Y.T.; Chen, W.; Qi, S.Y.; Cao, J.L.; Li, G.H. Assessment on the efficacy and safety of Aidi injection combined with vinorelbine and cisplatin for treatment of advanced nonsmall cell lung cancer. Chin. Med. J. 2016, 129, 723–730. [Google Scholar] [CrossRef]
  29. Chen, H.; Bai, J.; Fang, Z.-F.; Yu, S.-S.; Ma, S.-G.; Xu, S.; Li, Y.; Qu, J.; Ren, J.-H.; Li, L.; et al. Indole alkaloids and quassinoids from the stems of Brucea mollis. J. Nat. Prod. 2011, 74, 2438–2445. [Google Scholar] [CrossRef]
  30. Chen, Q.J.; Ouyang, M.A.; Tan, Q.W.; Zhang, Z.K.; Wu, Z.J.; Lin, Q.Y. Constituents from the seeds of Brucea javanica with inhibitory activity of Tobacco mosaic virus. J. Asian Nat. Prod. Res. 2009, 11, 539–547. [Google Scholar] [CrossRef]
  31. Hong-Wu, W.; Yan-Qing, L.; Zi-Jun, Y.; Shou-Lian, W.; Jin-Qing, Y.E. A gas chromatography-mass spectrometry analysis of the essential oils from Brucea javanica extracted with different methods. Fine Chem. 2011, 28, 668–670. [Google Scholar]
  32. Liu, J.H.; Jin, H.Z.; Zhang, W.D.; Yan, S.K.; Shen, Y.H. Chemical constituents of plants from the genus Brucea. Chem. Biodivers. 2009, 6, 57–70. [Google Scholar] [CrossRef] [PubMed]
  33. Chen, M.; Chen, R.; Wang, S.; Tan, W.; Hu, Y.; Peng, X.; Wang, Y. Chemical components, pharmacological properties, and nanoparticulate delivery systems of Brucea javanica. Int. J. Nanomed. 2013, 8, 85–92. [Google Scholar] [CrossRef] [PubMed]
  34. Cuendet, M.; Gills, J.J.; Pezzuto, J.M. Brusatol-induced HL-60 cell differentiation involves NF-kappaB activation. Cancer Lett. 2004, 206, 43–50. [Google Scholar] [CrossRef] [PubMed]
  35. Su, Z.; Hao, J.; Xu, Z.; Huang, R.; Zhang, N.; Qiu, S. A new quassinoid from fruits of Brucea javanica. Nat. Prod. Res. 2013, 27, 2016–2021. [Google Scholar] [CrossRef]
  36. Kim, I.H.; Takashima, S.; Hitotsuyanagi, Y.; Hasuda, T.; Takeya, K. New quassinoids, javanicolides C and D and javanicosides B--F, from seeds of Brucea javanica. J. Nat. Prod. 2004, 67, 863–868. [Google Scholar] [CrossRef]
  37. Ye, Q.M.; Bai, L.L.; Hu, S.Z.; Tian, H.Y.; Ruan, L.J.; Tan, Y.F.; Hu, L.P.; Ye, W.C.; Zhang, D.M.; Jiang, R.W. Isolation, chemotaxonomic significance and cytotoxic effects of quassinoids from Brucea javanica. Fitoterapia 2015, 105, 66–72. [Google Scholar] [CrossRef]
  38. Duan, Z.K.; Zhang, Z.J.; Dong, S.H.; Wang, Y.X.; Song, S.J.; Huang, X.X. Quassinoids: Phytochemistry and antitumor prospect. Phytochemistry 2021, 187, 112769. [Google Scholar] [CrossRef]
  39. Li, Z.; Ruan, J.Y.; Sun, F.; Yan, J.J.; Wang, J.L.; Zhang, Z.X.; Zhang, Y.; Wang, T. Relationship between structural characteristics and plant sources along with pharmacology research of quassinoids. Chem. Pharm. Bull. 2019, 67, 654–665. [Google Scholar] [CrossRef]
  40. Zhou, Z.; Shi, R.; Liu, B.; Zou, J.; Wang, L.; Xia, J. Quantitative determination of contents of three components in Brucea javanica by HPLC. Zhongguo Zhong Yao Za Zhi 2011, 36, 1979–1981. [Google Scholar]
  41. Hu, S.Z.; Jin, L.; Yu, T.; Tian, H.Y.; Jiang, R.W. Brusatol. Acta Crystallogr. Sect. E Crystallogr. Commun. 2012, 68, o1592–o1593. [Google Scholar] [CrossRef] [PubMed]
  42. Fukamiya, N.; Lee, K.H.; Muhammad, I.; Murakami, C.; Okano, M.; Harvey, I.; Pelletier, J. Structure-activity relationships of quassinoids for eukaryotic protein synthesis. Cancer Lett. 2005, 220, 37–48. [Google Scholar] [CrossRef] [PubMed]
  43. Hitotsuyanagi, Y.; Kim, I.H.; Hasuda, T.; Yamauchi, Y.; Takeya, K. A structure–activity relationship study of brusatol, an antitumor quassinoid. Tetrahedron 2006, 62, 4262–4271. [Google Scholar] [CrossRef]
  44. Luyengi, L.; Suh, N.; Fong, H.H.; Pezzuto, J.M.; Kinghorn, A.D. A lignan and four terpenoids from Brucea javanica that induce differentiation with cultured HL-60 promyelocytic leukemia cells. Phytochemistry 1996, 43, 409–412. [Google Scholar] [CrossRef]
  45. Smolarz, B.; Łukasiewicz, H.; Samulak, D.; Piekarska, E.; Kołaciński, R.; Romanowicz, H. Lung Cancer-epidemiology, pathogenesis, treatment and molecular aspect (review of literature). Int. J. Mol. Sci. 2025, 26, 2049. [Google Scholar] [CrossRef]
  46. Shalata, W.; Naamneh, R.; Najjar, W.; Asla, M.; Abu Gameh, A.; Abu Amna, M.; Saiegh, L.; Agbarya, A. Current and emerging therapeutic strategies for limited- and extensive-stage small-cell lung cancer. Med. Sci. 2025, 13, 142. [Google Scholar] [CrossRef] [PubMed]
  47. Imamura, K.; Fukamiya, N.; Okano, M.; Tagahara, K.; Lee, K.H. Bruceanols D, E, and F three new cytotoxic quassinoids from Brucea antidysenterica. J. Nat. Prod. 1993, 56, 2091–2097. [Google Scholar] [CrossRef]
  48. Fukamiya, N.; Okano, M.; Tagahara, K.; Aratani, T.; Lee, K.H. Antitumor agents, 93. Bruceanol C, a new cytotoxic quassinoid from Brucea antidysenterica. J. Nat. Prod. 1988, 51, 349–352. [Google Scholar] [CrossRef]
  49. Liu, J.Q.; Wang, C.F.; Li, X.Y.; Chen, J.C.; Li, Y.; Qiu, M.H. One new pregnane glycoside from the seeds of cultivated Brucea javanica. Arch. Pharm. Res. 2011, 34, 1297–1300. [Google Scholar] [CrossRef]
  50. Fukamiya, N.; Okano, M.; Miyamoto, M.; Tagahara, K.; Lee, K.H. Antitumor agents, 127. Bruceoside C, a new cytotoxic quassinoid glucoside, and related compounds from Brucea javanica. J. Nat. Prod. 1992, 55, 468–475. [Google Scholar] [CrossRef]
  51. Ohnishi, S.; Fukamiya, N.; Okano, M.; Tagahara, K.; Lee, K.H. Bruceosides D, E, and F, three new cytotoxic quassinoid glucosides from Brucea javanica. J. Nat. Prod. 1995, 58, 1032–1038. [Google Scholar] [CrossRef]
  52. Chen, H.; Ma, S.G.; Fang, Z.F.; Bai, J.; Yu, S.S.; Chen, X.G.; Hou, Q.; Yuan, S.P.; Chen, X. Tirucallane triterpenoids from the stems of Brucea mollis. Chem. Biodivers. 2013, 10, 695–702. [Google Scholar] [CrossRef] [PubMed]
  53. Su, B.N.; Chang, L.C.; Park, E.J.; Cuendet, M.; Santarsiero, B.D.; Mesecar, A.D.; Mehta, R.G.; Fong, H.H.; Pezzuto, J.M.; Kinghorn, A.D. Bioactive constituents of the seeds of Brucea javanica. Planta Med. 2002, 68, 730–733. [Google Scholar] [CrossRef]
  54. Tan, B.; Huang, Y.; Lan, L.; Zhang, B.; Ye, L.; Yan, W.; Wang, F.; Lin, N. Bruceine D induces apoptosis in human non-small cell lung cancer cells through regulating JNK pathway. Biomed. Pharmacother. 2019, 117, 109089. [Google Scholar] [CrossRef] [PubMed]
  55. Su, Z.; Huang, H.; Li, J.; Zhu, Y.; Huang, R.; Qiu, S.X. Chemical composition and cytotoxic activities of petroleum ether fruit extract of fruits of Brucea javanica (Simarubaceae). Trop. J. Pharm. Res. 2013, 12, 735–742. [Google Scholar] [CrossRef]
  56. Lau, F.Y.; Chui, C.H.; Gambari, R.; Kok, S.H.; Kan, K.L.; Cheng, G.Y.; Wong, R.S.; Teo, I.T.; Cheng, C.H.; Wan, T.S.; et al. Antiproliferative and apoptosis-inducing activity of Brucea javanica extract on human carcinoma cells. Int. J. Mol. Med. 2005, 16, 1157–1162. [Google Scholar] [CrossRef]
  57. Kim, S.H.; Liu, C.Y.; Fan, P.W.; Hsieh, C.H.; Lin, H.Y.; Lee, M.C.; Fang, K. The aqueous extract of Brucea javanica suppresses cell growth and alleviates tumorigenesis of human lung cancer cells by targeting mutated epidermal growth factor receptor. Drug Des. Dev. Ther. 2016, 10, 3599–3609. [Google Scholar] [CrossRef]
  58. Makong, Y.S.; Mouthé Happi, G.; Djouaka Bavoua, J.L.; Wansi, J.D.; Nahar, L.; Kamdem Waffo, A.F.; Martin, C.; Sewald, N.; Sarker, S.D. Cytotoxic stilbenes and canthinone alkaloids from Brucea antidysenterica (Simaroubaceae). Molecules 2019, 24, 4412. [Google Scholar] [CrossRef] [PubMed]
  59. Ren, D.; Villeneuve, N.F.; Jiang, T.; Wu, T.; Lau, A.; Toppin, H.A.; Zhang, D.D. Brusatol enhances the efficacy of chemotherapy by inhibiting the Nrf2-mediated defense mechanism. Proc. Natl. Acad. Sci. USA 2011, 108, 1433–1438. [Google Scholar] [CrossRef]
  60. Stoop, T.F.; Javed, A.A.; Oba, A.; Koerkamp, B.G.; Seufferlein, T.; Wilmink, J.W.; Besselink, M.G. Pancreatic cancer. Lancet 2025, 405, 1182–1202. [Google Scholar] [CrossRef]
  61. Connor, A.A.; Gallinger, S. Pancreatic cancer evolution and heterogeneity: Integrating omics and clinical data. Nat. Rev. Cancer 2022, 22, 131–142. [Google Scholar] [CrossRef] [PubMed]
  62. Xiang, Y.; Ye, W.; Huang, C.; Lou, B.; Zhang, J.; Yu, D.; Huang, X.; Chen, B.; Zhou, M. Brusatol inhibits growth and induces apoptosis in pancreatic cancer cells via JNK/p38 MAPK/NF-κb/Stat3/Bcl-2 signaling pathway. Biochem. Biophys. Res. Commun. 2017, 487, 820–826. [Google Scholar] [CrossRef] [PubMed]
  63. Lau, S.T.; Lin, Z.X.; Liao, Y.; Zhao, M.; Cheng, C.H.; Leung, P.S. Bruceine D induces apoptosis in pancreatic adenocarcinoma cell line PANC-1 through the activation of p38-mitogen activated protein kinase. Cancer Lett. 2009, 281, 42–52. [Google Scholar] [CrossRef]
  64. Liu, L.; Lin, Z.X.; Leung, P.S.; Chen, L.H.; Zhao, M.; Liang, J. Involvement of the mitochondrial pathway in bruceine D-induced apoptosis in Capan-2 human pancreatic adenocarcinoma cells. Int. J. Mol. Med. 2012, 30, 93–99. [Google Scholar] [CrossRef]
  65. Zhang, P.; Tao, W.; Lu, C.; Fan, L.; Jiang, Q.; Yang, C.; Shang, E.; Cheng, H.; Che, C.; Duan, J.; et al. Bruceine A induces cell growth inhibition and apoptosis through PFKFB4/GSK3β signaling in pancreatic cancer. Pharmacol. Res. 2021, 169, 105658. [Google Scholar] [CrossRef] [PubMed]
  66. Abedizadeh, R.; Majidi, F.; Khorasani, H.R.; Abedi, H.; Sabour, D. Colorectal cancer: A comprehensive review of carcinogenesis, diagnosis, and novel strategies for classified treatments. Cancer Metastasis Rev. 2024, 43, 729–753. [Google Scholar] [CrossRef]
  67. Osumi, H.; Shinozaki, E.; Yamaguchi, K.; Zembutsu, H. Clinical utility of circulating tumor DNA for colorectal cancer. Cancer Sci. 2019, 110, 1148–1155. [Google Scholar] [CrossRef]
  68. Liu, J.H.; Zhao, N.; Zhang, G.J.; Yu, S.S.; Wu, L.J.; Qu, J.; Ma, S.G.; Chen, X.G.; Zhang, T.Q.; Bai, J.; et al. Bioactive quassinoids from the seeds of Brucea javanica. J. Nat. Prod. 2012, 75, 683–688. [Google Scholar] [CrossRef]
  69. Oh, E.T.; Kim, C.W.; Kim, H.G.; Lee, J.S.; Park, H.J. Brusatol-mediated inhibition of c-Myc increases HIF-1α degradation and causes cell death in colorectal cancer under hypoxia. Theranostics 2017, 7, 3415–3431. [Google Scholar] [CrossRef]
  70. Lu, Y.; Wang, B.; Shi, Q.; Wang, X.; Wang, D.; Zhu, L. Brusatol inhibits HIF-1 signaling pathway and suppresses glucose uptake under hypoxic conditions in HCT116 cells. Sci. Rep. 2016, 6, 39123. [Google Scholar] [CrossRef]
  71. Evans, J.P.; Winiarski, B.K.; Sutton, P.A.; Jones, R.P.; Ressel, L.; Duckworth, C.A.; Pritchard, D.M.; Lin, Z.X.; Fretwell, V.L.; Tweedle, E.M.; et al. The Nrf2 inhibitor brusatol is a potent antitumour agent in an orthotopic mouse model of colorectal cancer. Oncotarget 2018, 9, 27104–27116. [Google Scholar] [CrossRef]
  72. Bagheri, E.; Hajiaghaalipour, F.; Nyamathulla, S.; Salehen, N.A. Ethanolic extract of Brucea javanica inhibit proliferation of HCT-116 colon cancer cells via caspase activation. RSC Adv. 2018, 8, 681–689. [Google Scholar] [CrossRef] [PubMed]
  73. Chen, H.M.; Lai, Z.Q.; Liao, H.J.; Xie, J.H.; Xian, Y.F.; Chen, Y.L.; Ip, S.P.; Lin, Z.X.; Su, Z.R. Synergistic antitumor effect of brusatol combined with cisplatin on colorectal cancer cells. Int. J. Mol. Med. 2018, 41, 1447–1454. [Google Scholar] [CrossRef]
  74. Donne, R.; Lujambio, A. The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma. Hepatology 2023, 77, 1773–1796. [Google Scholar] [CrossRef]
  75. Gao, S.; Jiang, X.; Wang, L.; Jiang, S.; Luo, H.; Chen, Y.; Peng, C. The pathogenesis of liver cancer and the therapeutic potential of bioactive substances. Front. Pharmacol. 2022, 13, 1029601. [Google Scholar] [CrossRef]
  76. Ye, R.; Dai, N.; He, Q.; Guo, P.; Xiang, Y.; Zhang, Q.; Hong, Z.; Zhang, Q. Comprehensive anti-tumor effect of Brusatol through inhibition of cell viability and promotion of apoptosis caused by autophagy via the PI3K/Akt/mTOR pathway in hepatocellular carcinoma. Biomed. Pharmacother. 2018, 105, 962–973. [Google Scholar] [CrossRef]
  77. Chen, J.H.; Kim, S.H.; Fan, P.W.; Liu, C.Y.; Hsieh, C.H.; Fang, K. The aqueous extract of Chinese medicinal herb Brucea javanica suppresses the growth of human liver cancer and the derived stem-like cells by apoptosis. Drug Des. Dev. Ther. 2016, 10, 2003–2013. [Google Scholar] [CrossRef]
  78. Smyth, E.C.; Nilsson, M.; Grabsch, H.I.; van Grieken, N.C.; Lordick, F. Gastric cancer. Lancet 2020, 396, 635–648. [Google Scholar] [CrossRef] [PubMed]
  79. Konstantinopoulos, P.A.; Matulonis, U.A. Clinical and translational advances in ovarian cancer therapy. Nat. Cancer 2023, 4, 1239–1257. [Google Scholar] [CrossRef] [PubMed]
  80. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA A Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  81. Zhang, D.; Li, S.; Zhang, X.; Peng, J.; Zhang, S. What predicts the clinical benefits of PARP inhibitors in platinum-sensitive recurrent ovarian cancer: A real-world single-center retrospective cohort study from China. Front. Oncol. 2022, 12, 955124. [Google Scholar] [CrossRef]
  82. Qi, Y.J.; Su, G.H.; You, C.; Zhang, X.; Xiao, Y.; Jiang, Y.Z.; Shao, Z.M. Radiomics in breast cancer: Current advances and future directions. Cell Rep. Med. 2024, 5, 101719. [Google Scholar] [CrossRef]
  83. Pan, L.; Chin, Y.W.; Chai, H.B.; Ninh, T.N.; Soejarto, D.D.; Kinghorn, A.D. Bioactivity-guided isolation of cytotoxic constituents of Brucea javanica collected in Vietnam. Bioorganic Med. Chem. 2009, 17, 2219–2224. [Google Scholar] [CrossRef]
  84. Chumkaew, P.; Srisawat, T. Antimalarial and cytotoxic quassinoids from the roots of Brucea javanica. J. Asian Nat. Prod. Res. 2017, 19, 247–253. [Google Scholar] [CrossRef] [PubMed]
  85. Haryanti, S.; Sholikhah, I.Y.M.; Widiyastuti, Y. Cytotoxic and anti-migratory effects of Brucea javanica fruits extract on 4T1 breast cancer cells. AIP Conf. Proc. 2019, 2202, 020097. [Google Scholar] [CrossRef]
  86. Su, Z.; Ma, Z.; Liu, K.; Li, T.; Zhou, B. Quassilactones A and B, structural characterization of a new class of norquassinoids from Brucea javanica. J. Nat. Med. 2020, 74, 599–605. [Google Scholar] [CrossRef]
  87. Ciantra, Z.; Paraskevopoulou, V.; Aifantis, I. The rewired immune microenvironment in leukemia. Nat. Immunol. 2025, 26, 351–365. [Google Scholar] [CrossRef] [PubMed]
  88. Kupchan, S.M.; Britton, R.W.; Ziegler, M.F.; Sigel, C.W. Bruceantin, a new potent antileukemic simaroubolide from Brucea antidysenterica. J. Org. Chem. 1973, 38, 178–179. [Google Scholar] [CrossRef]
  89. Kupchan, S.M.; Britton, R.W.; Lacadie, J.A.; Ziegler, M.F.; Sigel, C.W. Tumor inhibitors. 100. Isolation and structural elucidation of bruceantin and bruceantinol, new potent antileukemic quassinoids from Brucea antidysenterica. J. Org. Chem. 1975, 40, 648–654. [Google Scholar] [CrossRef]
  90. Okano, M.; Lee, K.H.; Hall, I.H.; Boettner, F.E. Antitumor agents. 39. Bruceantinoside-A and -B, novel antileukemic quassinoid glucosides from Brucea antidysenterica. J. Nat. Prod. 1981, 44, 470–474. [Google Scholar] [CrossRef]
  91. Lee, K.H.; Hayashi, N.; Okano, M.; Nozaki, H.; Ju-Ichi, M. Antitumor agents, 65. Brusatol and cleomiscosin-A, antileukemic principles from Brucea javanica. J. Nat. Prod. 1984, 47, 550–551. [Google Scholar] [CrossRef] [PubMed]
  92. Okano, M.; Fukamiya, N.; Aratani, T.; Juichi, M.; Lee, K.H. Antitumor agents, 74. Bruceanol-A and -B, two new antileukemic quassinoids from Brucea antidysenterica. J. Nat. Prod. 1985, 48, 972–975. [Google Scholar] [CrossRef] [PubMed]
  93. Sakaki, T.; Yoshimura, S.; Tsuyuki, T.; Takahashi, T.; Honda, T. Yadanzioside P, a new antileukemic quassinoid glycoside from Brucea javanica (L.) Merr with the 3-O-(beta-D-glucopyranosyl) bruceantin structure. Chem. Pharm. Bull. 1986, 34, 4447–4450. [Google Scholar] [CrossRef]
  94. Fukamiya, N.; Okano, M.; Tagahara, K.; Aratani, T.; Muramoto, Y.; Lee, K.H. Antitumor agents, 90. Bruceantinoside C, a new cytotoxic quassinoid glycoside from Brucea antidysenterica. J. Nat. Prod. 1987, 50, 1075–1079. [Google Scholar] [CrossRef]
  95. Toyota, T.; Fukamiya, N.; Okano, M.; Tagahara, K.; Chang, J.J.; Lee, K.H. Antitumor agents, 118. The isolation and characterization of bruceanic acid A, its methyl ester, and the new bruceanic acids B, C, and D, from Brucea antidysenterica. J. Nat. Prod. 1990, 53, 1526–1532. [Google Scholar] [CrossRef]
  96. Kim, I.H.; Hitotsuyanagi, Y.; Takeya, K. Quassinoid glucosides from seeds of Brucea amarissima. Phytochemistry 2004, 65, 3167–3173. [Google Scholar] [CrossRef] [PubMed]
  97. Mata-Greenwood, E.; Cuendet, M.; Sher, D.; Gustin, D.; Stock, W.; Pezzuto, J.M. Brusatol-mediated induction of leukemic cell differentiation and G(1) arrest is associated with down-regulation of c-myc. Leukemia 2002, 16, 2275–2284. [Google Scholar] [CrossRef]
  98. Sakaki, T.; Yoshimura, S.; Ishibashi, M.; Tsuyuki, T.; Takahashi, T.; Honda, T.; Nakanishi, T. Structures of new quassinoid glycosides, yadanziosides A, B, C, D, E, G, H, and new quassinoids, dehydrobrusatol and dehydrobruceantinol from Brucea javanica (L.) Merr. Bull. Chem. Soc. Jpn. 1985, 58, 2680–2686, Erratum in Bull. Chem. Soc. Jpn. 1986, 59, 3541–3546. [Google Scholar] [CrossRef]
  99. Anderson, M.M.; O’Neill, M.J.; Phillipson, J.D.; Warhurst, D.C. In vitro cytotoxicity of a series of quassinoids from Brucea javanica fruits against KB cells. Planta Med. 1991, 57, 62–64. [Google Scholar] [CrossRef]
  100. Chumkaew, P.; Srisawat, T. New neolignans from the seeds of Myristica fragrans and their cytotoxic activities. J. Nat. Med. 2019, 73, 273–277. [Google Scholar] [CrossRef]
  101. Majid, M.Z.; Zaini, Z.M.; Razak, F.A. Apoptosis-inducing effect of three medicinal plants on oral cancer cells KB and ORL-48. Sci. World J. 2014, 2014, 125353. [Google Scholar] [CrossRef]
  102. Liu, X.; Xu, H.; Zhang, Y.; Wang, P.; Gao, W. Brusatol inhibits amyloid-β-induced neurotoxicity in U-251 cells via regulating the Nrf2/HO-1 pathway. J. Cell. Biochem. 2019, 120, 10556–10563. [Google Scholar] [CrossRef]
  103. Tang, X.; Fu, X.; Liu, Y.; Yu, D.; Cai, S.J.; Yang, C. Blockade of glutathione metabolism in IDH1-mutated glioma. Mol. Cancer Ther. 2020, 19, 221–230. [Google Scholar] [CrossRef]
  104. Lee, J.; Mawla, I.; Kim, J.; Loggia, M.L.; Ortiz, A.; Jung, C.; Chan, S.T.; Gerber, J.; Schmithorst, V.J.; Edwards, R.R.; et al. Machine learning-based prediction of clinical pain using multimodal neuroimaging and autonomic metrics. Pain 2019, 160, 550–560. [Google Scholar] [CrossRef]
  105. Xie, J.; Lai, Z.; Zheng, X.; Liao, H.; Xian, Y.; Li, Q.; Wu, J.; Ip, S.; Xie, Y.; Chen, J.; et al. Apoptotic activities of brusatol in human non-small cell lung cancer cells: Involvement of ROS-mediated mitochondrial-dependent pathway and inhibition of Nrf2-mediated antioxidant response. Toxicology 2021, 451, 152680. [Google Scholar] [CrossRef]
  106. Moyer, A.; Tanaka, K.; Cheng, E.H. Apoptosis in cancer biology and therapy. Annu. Rev. Pathol. Mech. Dis. 2025, 20, 303–328. [Google Scholar] [CrossRef] [PubMed]
  107. Lu, Z.; Lai, Z.Q.; Leung, A.W.N.; Leung, P.S.; Li, Z.S.; Lin, Z.X. Exploring brusatol as a new anti-pancreatic cancer adjuvant: Biological evaluation and mechanistic studies. Oncotarget 2017, 8, 84974–84985. [Google Scholar] [CrossRef]
  108. Wu, Z.; Xu, Y.; Xu, J.; Lu, J.; Cai, L.; Li, Q.; Wang, C.; Su, Z. Brusatol inhibits tumor growth and increases the efficacy of cabergoline against pituitary adenomas. Oxidative Med. Cell. Longev. 2021, 2021, 6696015. [Google Scholar] [CrossRef] [PubMed]
  109. Guo, S.; Zhang, J.; Wei, C.; Lu, Z.; Cai, R.; Pan, D.; Zhang, H.; Liang, B.; Zhang, Z. Anticancer effects of brusatol in nasopharyngeal carcinoma through suppression of the Akt/mTOR signaling pathway. Cancer Chemother. Pharmacol. 2020, 85, 1097–1108. [Google Scholar] [CrossRef] [PubMed]
  110. Yang, J.; Li, S.; Xie, C.; Ye, H.; Tang, H.; Chen, L.; Peng, A. Anti-inflammatory activity of ethyl acetate fraction of the seeds of Brucea javanica. J. Ethnopharmacol. 2013, 147, 442–446. [Google Scholar] [CrossRef] [PubMed]
  111. Chen, H.; Jiang, T.; Chen, H.; Su, J.; Wang, X.; Cao, Y.; Li, Q. Brusatol reverses lipopolysaccharide-induced epithelial-mesenchymal transformation and induces apoptosis through PI3K/Akt/NF-κB pathway in human gastric cancer SGC-7901 cells. Anticancer. Drugs 2021, 32, 394–404. [Google Scholar] [CrossRef]
  112. Ya-Fang, T.; Juan, L.I.; Shu-Zhi, H.U.; Ren-Wang, J. Inhibitory effects of brusatol on human prostate cancer cells DU145 and its molecular mechanism. Guihaia 2015, 35, 431–436. [Google Scholar]
  113. Xiao, Z.; Chow, S.C.; Li, C.H.; Tang, S.C.; Tsui, S.K.W.; Lin, Z.; Chen, Y. Role of microRNA-95 in the anticancer activity of Brucein D in hepatocellular carcinoma. Eur. J. Pharmacol. 2014, 728, 141–150. [Google Scholar] [CrossRef]
  114. Glaviano, A.; Singh, S.K.; Lee, E.H.C.; Okina, E.; Lam, H.Y.; Carbone, D.; Reddy, E.P.; O’Connor, M.J.; Koff, A.; Singh, G.; et al. Cell cycle dysregulation in cancer. Pharmacol. Rev. 2025, 77, 100030. [Google Scholar] [CrossRef]
  115. Min, W.; Qili, L.; Shuai, X.U.; Pinyu, L.I.; Juntao, L.I. Study on the mechanism of the influence of Brusatol for the Nrf2-Notch1 axis of non-small cell lung cancer. China Med. Her. 2018, 15, 16–19. [Google Scholar]
  116. Cheng, Z.; Yuan, X.; Qu, Y.; Li, X.; Wu, G.; Li, C.; Zu, X.; Yang, N.; Ke, X.; Zhou, J.; et al. Bruceine D inhibits hepatocellular carcinoma growth by targeting β-catenin/jagged1 pathways. Cancer Lett. 2017, 403, 195–205. [Google Scholar] [CrossRef]
  117. Luo, C.; Wang, Y.; Wei, C.; Chen, Y.; Ji, Z. The anti-migration and anti-invasion effects of Bruceine D in human triple-negative breast cancer MDA-MB-231 cells. Exp. Ther. Med. 2020, 19, 273–279. [Google Scholar] [CrossRef]
  118. Chen, X.; Li, S.; Li, D.; Li, M.; Su, Z.; Lai, X.; Zhou, C.; Chen, S.; Li, S.; Yang, X.; et al. Ethanol extract of Brucea javanica seed inhibit triple-negative breast cancer by restraining autophagy via PI3K/Akt/mTOR pathway. Front. Pharmacol. 2020, 11, 606. [Google Scholar] [CrossRef]
  119. Wang, M.; Shi, G.; Bian, C.; Nisar, M.F.; Guo, Y.; Wu, Y.; Li, W.; Huang, X.; Jiang, X.; Bartsch, J.W.; et al. UVA irradiation enhances Brusatol-mediated inhibition of melanoma growth by downregulation of the Nrf2-mediated antioxidant response. Oxidative Med. Cell. Longev. 2018, 2018, 9742154. [Google Scholar] [CrossRef] [PubMed]
  120. Bovilla, V.R.; Kuruburu, M.G.; Bettada, V.G.; Krishnamurthy, J.; Sukocheva, O.A.; Thimmulappa, R.K.; Shivananju, N.S.; Balakrishna, J.P.; Madhunapantula, S.V. Targeted inhibition of anti-inflammatory regulator Nrf2 results in breast cancer retardation in vitro and in vivo. Biomedicines 2021, 9, 1119. [Google Scholar] [CrossRef] [PubMed]
  121. Liu, P.; Zhang, B.; Li, Y.; Yuan, Q. Potential mechanisms of cancer prevention and treatment by sulforaphane, a natural small molecule compound of plant-derived. Mol. Med. 2024, 30, 94. [Google Scholar] [CrossRef] [PubMed]
  122. Zanotelli, M.R.; Zhang, J.; Reinhart-King, C.A. Mechanoresponsive metabolism in cancer cell migration and metastasis. Cell Metab. 2021, 33, 1307–1321. [Google Scholar] [CrossRef]
  123. Abdelwahed, K.S. Pseurotin a as a Novel PCSK9 Axis Lead Modulator for the Control of Breast and Prostate Malignancy Recurrences. Ph.D. Thesis, University of Louisiana at Monroe, Monroe, LA, USA, 2021. [Google Scholar]
  124. Marcucci, F.; Stassi, G.; De Maria, R. Epithelial-mesenchymal transition: A new target in anticancer drug discovery. Nat. Rev. Drug Discov. 2016, 15, 311–325. [Google Scholar] [CrossRef]
  125. Lee, J.H.; Mohan, C.D.; Deivasigamani, A.; Jung, Y.Y.; Rangappa, S.; Basappa, S.; Chinnathambi, A.; Alahmadi, T.A.; Alharbi, S.A.; Garg, M.; et al. Brusatol suppresses STAT3-driven metastasis by downregulating epithelial-mesenchymal transition in hepatocellular carcinoma. J. Adv. Res. 2020, 26, 83–94. [Google Scholar] [CrossRef]
  126. He, T.; Zhou, F.; Su, A.; Zhang, Y.; Xing, Z.; Mi, L.; Li, Z.; Wu, W. Brusatol: A potential sensitizing agent for cancer therapy from Brucea javanica. Biomed. Pharmacother. 2023, 158, 114134. [Google Scholar] [CrossRef]
  127. Déry, M.A.; Michaud, M.D.; Richard, D.E. Hypoxia-inducible factor 1: Regulation by hypoxic and non-hypoxic activators. Int. J. Biochem. Cell Biol. 2005, 37, 535–540. [Google Scholar] [CrossRef]
  128. Huang, Y.; Yang, Y.; Xu, Y.; Ma, Q.; Guo, F.; Zhao, Y.; Tao, Y.; Li, M.; Guo, J. Nrf2/HO-1 axis regulates the angiogenesis of gastric cancer via targeting VEGF. Cancer Manag. Res. 2021, 13, 3155–3169. [Google Scholar] [CrossRef] [PubMed]
  129. Bian, Y.; Li, Q.; Li, Q.; Pan, R. Silencing of BRF2 inhibits the growth and metastasis of lung cancer cells. Mol. Med. Rep. 2020, 22, 1767–1774. [Google Scholar] [CrossRef]
  130. Tsai, Y.F.; Chou, H.C.; Liou, M.H.; Liao, E.C.; Cheng, C.T.; Chang, S.J.; Chan, H.L. Role of IGFBP-2 in oral cancer metastasis. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166143. [Google Scholar] [CrossRef] [PubMed]
  131. Zhang, K.; Zhu, Z.; Jia, R.; Wang, N.A.; Shi, M.; Wang, Y.; Xiang, S.; Zhang, Q.; Xu, L. CD151-enriched migrasomes mediate hepatocellular carcinoma invasion by conditioning cancer cells and promoting angiogenesis. J. Exp. Clin. Cancer Res. 2024, 43, 160. [Google Scholar] [CrossRef]
  132. Guo, C.; Wei, L.I.; Xiao-Hui, L.I. The inhibitory effects of brusatol on the migration of human non-small cell lung carcinoma A549 cells and its molecular mechanism. Chin. J. Gerontol. 2016, 36, 3897–3899. [Google Scholar]
  133. So, T.H.; Chan, S.K.; Lee, V.H.; Chen, B.Z.; Kong, F.M.; Lao, L.X. Chinese Medicine in cancer treatment—How is it practised in the east and the West? Clin. Oncol. 2019, 31, 578–588. [Google Scholar] [CrossRef]
  134. Xiang, Y.; Guo, Z.; Zhu, P.; Chen, J.; Huang, Y. Traditional Chinese Medicine as a cancer treatment: Modern perspectives of ancient but advanced science. Cancer Med. 2019, 8, 1958–1975. [Google Scholar] [CrossRef] [PubMed]
  135. Dan, X.; Shou-Xiang, Y.; Peng, J.; Pharmacy, D.O. Literature analysis on adverse drug reactions induced by Brucea Jananica Oil Injection. Drug Eval. Res. 2017, 40, 266–269. [Google Scholar]
  136. Li, K.W.; Liang, Y.Y.; Wang, Q.; Li, Y.; Zhou, S.J.; Wei, H.C.; Zhou, C.Z.; Wan, X.H. Brucea javanica: A review on anticancer of its pharmacological properties and clinical researches. Phytomedicine 2021, 86, 153560. [Google Scholar] [CrossRef]
  137. Rong, S.U.N.; Qian, Y. Research development on toxicity of fructus Bruceae based on efficacy and material basis. Chin. J. Pharmacovigil. 2010, 7, 159. [Google Scholar]
  138. Hang, Y.; Feng, P.P.; Jiang, H.H.; Liu, K.; Zhang, D.; Li, S.Y. High-throughput and rapid characteristics analysis of Brucea javanica oil and its oral drugs by ultra high resolution mass spectrometry. J. Instrum. Anal. 2023, 42, 531–540. [Google Scholar] [CrossRef]
  139. Zhang, H.; Yang, J.Y.; Zhou, F.; Wang, L.H.; Zhang, W.; Sha, S.; Wu, C.F. Seed oil of Brucea javanica induces apoptotic death of acute myeloid leukemia cells via coth the death receptors and the mitochondrial-related pathways. Evid. Based Complement. Altern. Med. 2011, 2011, 965016. [Google Scholar] [CrossRef]
  140. Shi, W.R.; Liu, Y.; Wang, X.T.; Huang, Q.Y.; Cai, X.R.; Wu, S.R. Antitumor efficacy and mechanism in hepatoma H22-bearing mice of Brucea javanica oil. Evid. Based Complement. Altern. Med. 2015, 2015, 217494. [Google Scholar] [CrossRef] [PubMed]
  141. Yan, Z.; Zhang, B.; Huang, Y.; Qiu, H.; Chen, P.; Guo, G.F. Involvement of autophagy inhibition in Brucea javanica oil emulsion-induced colon cancer cell death. Oncol. Lett. 2015, 9, 1425–1431. [Google Scholar] [CrossRef]
  142. Wang, D.; Qu, X.; Zhuang, X.; Geng, G.; Hou, J.; Xu, N.; Li, W.; Hu, T.; Chen, Y.S. Seed oil of Brucea javanica induces cell cycle arrest and apoptosis via reactive oxygen species-mediated mitochondrial dysfunction in human lung cancer cells. Nutr. Cancer 2016, 68, 1394–1403. [Google Scholar] [CrossRef]
  143. Nan, Z.; Feng-Juan, H.; Gui-Yuan, W.; Yu-Hua, L.I. Effect of Brucea javanica fruit oil emulsion combination with cisplatin on the growth of human ovarian cancer SKOV3 cells nude mouse orthotopic transplantation tumor. Lett. Biotechnol. 2015, 26, 519–523. [Google Scholar]
  144. Zhao, N.; Li, Y.H.; Wu, X.K.; Wang, G.Y.; Cai, D.Y.; Han, F.J. Effect of Brucea javanica fruit oil emulsion combined cisplatin on the growth inhibition of transplanted tumor in human ovarian cancer SKOV3 nude mice: An experimental study. Zhongguo Zhong Xi Yi Jie He Za Zhi 2015, 35, 57–62. [Google Scholar]
  145. Zhonghui, S. Experiment study of oleum fructus bruceae vein emulsion on ovarian cancer cell strain. Mod. J. Integr. Tradit. Chin. West. Med. 2009, 18, 1591–1592. [Google Scholar]
  146. Wang, Y.; Chen, B.; Xiao, M.; Wang, X.; Peng, Y. Brucea javanica oil emulsion promotes autophagy in ovarian cancer cells through the miR-8485/LAMTOR3/mTOR/ATG13 signaling axis. Front. Pharmacol. 2022, 13, 935155. [Google Scholar] [CrossRef]
  147. Pan, P.; Yang, B.X.; Ge, X.L. Brucea javanica seed oil enhances the radiosensitivity of esophageal cancer by inhibiting hypoxia-inducible factor 1α, in vitro and in vivo. Oncol. Lett. 2018, 15, 3870–3875. [Google Scholar] [CrossRef] [PubMed]
  148. Qiu, Z.H.; Zhang, W.W.; Zhang, H.H.; Jiao, G.H. Brucea javanica oil emulsion improves the effect of radiotherapy on esophageal cancer cells by inhibiting cyclin D1-CDK4/6 axis. World J. Gastroenterol. 2019, 25, 2463–2472. [Google Scholar] [CrossRef] [PubMed]
  149. Ye, L.; Zhao, J.F.; Wang, Y.M.; Chen, W.H.; Qian, S.; Zhou, Z.G.; Xu, M. Brucea javanica oil emulsion suppresses tumor growth in human cervical cancer cells through inhibition of the E6 oncogene and induction of apoptosis. Transl. Cancer Res. 2020, 9, 918–929. [Google Scholar] [CrossRef]
  150. Wang, T.; Dou, Y.; Lin, G.; Li, Q.; Nie, J.; Chen, B.; Xie, J.; Su, Z.; Zeng, H.; Chen, J.; et al. The anti-hepatocellular carcinoma effect of Brucea javanica oil in ascitic tumor-bearing mice: The detection of brusatol and its role. Biomed. Pharmacother. 2021, 134, 111122. [Google Scholar] [CrossRef]
  151. Su, J.; Chen, X.; Xiao, Y.; Li, D.; Li, M.; Li, H.; Huang, J.; Lai, Z.; Su, Z.; Xie, Y.; et al. Bruceae fructus oil inhibits triple-negative breast cancer by restraining autophagy: Dependence on the gut microbiota-mediated amino acid regulation. Front. Pharmacol. 2021, 12, 727082. [Google Scholar] [CrossRef] [PubMed]
  152. Lou, G.G.; Yao, H.P.; Xie, L.P. Brucea javanica oil induces apoptosis in T24 bladder cancer cells via upregulation of caspase-3, caspase-9, and inhibition of NF-kappaB and COX-2 expressions. Am. J. Chin. Med. 2010, 38, 613–624. [Google Scholar] [CrossRef]
  153. Zhang, H.; Yin, S.L.; Wang, L.H.; Jia, L.N.; Su, G.Y.; Liu, X.Q.; Zhou, F.; Breslin, P.; Meng, R.; Li, Q.Y.; et al. Seed oil of Brucea javanica induces apoptosis through the PI3K/Akt signaling pathway in acute lymphocytic leukemia Jurkat cells. Chin. J. Nat. Med. 2021, 19, 608–620. [Google Scholar] [CrossRef]
  154. Jin, W.; Han, H.; Zhou, S.; Wang, Y.; Dong, T.; Zhao, C. Therapeutic efficacy of Brucea javanica oil emulsion (BJOE) combined with transcatheter hepatic arterial chemoembolization (TACE) in patients with primary liver cancer. Int. J. Clin. Exp. Med. 2015, 8, 18954–18962. [Google Scholar] [PubMed]
  155. Chen, J.; Chen, S.; Yang, X.; Wang, S.; Wu, W. Efficacy and safety of Brucea javanica oil emulsion injection as adjuvant therapy for cancer: An overview of systematic reviews and meta-analyses. Phytomedicine 2022, 102, 154141. [Google Scholar] [CrossRef] [PubMed]
  156. Liu, S.; Zhang, K.; Hu, X. Comparative efficacy and safety of Chinese medicine injections combined with capecitabine and oxaliplatin chemotherapies in treatment of colorectal cancer: A bayesian network meta-analysis. Front. Pharmacol. 2022, 13, 1004259. [Google Scholar] [CrossRef]
  157. Wu, J.R.; Liu, S.Y.; Zhu, J.L.; Zhang, D.; Wang, K.H. Efficacy of Brucea javanica oil emulsion injection combined with the chemotherapy for treating gastric cancer: A systematic review and meta-analysis. Evid. Based Complement. Altern. Med. 2018, 2018, 6350782. [Google Scholar] [CrossRef]
  158. Wu, Z.H.; Zhang, H.F.; Li, J.Y.; Diao, Y.R.; Huang, M.J.; Gao, D.Y.; Liang, C.H.; Luo, Z.Q. Effectiveness and safety of Brucea javanica oil assisted TACE versus TACE in the treatment of liver cancer: A systematic review and meta-analysis of randomized controlled trials. Front. Pharmacol. 2024, 15, 1337179. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Chemical structures of anticancer active compounds in BJ.
Figure 1. Chemical structures of anticancer active compounds in BJ.
Nutrients 17 03285 g001
Figure 2. BJ shows significant antitumor effect against various cancers.
Figure 2. BJ shows significant antitumor effect against various cancers.
Nutrients 17 03285 g002
Figure 3. Molecular mechanism of BJ-induced cancer cell apoptosis.
Figure 3. Molecular mechanism of BJ-induced cancer cell apoptosis.
Nutrients 17 03285 g003
Figure 4. Molecular mechanism of BJ-induced cell cycle arrest.
Figure 4. Molecular mechanism of BJ-induced cell cycle arrest.
Nutrients 17 03285 g004
Figure 5. Clinical and experimental study of BJ-related preparations.
Figure 5. Clinical and experimental study of BJ-related preparations.
Nutrients 17 03285 g005
Table 1. Cytotoxic activity (IC50 values) of BJ extracts and major bioactive compounds against various cancer cell lines.
Table 1. Cytotoxic activity (IC50 values) of BJ extracts and major bioactive compounds against various cancer cell lines.
Cancer Cell LineCompound/ExtractIC50 ReferenceCancer Cell LineCompound/ExtractIC50 Reference
HL-60Brusatol0.06 μmol/L[49]MCF-7Brusatol0.08 μmol/L[49]
Bruceine B0.27 μmol/LBruceine B0.54 μmol/L
Bruceine D1.14 μmol/LBruceine D6.75 μmol/L
Bruceine E4.48 μmol/LBruceine E17.77 μmol/L
Yadanziolide A26.32 μmol/LYadanziolide A14.61 μmol/L
SMMC-7721Brusatol<0.064 μmol/L[49]BJ dichloromethane extract55.1 μg/mL[58]
Bruceine B0.15 μmol/LBJ methanol extract80.5 μg/mL
Bruceine D0.88 μmol/LBJ ethanol extract15.12 μg/mL[55]
Bruceine E4.27 μmol/LBJ ethyl acetatel extract3.28 μg/mL
Yadanziolide A12.35 μmol/LBJ petroleum ether extract15.15 μg/mL
A-549Bruceine B0.24 μmol/L[49]BJ n-butyl alcohol extract30.92 μg/mL
Bruceine D3.30 μmol/LSW480Brusatol0.10 μmol/L[49]
Bruceine E7.62 μmol/LBruceine B0.30 μmol/L
Yadanziolide A17.05 μmol/LBruceine D7.78 μmol/L
BJ dichloromethane extract50.0 μg/mL[58]Bruceine E28.48 μmol/L
BJ methanol extract75.2 μg/mLPANC-1Brusatol0.36 mmol/L[21]
BJ water extract50 μg/mL[56]SW1990Brusatol0.10 mmol/L[21]
BJ ethanol extract8.79 μg/mL[55]Hs68 Bruceine D>30 μmol/L[63]
BJ ethyl acetatel extract0.02 μg/mLHCT-8Bruceine B2 μmol/L[68]
BJ petroleum ether extract9.14 μg/mLBruceine D2 μmol/L
BJ n-butyl alcohol extract17.47 μg/mLBruceine E6.70 μmol/L
Hep3BBrusatol0.69 μmol/L[76]Bruceine H1.30 μmol/L
BJ water extract50 μg/mL[56]HCT116Brusatol15 nmol/L[69]
BJ water extract4 mg/mL[77]BJ ethanol extract8.90 μg/mL[72]
Huh7Brusatol0.34 μmol/L[76]CT26Brusatol373 nmol/L[70]
LM3Brusatol12.49 μmol/L[76]HT29 BJ ethanol extract48 μg/mL[20]
Bel-7404Brusatol18.04 nmol/L[76]BGC-823javanicolide H0.52 μmol/L[68]
HepG2Bruceine B0.81 μmol/L[68]SKOV3Bruceine B0.12 μmol/L[68]
Bruceine D1.2 μmol/LBruceine D0.76 μmol/L
Bruceine E2.9 μmol/LBruceine E2.2 μmol/L
Bruceine H2.8 μmol/LBruceine H0.33 μmol/L
javanicolide H>10 μmol/Ljavanicolide H 0.23 μmol/L
javanicolide E>10 μmol/Ljavanicolide E1.49 μmol/L
Bel-7402Yadanziolide B4.24 μmol/L[29]MDA-MB231BJ water extract50 μg/mL[56]
Bel-7404Bruceantinol10 μmol/L[35]Brusatol0.081 μmol/L[37]
P-388javanicoside B5.6 μg/mL[96]Bruceantinol0.088 μmol/L
javanicolides C>100 μg/mLBruceine A0.228 μmol/L
javanicolides D18 μg/mLBruceantarin0.238 μmol/L
javanicosides C18 μg/mLHeLaquassilactones A78.95 μmol/L[86]
javanicosides D89 μg/mLquassilactones B92.57 μmol/L
javanicosides E16 μg/mLKBBJ extract24.37 μg/mL[101]
javanicosides F50 μg/mLORL-48BJ extract6.67 μg/mL[101]
H1650Brusatol24.27 ng/mL[105]U251Bruceanol20 nmol/L[102]
PC9Brusatol18.40 ng/mL[105]HCC827Brusatol73.3 ng/mL[105]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Xu, W.; Yang, H.; Zhou, Y.; Guo, R.; Liu, J.; Wei, F.; Lin, Y. Potential of Traditional Chinese Medicine Brucea javanica in Cancer Treatment: A Review of Chemical Constituents, Pharmacology, and Clinical Applications. Nutrients 2025, 17, 3285. https://doi.org/10.3390/nu17203285

AMA Style

Xu W, Yang H, Zhou Y, Guo R, Liu J, Wei F, Lin Y. Potential of Traditional Chinese Medicine Brucea javanica in Cancer Treatment: A Review of Chemical Constituents, Pharmacology, and Clinical Applications. Nutrients. 2025; 17(20):3285. https://doi.org/10.3390/nu17203285

Chicago/Turabian Style

Xu, Weiyin, Hongmei Yang, Yanan Zhou, Rixin Guo, Jing Liu, Feng Wei, and Yongqiang Lin. 2025. "Potential of Traditional Chinese Medicine Brucea javanica in Cancer Treatment: A Review of Chemical Constituents, Pharmacology, and Clinical Applications" Nutrients 17, no. 20: 3285. https://doi.org/10.3390/nu17203285

APA Style

Xu, W., Yang, H., Zhou, Y., Guo, R., Liu, J., Wei, F., & Lin, Y. (2025). Potential of Traditional Chinese Medicine Brucea javanica in Cancer Treatment: A Review of Chemical Constituents, Pharmacology, and Clinical Applications. Nutrients, 17(20), 3285. https://doi.org/10.3390/nu17203285

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop