Skip to main content

Insight into norcantharidin, a small-molecule synthetic compound with potential multi-target anticancer activities

Abstract

Norcantharidin (NCTD) is a demethylated derivative of cantharidin, which is an anticancer active ingredient of traditional Chinese medicine, and is currently used clinically as a routine anti-cancer drug in China. Clarifying the anticancer effect and molecular mechanism of NCTD is critical for its clinical application. Here, we summarized the physiological, chemical, pharmacokinetic characteristics and clinical applications of NCTD. Besides, we mainly focus on its potential multi-target anticancer activities and underlying mechanisms, and discuss the problems existing in clinical application and scientific research of NCTD, so as to provide a potential anticancer therapeutic agent for human malignant tumors.

Background

Since Tu Youyou was awarded the 2015 Nobel Prize in physiology or medicine for the discovery of artemisinin used for malaria treatment, traditional Chinese medicines (TCMs) and natural medicine are getting more attention. A growing body of evidences indicate that TCMs contain anticancer ingredient. Norcantharidin (NCTD), a demethylated derivative of cantharidin which is an active ingredient of TCM—Mylabris [1,2,3], is currently used clinically as an optional anticancer drug in China, because of its relatively synthesized facility, potential anticancer activity, and less side-effects such as myelosuppression, gastrointestinal and urinary tract toxicity [1,2,3,4,5]. Increasing evidences show that NCTD not only effectively inhibited the proliferation of many tumor cells in vitro and in vivo, including hepatoma HepG2 [6,7,8], SMMC-7721 [8, 9] and BEL-7402 [10, 11], gallbladder cancer GBC-SD cells [12, 13], colon cancer CT26 and HT29 cells [14, 15], breast cancer cells [16, 17], leukemia K562 [18] and HL-60 cells [4, 5, 19], melanoma A375 cells [20], and oral cancer KB cells [21], but also decreased tumor growth and prolonged survival in animal models in vivo [17, 22]. As an efficacious anticancer drug, it has been used to treat hepatic cancer, gastric cancer and leucopenia patients in China for many years. To deepen the understanding of the characteristics and clinical application of NCTD is of great significance for NCTD to work as an anticancer drug in clinic. Here, we review the physiological, chemical, pharmacokinetic characteristics and clinical uses, especially, potential multi-target anticancer activities such as inducing apoptosis, inhibiting proliferation, blocking invasion/metastasis, antiangiogenesis, anti-vasculogenic mimicry, anti-lymphangiogenesis and underlying mechanisms of NCTD, so as to provide a potential anticancer therapeutic agent for human malignant tumors.

Physiological, chemical and pharmacokinetic characteristics

Norcantharidin (NCTD, 7-oxabicyclo[2.2.1] heptane-2,3-dicarboxylic anhydride) is a demethylated analogue of cantharidin (CTD). The molecular formula is C8H8O4 and the molecular formula is 168.15 g/mol. NCTD can not only be extracted from TCM Mylabris (Spanish fly) [1,2,3,4] (Fig. 1), but also can be synthesized from furan and maleic anhydride via the Diels–Alder reaction [23] (Fig. 2). It is a colorless, odorless, slightly irritating crystalline powder, being slightly soluble in water and ethanol, and soluble in hot water and acetone. This small-molecule synthetic compound has low-cytotoxic features and few side effects such as less marrow suppression (myelosuppression), low toxicity of gastrointestinal and urinary tract, because of removing 1,2 methyl groups on the chemical structure of CTD [1,2,3,4,5].

Fig. 1
figure 1

The origin, evolvement and molecular formula of norcantharidin (NCTD). Mylabris, also known as Spanish fly, is a traditional Chinese medicine. Cantharidin (CTD), a 7-oxabicyclo [2.2.1] heptane-2, 3-dicarboxylic acid derivative, a natural toxin and the active ingredient with antitumor properties extracted from a traditional Chinese medicine Mylabris. NCTD (7-oxabicyclo [2.2.1] heptane-2, 3-dicarboxylic anhydride), with a molecular formula of C8H8O4 and formula weight of 168.15 g/mol, is the demethylated analog and the low-cytotoxic derivative of CTD with antitumor properties

Fig. 2
figure 2

Synthesis of NCTD by furan and maleic anhydride through Diels–Alder reaction. NCTD can be synthesized by furan and maleic anhydride through Diels–Alder reaction under appropriate conditions

In pharmacokinetics, radionuclide and whole-body autoradiography showed that NCTD was rapidly absorbed by intragastric administration in mice with 3H-norcantharidin, reached a higher concentration within 15 min and 2 h after dosing in the kidney, liver, tumor, stomach, intestines, heart and lung. NCTD was highly distributed in the bile duct, liver, kidney, heart and lung by intravenous administration, reached the peak concentration in liver and cancer tissues within 15 min after dosing. After 6 h, the concentration decreased significantly by being excreted from the urethra. Most of drugs were excreted from the kidneys within 24 h, and were rarely accumulated in the various organs of the body [24]. Thus, NCTD is less likely to cause drug accumulation poisoning.

Clinical uses

As an efficacious anticancer drug, NCTD has been used to treat cancer patients clinically in China for many years. Two thousand years ago, Mylabris (Spanish fly), a traditional Chinese medicine, was used to treat “abdominal mass” in China [1,2,3,4]. Later, an active ingredient of Mylabris—CTD was artificially extracted and be used to treat many human tumors as a natural toxin [1,2,3,4]. Afterwards, in order to alleviate side effects of CTD such as gastrointestinal and urinary tract toxicity, NCTD was extracted from CTD, or was synthesized from furan and maleic anhydride [1,2,3,4, 23]. Now, NCTD is clinically used as a routine anticancer drug in China.

Clinical indications of NCTD include: (1) It is used to treat patients with digestive tumors, such as hepatocellular cancer, esophageal cancer, gastric cancer, and colorectal cancer and it shows better curative effect; (2) It is used to treat other cancer patients, such as lung, breast and ovarian cancers and has certain curative effect; (3) Also, it is used as premedication or in combination with other antineoplastic drugs. In addition, NCTD can also be used for hepatitis, liver cirrhosis and leukopenia.

Usage of NCTD includes oral, intravenous administration and local injection. For oral, 5–15 mg (most dose can be added to 30 mg) NCTD is used for one time, 3 times a day, 1 months for 1 courses, generally 3 courses. For intravenous infusion or intravenous drip, 10–20 mg a day, added to the 5% glucose injection 250–500 ml, in a slow drop by intravenous drip; or added to the 5% glucose injection 10–20 ml, by slow intravenous injection; 1 month for 1 treatment course. And for local injection, 20–40 mg/times, once a week, 2–4 times for 1 courses.

Growing clinical evidences demonstrated that NCTD was an efficacious anticancer drug for cancer patients. Table 1 illustrates the clinical uses of NCTD and the related results [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48]. No matter NCTD is used alone via oral, intravenous administration, intro-tumor injection, or in combination with chemotherapy, radiotherapy and other therapies such as interventional therapy (IVT), transcatheter arterial chemoembolization (TACE) and TCMs can reduce tumors, improve symptoms and life quality, alleviate side effects, and prolong survival time in most patients with mid-advanced stage tumors such as hepatocellular cancer, esophageal cancer, gastric cancer, lung cancer, ovarian cancer, non-Hodgkin lymphoma and so on [25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48]. Thus, NCTD is believed as a useful adjunct anticancer drug in clinical treatment of mid-advanced stage tumors and in the prevention of post-operational recurrent tumors.

Table 1 Clinical uses of NCTD in treatment of cancer patients and the related results and outcomes

Multi-target anticancer activities and underlying mechanisms

The multi-target anticancer activities and underlying mechanisms of NCTD in treatment of different cancer models and cell lines have been reported. Here, we systematically review the potential anticancer activities and underlying molecular mechanisms of NCTD in vitro and in vivo.

Inhibiting proliferation and inducing apoptosis

In recent years, a large number of researches have been carried out to study the effects of NCTD on inhibiting proliferation and inducing apoptosis in different cancer models (Table 2). NCTD has a cytotoxic effect on a variety of tumor cells. Significant anti-proliferative and apoptotic effects are observed in NCTD-treated tumor cells [7, 49, 50]. At the same time, relevant studies have confirmed that NCTD has no myelosuppression and can induce hematopoiesis via bone marrow stimulation while exerting its anticancer activity [4, 5]. NCTD has no effect on the viability of normal peripheral blood mononuclear cells (MNC) [51, 52]. These are incomparable advantages over many traditional anticancer drugs. In addition, NCTD has a synergistic effect with a variety of anticancer drugs, such as cisplatin and gefitinib [53, 54].

Table 2 Relevant researches of NCTD on inhibiting proliferation and inducing apoptosis

The anti-proliferation and pro-apoptotic effects of NCTD depend on the complex interactions between different molecules (Fig. 3). On the one hand, the inhibitory effect of NCTD on proliferation is mainly achieved through cell cycle arrest and inhibition of DNA synthesis by inhibiting the expression of cyclins, cyclin-dependent kinases (CDKs) and increasing the expression of cyclin-dependent kinase inhibitors (CDKIs, such as p21Cip/Waf1, p27kip1); On the other hand, NCTD can also induce apoptosis by increasing the expression of pro-apoptotic protein such as P53, Bax, Caspases, and reducing the expression of anti-apoptotic proteins such as Bcl-2 (B-cell lymphoma-2) and survivin. These mechanisms have been confirmed in a variety of tumor cell lines such as leukemia K562 and HL-60 [18, 55], hepatoma HepG2, SMMC-7721 and BEL-7402 [56,57,58], colorectal cancer CT26 and HCT-15 cells [59, 60], etc. It is generally believed that serine/threonine protein phosphatases, such as protein phosphatase type 1 (PP1), protein phosphatase-2A (PP2A) and protein phosphatase-2B (PP2B), play important roles in intracellular signal transduction, whose inhibition is an excellent target for the development of novel anti-cancer agents [5, 61, 62]. Some studies have confirmed that NCTD, as a PP2A inhibitor, can inhibit cancer cell proliferation and induce apoptosis by inhibiting the activity of PP2A [5, 62, 63]. In addition, DNA replication-initiation protein Cdc6 (cell division cycle protein 6) is an effective target to disturb DNA replication [64]. Other studies have found that NCTD can inhibit cell proliferation by inducing Cdc6 degradation [65, 66]. In gallbladder cancer, it was reported that NCTD inhibited the expression of GBC-SD cell proliferation-related gene proteins PCNA (proliferating cell nuclear antigen) and Ki-67, this may be one of the mechanisms by which NCTD inhibit the proliferation and growth of tumor cells [12, 67].

Fig. 3
figure 3

The “multi-points priming” mechanisms of NCTD on inhibiting proliferation and inducing apoptosis. NCTD: norcantharidin; PI3K: phosphoinositide 3 kinase; NF-κB: nuclear factor-kappa B; MAPK: mitogen-activated protein kinase; JNK: Jun N-terminal kinase; PP1: protein phosphatase type 1; PP2A: protein phosphatase 2A; PP2B: protein phosphatase 2B; Cdc6: cell division cycle protein 6; CD1: cyclin D1; CDKs: cyclin-dependent kinases; CDKIs: cyclin-dependent kinase inhibitors; Bcl-2: B-cell lymphoma-2; (−): Inhibition; (+): Promotion or inducing

NCTD inhibited proliferation and induced apoptosis in cancer cells is dose- and time-dependent [51, 55], and is regulated by both extrinsic and intrinsic signaling pathways [34]. MAPK (mitogen-activated protein kinase) can be divided into four subfamilies: ERK (extracellular regulated protein kinases), p38, JNK (Jun N-terminal kinase) and ERK5. MAPK-related signaling pathways are widely involved in NCTD-induced apoptosis [68]. For instance, NCTD-induced apoptosis in leukemia HL-60 cells is regulated by activating JNK signaling [19], and apoptosis in hepatocellular cancer HepG2 cells induced by NCTD is dependent on ERK and JNK activity [6]. The Wnt/β-catenin signaling pathway is considered to be another target for antitumor drugs [69]. Some studies have shown that NCTD can reduce the proliferation of leukemia Jurkat cells by inhibiting Wnt/β-catenin signaling [70]. Due to the ability to cross the blood–brain barrier, NCTD can also significantly inhibit the growth of medulloblastoma through Wnt/β-catenin signaling pathway [71]. In addition, NCTD can inhibit the expression of the proliferation-related protein cyclin D1, downregulate the expression of anti-apoptotic protein, and upregulate the expression of pro-apoptotic protein by blocking PI3K (phosphoinositide 3 kinase)/Akt/NF-κB (nuclear factor-kappa B) pathway [72, 73]. So, the PI3K/Akt/NF-κB pathway has been shown to be another signal pathway for the regulation of NCTD-mediated anti-proliferation and pro-apoptosis.

Inhibiting tumor invasion/metastasis

Two major protein families are involved in NCTD against tumor invasion and metastasis, including matrix metalloproteinases (MMPs) and adhesion molecules [74]. The MMP family, particularly MMP-2 and MMP-9, has gelatinase activity and is capable of proteolytic cleavage of plasminogen in extracellular matrix [75]. Cell adhesion molecules such as α-catenin and b-catenin have the function of adhering tumor cells to other cellular and matrix components [76], both of them play an important role in local invasion and distant metastasis.

It has been confirmed that NCTD has anti-invasion and anti-metastasis effects in many kinds of tumor cells (Table 3). Some experiments indicated that NCTD reduces the activity of MMP-2 and MMP-9 by upregulating the transcription factor STAT1 (signal transducers and activators of transcription 1) and inhibiting the transactivation of Sp1 (specificity protein 1), thereby inhibiting the invasion and metastasis of tumor cells [77, 78]. Another study showed that NCTD has the ability to reduce the expression of α-catenin and β-catenin in colorectal cancer CT26 cells, suggesting that the anti-invasive and anti-metastatic activity of NCTD may be related to the regulation of these adhesion molecules [75]. Furthermore, epithelial–mesenchymal transition (EMT) is widely involved in the invasion and metastasis of malignant epithelial tumors [79]. NCTD inhibits the EMT process in non-small cell lung cancer, colorectal cancer and hepatocellular cancer cells via the αvβ6-ERK-Ets1 (E-Twenty-Six-1) signaling pathway blocking and NCTD-mediated Yes-associated protein (YAP) inhibition [78, 80, 81]. These regulatory mechanism of NCTD against tumor invasion and metastasis is detailed in Fig. 4.

Table 3 Relevant researches of NCTD against invasion and metastasis for multiple cell lines in different cancer models
Fig. 4
figure 4

Underlying regulatory targets of NCTD against invasion and metastasis. NCTD: norcantharidin; YAP: Yes-associated protein; ERK: extracellular regulated protein kinases; Ets1: E-Twenty-Six-1; Sp1: specificity protein 1; STAT1: signal transducers and activators of transcription 1; MMPs: matrix metalloproteinases; EMT: epithelial–mesenchymal transition. (−): Inhibition; (+): Promotion or inducing

Anti-angiogenesis and anti-vasculogenic mimicry

Angiogenesis and effective blood supply are basic conditions for tumor growth and metastasis [82]. Multiple angiogenic growth factors and cytokines play important roles in regulating tumor angiogenesis, such as vascular endothelial growth factor (VEGF) and its corresponding receptor, thrombospondin (TSP), angiogenin (Ang), and tissue metalloproteinase inhibitor (TIMP) family. In gallbladder and colorectal cancer, it has been confirmed that NCTD can inhibit angiogenesis, induce apoptosis of vascular endothelial cells, downregulate the expression of angiogenic factors such as VEGF, VEGFR-2 (vascular endothelial growth factor receptor-2), Ang-2, and upregulate the expression of anti-angiogenic factors such as TSP and TIMP-2 [83,84,85,86]. So, NCTD may be a potential anti-angiogenic drug for cancer treatment.

Tumor vasculogenic mimicry (VM) refers to a novel tumor blood supply pattern that occurs in certain highly aggressive malignancies and is associated with poor clinical outcomes and poor prognosis [87]. TIMP-2 has anti-VM activity in some highly aggressive malignancies [88]. Furthermore, the PI3-K (phosphatidylinositol 3-kinase)/MMPs (matrix metalloproteinases)/Ln-5γ2 (laminin 5γ2) and EphA2 (ephrin type a receptor 2)/FAK (focal adhesion kinase)/Paxillin signaling pathways are two critical pathways for the control of VM formation [89], while MMP-2 and MT1-MMP (membrane type 1-matrix metalloproteinase) are key molecules and important mediators of these two pathways, regulating VM formation in invasive malignant cells [90]. NCTD is believed as a potential anti-VM active drug, its anti-VM mechanisms mainly involves two aspects: NCTD downregulates the expression of MMP-2 and MT1-MMP via inhibiting EphA2/FAK/Paxillin signaling pathway, thereby enhancing the anti-VM activity of TIMP-2; in turn, a decrease in MMP-2 and MT1-MMP activity inhibits PI3-K/MMPs/Ln-5γ2 signaling and exerts an anti-VM effect on malignant cells [13, 91,92,93].

Anti-lymphangiogenesis

Lymphatic metastasis is one of the important metastatic pathways of tumors, and tumor lymphatic tube formation (lymphangiogenesis) plays an important role in tumor growth, metastasis and prognosis [94]. Lymphatic endothelial growth factors, including two members of the VEGF family, VEGF-C and VEGF-D, as well as their cognate receptor VEGFR-3, are the main regulators of tumor lymphangiogenesis and is of great significance in tumor lymph node metastasis [95,96,97]. In recent years, some researchers have reported that NCTD is an effective lymphangiogenesis inhibitor. The basic mechanism of NCTD anti-lymphangiogenesis refers to directly or indirectly downregulate the expression of VEGF-C, VEGF-D and VEGFR-3 at protein and mRNA levels, which has been proved in human lymphatic endothelial cells (HLECs) and human colonic adenocarcinomas (HCACs) [98,99,100]. In addition, NCTD in combine with sorafenib or mF4-31C1 enhanced the ability of anti-lymphangiogenesis in human colonic adenocarcinomas [100].

The relevant researches and mechanisms of NCTD inhibiting tumor vascularization (Angiogenesis, VM and lymphangiogenesis) are summarized in Table 4 and Fig. 5.

Table 4 Relevant studies of NCTD anti-angiogenesis, anti-VM, and anti-lymphangiogenesis
Fig. 5
figure 5

The “more targets” mechanisms of NCTD against tumor vascularization (angiogeneses, VM and lymphangiogenesis). NCTD: norcantharidin; TSP: thrombospondin; Ang-2: angiogenin-2; VEGF: vascular endothelial growth factor; VEGFR: vascular endothelial growth factor receptor; EphA2: ephrin type a receptor 2; FAK: focal adhesion kinase; PI3-K: phosphatidylinositol 3-kinase; MMPs: matrix metalloproteinases; Ln-5γ2: laminin 5γ2; TIMP: tissue metalloproteinase inhibitor. (−): Inhibition; (+): Promotion or inducing

Overcoming multi-drug resistance

Multi-drug resistance (MDR) refers to tumor cells develop resistance to anti-tumor drugs, as well as producing cross-resistance to other antineoplastics with different structures and mechanisms [101]. As one of the main problems in clinical tumor chemotherapy, MDR directly affects the efficacy of chemotherapy drugs and even lead to treatment failure [102].

In human breast cancer cells, NCTD may overcome MDR through inhibiting sonic hedgehog (Shh) signaling and its downstream MDR-1/P-gp expression [103], which has been shown to increase resistance to a variety of structurally unrelated antitumor drugs [104]. Bcl-2 family proteins Bcl-2 and Bcl-xL are resistant to multiple chemotherapeutic agents in a variety of cell lines [105,106,107], and it was reported that NCTD downregulated the expression of Bcl-2 and Bcl-xL in oral cancer cells [108]. In addition, Bcl-2 family inhibitors ABT-737 and ABT-263 are two promising anticancer agents with anticancer activity against a variety of cancer cells [109, 110]. NCTD significantly enhances ABT-263 and ABT-737-mediated anticancer activity, and overcomes the increased ABT-737 resistance caused by elevated Mcl-1 levels in cancer cells [111,112,113]. Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) are widely used in anti-tumor therapy for non-small cell lung cancer (NSCLC) [114]. HGF (hepatocyte growth factor) overexpression is a major factor contributing to acquired resistance caused by EGFR-TKI [115]. NCTD can overcome HGF-induced EGFR-TKI resistance in EGFR-mutant lung cancer cells by inhibition of the Met/PI3K/Akt pathway [116]. Therefore, NCTD may be a potential agent to reverse MDR (Table 5).

Table 5 Summary of related research on NCTD overcoming multidrug resistance

Promoting tumor cell demethylation

Tumorigenesis is a process of interaction between genetic and epigenetic mechanisms. DNA methylation is an important epigenetic regulator closely related to the occurrence and development of tumors [117]. Abnormal DNA methylation is involved in the pathogenesis of tumors. DNA hypomethylation promotes gene expression, while DNA hypermethylation inhibits gene expression [118, 119]. Hypermethylation of RASSF1A (a tumor suppressor gene) results in loss of function in human tumor cells [120]. It was reported that NCTD can inhibit RASSF1A methylation and inducing its re-expression in hepatocellular cancers [121]. Moreover, the Wnt/β-catenin signaling pathway is closely related to a variety of neoplastic diseases and is activated in tumor formation [122, 123]. Wnt inhibitory factor-1 (WIF-1), as a Wnt antagonist, has the function of inhibiting Wnt signal transduction. And due to hypermethylation of the promoter, WIF-1 silencing occurs in some tumor cells [124]. Studies have demonstrated that NCTD can activate WIF-1 to inhibit Wnt signaling pathway through promoter demethylation in NSCLC and glioma cells [125, 126] (Table 6).

Table 6 Studies of NCTD on promoting demethylation, modulating immune response and some other anticancer activities

Modulating immune responses

The immune system plays a very important role in the development of tumors. The inflammatory response is a common and serious complication due to the continued damage to the immune system by the cancer itself and anti-cancer drugs. NCTD positively regulates macrophage-mediated immune responses via the AKT/NF-κB signaling pathway, helping to clear invading pathogens [127]; NCTD also reduces tissue inflammation by suppressing PBMC (human peripheral blood mononuclear cells) proliferation and cytokine gene expression and production [128]. In addition, the increased production of IL-10 will block the effect of specific T lymphocytes on tumor cells [129], and NCTD inhibits the production of IL-10 in PBMC induced by PHA (phytohemagglutinin) [128] (Table 6).

Others

NCTD has also been reported to have some other anticancer activities, including inhibition of tumor glucose oxidative metabolism [130]; inhibition of NAT (N-acetyltransferase) activity [131]; regulation of macrophage polarization [175]; regulation of leukemia stem cell activity [4] (Table 6). Due to the lack of relevant researches, it is necessary to further verify the relevant mechanisms and applications in the clinic.

Discussion

In recent years, the anti-tumor effect of TCMs has aroused extensive attention. However, due to the complexity of components, difficulty in extraction and high toxicity, the clinical application of many anti-tumor TCMs is limited. NCTD, as a demethylation product of CTD, can be extracted from CTD or synthesized artificially at a low cost. In addition, its physical and chemical properties are clear, so it is convenient for basic and clinical research. These prerequisites are helpful for the promotion of NCTD in clinical practice.

On the basis of summarizing the relevant literature, we found that there are two main ways of clinical application of NCTD. First of all, NCTD can be used as an anti-tumor drug alone in the treatment of liver cancer, gastric cancer and other tumors, especially for advanced malignant tumors that have lost the opportunity of operation. Secondly, it is used as an adjuvant of other anti-tumor drugs, which is currently the most important way for NCTD applied in clinic. Some studies have shown that the combination of NCTD with other anticancer drugs, or as an adjuvant to chemotherapy or interventional therapy, can help to improve the efficacy, increase the tolerance of patients, reduce side effects, and improve the prognosis [28, 30, 33].

Adverse reactions and serious complications of NCTD are rare. Gastrointestinal symptoms such as nausea and vomiting may occur when the oral dose or injection is excessive. A study has shown that patients with advanced liver cancer who take NCTD more than 45 mg/day will have significant gastrointestinal response [25]. It has also been reported that when the dosage of NCTD reaches 600 mg, the patients may have slight gastrointestinal symptoms, but it will be relieved soon after the drug is stopped or the alkaline agent is taken [27]. A large number of clinical studies have proven that patients treated with NCTD have no obvious symptoms of urinary irritation, no adverse effects on liver and renal function, and no myelosuppression [27, 28, 32].

Among the three routes of administration, oral administration and intravenous administration are simple and safe. The disadvantage is that the drug is eliminated quickly in the body, resulting in poor anti-tumor effect. It is reported that the half-life of NCTD in blood is short, only about 0.26 h [17]. Local injection is mainly used for some solid tumors, especially for advanced liver cancer which can not be treated by surgery. Compared with the former two, this method has better curative effect. However, due to the invasive operation, there are some risks such as bleeding, cancer rupture and so on.

NCTD has the disadvantages of poor water solubility, short half-life and low tumor targeting efficiency, which limits its clinical application [132, 176]. Therefore, a variety of NCTD analogues have been developed to improve the clinical applicability and efficacy. These NCTD analogues can be divided into two categories: new NCTD reagents and drug delivery systems. For example, it has been reported a new type of NCTD conjugate recently, called CNC conjugates (NCTD-conjugated carboxymethyl chitosan). Compared with the same dose of free NCTD, CNC conjugates have higher therapeutic concentration and longer half-life. It can not only enhance the inhibitory effect on cancer cells, but also reduce side effects [177, 178]. In addition, some other NCTD derivatives and liposomes, such as NOC15 (N-farnesyloxy-norcantharimide) [179] and SG-NCTD-LIP (NCTD-loaded liposomes modified with stearyl glycyrrhetinate) [176], also can effectively improve the anticancer activity and reduce the toxicity of NCTD. However, although these studies have shown that NCTD analogues have a very broad application prospect, most of the existing NCTD analogues have no obvious selectivity for tumors and targets. And it should be noted that most of the relevant researches are in the stage of basic research at present, whether these NCTD analogues can be applied to clinical needs to be confirmed by a large number of clinical experiments.

Conclusions

Collectively, NCTD, as a demethylation derivative of traditional Chinese medicine, has been clinically used to treat cancer patients, and is gradually believed as a useful adjunct anticancer drug, especially for the patients with mid-advanced and postoperational recurrent cancers. The underlying molecular mechanisms of NCTD anticancer activities maybe “multi-factor”, “more targets” and “multi-points priming” mechanisms, include inhibiting proliferation, inducing apoptosis, inhibiting tumor invasion and metastasis, anti-neoangiogenesis (including anti-angiogenesis and anti-VM), anti-lymphangiogenesis, overcoming multiple drug resistance, promoting tumor cell demethylation, modulating immune responses and so on. Numerous clinical applications and drug experiments have also demonstrated that NCTD has effective and “multi-factor” anticancer activities, especially in apoptotic inducement in human cancer cells by “more targets” and “multi-points priming” mechanisms. But other mechanisms of NCTD’s anticancer effects such as anti-angiogenesis, anti-VM, anti-lymphangiogenesis as well as overcoming multiple drug resistance are seldom reported. It is necessary to improve the relevant research, which is of great significance for the development of NCTD as a potential chemotherapeutic agent.

Availability of data and materials

All available data and material can be accessed.

Abbreviations

TCM:

Traditional Chinese medicine

NCTD:

Norcantharidin

CTD:

Cantharidin

IVT:

Interventional therapy

TACE:

Transcatheter arterial chemoembolization

MNC:

Mononuclear cells

Bcl-2:

B-cell lymphoma-2

PP2A:

Protein phosphatase 2A

Cdc6:

Cell division cycle protein 6

PCNA:

Proliferating cell nuclear antigen

MAPK:

Mitogen-activated protein kinase

ERK:

Extracellular regulated protein kinases

JNK:

Jun N-terminal kinase

PI3K:

Phosphoinositide 3 kinase

NF-κB:

Nuclear factor-kappa B

MMPs:

Matrix metalloproteinases

STAT1:

Signal transducers and activators of transcription 1

Sp1:

Specificity protein 1

EMT:

Epithelial–mesenchymal transition

YAP:

Yes-associated protein

VM:

Vasculogenic mimicry

VEGF:

Vascular endothelial growth factor

TSP:

Thrombospondin

Ang:

Angiogenin

TIMP:

Tissue metalloproteinase inhibitor

VEGFR:

Vascular endothelial growth factor receptor

Ln-5γ2:

Laminin 5γ2

EphA2:

Ephrin type a receptor 2

FAK:

Focal adhesion kinase

MT1-MMP:

Membrane type 1-matrix metalloproteinase

HLECs:

Human lymphatic endothelial cells

HCACs:

Human colonic adenocarcinomas

MDR:

Multi-drug resistance

Shh:

Sonic hedgehog

EGFR-TKIs:

Epidermal growth factor receptor- tyrosine kinase inhibitors

NSCLC:

Non-small cell lung cancer

HGF:

Hepatocyte growth factor

WIF-1:

Wnt inhibitory factor-1

PBMC:

Peripheral blood mononuclear cells

PHA:

Phytohemagglutinin

NAT:

N-acetyltransferase

CNC conjugates:

NCTD-conjugated carboxymethyl chitosan

NOC15:

N-farnesyloxy-norcantharimide

SG-NCTD-LIP:

NCTD-loaded liposomes modified with stearyl glycyrrhetinate

References

  1. Wang GS. Medical uses of Mylabris in ancient China and recent studies. J Ethnopharmacol. 1989;26:147–62.

    Article  CAS  PubMed  Google Scholar 

  2. Jiang Z, Chi J, Han B, Liu W. Preparation and pharmacological evaluation of norcantharidin-conjugated carboxymethyl chitosan in mice bearing hepatocellular carcinoma. Carbohydr Polym. 2017;174:282–90.

    Article  CAS  PubMed  Google Scholar 

  3. Chang C, Zhu YQ, Mei JJ, Liu SQ, Luo J. Involvement of mitochondrial pathway in NCTD-induced cytotoxicity in human hepG2 cells. J Exp Clin Cancer Res. 2010;29(1):145.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  4. Dorn DC, Kou CA, Png KJ, Moore MA. The effect of cantharidins on leukemic stem cells. Int J Cancer. 2009;124(9):2186–99.

    Article  CAS  PubMed  Google Scholar 

  5. Liu XH, Blazsek I, Comisso M, Legras S, Marion S, Quittet P, et al. Effects of norcantharidin, a protein phosphatase type-2A inhibitor, on the growth of normal and malignant haemopoietic cells. Eur J Cancer. 1995; null: 953–63.

    Article  Google Scholar 

  6. Chen YN, Cheng CC, Chen JC, Tsauer W, Hsu SL. Norcantharidin-induced apoptosis is via the extracellular signal-regulated kinase and c-Jun-NH2-terminal kinase signaling pathways in human hepatoma HepG2 cells. Br J Pharmacol. 2003;140(3):461–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Chen YN, Chen JC, Yin SC, Wang GS, Tsauer W, Hsu SF, et al. Effector mechanisms of norcantharidin-induced mitotic arrest and apoptosis in human hepatoma cells. Int J Cancer. 2002;100(2):158–65.

    Article  CAS  PubMed  Google Scholar 

  8. Wang Q, Zhang L, Hu W, Hu ZH, Bei YY, Xu JY, et al. Norcantharidin- associated galactosylated chitosan nanoparticles for hepatocyte-targeted delivery. Nanomedicine. 2010;6(2):371–81.

    Article  CAS  PubMed  Google Scholar 

  9. Wan XY, Zhai XF, Jiang YP, Han T, Zhang QY, Xin HL. Antimetastatic effects of norcantharidin on hepatocellular carcinoma cells by up-regulating FAM46C expression. Am J Transl Res. 2017;9(1):155.

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Yang H, Guo W, Xu B, Li M, Cui J. Anticancer activity and mechanisms of norcantharidin-Nd3II on hepatoma. Anticancer Drugs. 2007;18(10):1133–7.

    Article  CAS  PubMed  Google Scholar 

  11. Li G, Zhang S, Lü JF, Wu Y, Li J, Zhang G, et al. Molecular mechanism of norcantharidin inducing apoptosis in liver cancer cells. Zhonghua Yi Xue Za Zhi. 2010;90(30):2145.

    CAS  PubMed  Google Scholar 

  12. Fan YZ, Fu JY, Zhao ZM, Chen CQ. Effect of norcantharidin on proliferation and invasion of human gallbladder carcinoma GBC-SD cells. World J Gastroenterol. 2005;11(16):2431–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Wang H, Sun W, Zhang WZ, Ge CY, Zhang JT, Liu ZY, et al. Inhibition of tumor vasculogenic mimicry and prolongation of host survival in highly aggressive gallbladder cancers by norcantharidin via blocking the ephrin type a receptor 2/focal adhesion kinase/paxillin signaling pathway. PLoS ONE. 2014;9(1):64.

    Google Scholar 

  14. Chen YJ, Tsai YM, Kuo CD, Ku KL, Shie HS, Liao HF. Norcantharidin is a small-molecule synthetic compound with anti-angiogenesis effect. Life Sci. 2009;85(17–18):642–51.

    Article  CAS  PubMed  Google Scholar 

  15. Yeh CB, Su CJ, Hwang JM, Chou MC. Therapeutic effects of cantharidin analogues without bridging ether oxygen on human hepatocellular carcinoma cells. Eur J Med Chem. 2010;45(9):3981–5.

    Article  CAS  PubMed  Google Scholar 

  16. He Q, Xue S, Tan Y, Zhang L, Shao Q, Xing L, et al. Dual inhibition of Akt and ERK signaling induces cell senescence in triple-negative breast cancer. Cancer Lett. 2019;448:94–104.

    Article  CAS  PubMed  Google Scholar 

  17. Li Y, Xiao Y, Lin HP, Reichel D, Bae Y, Lee EY, et al. In vivo β-catenin attenuation by the integrin α5-targeting nano-delivery strategy suppresses triple negative breast cancer stemness and metastasis. Biomaterials. 2019;188:160–72.

    Article  CAS  PubMed  Google Scholar 

  18. Yi S, Wass J, Vincent P, Iland H. Inhibitory effect of norcantharidin on K562 human myeloid leukemia cells in vitro. Leuk Res. 1991;15(10):883.

    Article  CAS  PubMed  Google Scholar 

  19. Wang SC, Chow JM, Chien MH, Lin CW, Chen HY, Hsiao PC, et al. Cantharidic acid induces apoptosis of human leukemic HL-60 cells via c-Jun N-terminal kinase-regulated caspase-8/-9/-3 activation pathway. Environ Toxicol. 2018;33:514–22.

    Article  CAS  PubMed  Google Scholar 

  20. Liu S, Yu H, Kumar SM, Martin JS, Bing Z, Sheng W, et al. Norcantharidin induces melanoma cell apoptosis through activation of TR3 dependent pathway. Cancer Biol Ther. 2011;12(11):1005–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kok SH, Hong CY, Kuo MY, Lee CH, Lee JJ, Lou IU, et al. Comparisons of norcantharidin cytotoxic effects on oral cancer cells and normal buccal keratinocytes. Oral Oncol. 2003;39(1):19–26.

    Article  CAS  PubMed  Google Scholar 

  22. Mei L, Sang W, Cui K, Zhang Y, Chen F, Li X, et al. Norcantharidin inhibits proliferation and promotes apoptosis via c-Met/Akt/mTOR pathway in human osteosarcoma cells. Cancer Sci. 2019;110:582–95.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Tu GG, Zhan JF, Lv QL, Wang JQ, Kuang BH, Li SH. Synthesis and antiproliferative assay of norcantharidin derivatives in cancer cells. Med Chem. 2014;10:376–81.

    Article  CAS  PubMed  Google Scholar 

  24. Wei CM, Wang BJ, Ma Y, Sun ZP, Li XL, Guo RC. Pharmacokinetics and biodistribution of ~ 3H-norcantharidin in mice. Acta Pharm Sin. 2007;42(5):516.

    CAS  Google Scholar 

  25. Wang GS, Zhong HY, Huang JK, Lu FX, Yang KZ, Liu ZC, et al. Treatment of 244 cases of primary hepatocellular carcinoma with norcantharidin. Chin Pharm J. 1986;02:90–3 (in Chinese).

    Google Scholar 

  26. Yang MY, Wu Z, Liang BY, Yu QP, Jian GF, Lin JM, et al. Clinical observation of oral and tumor center injection of norcantharidin in the treatment of primary liver cancer. J Pharm Res. 1992;01:45–7 (in Chinese).

    Google Scholar 

  27. Huang DT. Treatment for 41 cases of primary liver cancer with norcantharidin. Acta Med Sin. 1996;01:39–40.

    Google Scholar 

  28. Zhou YY, Yang HY, Liu GX, Deng WJ. Norcantharidin combined with chemotherapy for advanced hepatocellular carcinoma. Chin J Clin Oncol Rehabil. 1997;03:75 (in Chinese).

    Google Scholar 

  29. Liu A, Shan DQ. Combination of Ganfule and norcantharidin in the treatment of advanced hepatocellular carcinoma. Neimongol J Tradit Chin Med. 1998;04:6 (in Chinese).

    Google Scholar 

  30. Ling CQ, Chen J, Chen Z, Huang XQ, Gao XF, Zheng XM, et al. Clinical study of intratumoral injecting sustained-release norcantharidin-poloxamer 407 preparation in treatment of primary liver cancer. Acad J Second Mil Med Univ. 2000;11:1074–6.

    Google Scholar 

  31. Chen Z, Zhai XF, Jiang D, Ling CQ. Comparing therapeutic effects of intratumoral injection of norcantharidin poloxamer 407 slow released preparation and absolute ethanol on primary liver cancer. Acad J Second Mil Med Univ. 2001;07:606–8.

    Google Scholar 

  32. Zheng YL, Shi CL, He YX. Clinical study of norcantharidin in the treatment of liver cancer. Hebei Med. 2005;09:820–1 (in Chinese).

    Google Scholar 

  33. Luan ZP, Li XD, Ma M. Clinical study of norcantharidin injection combined with fluorouracil in the treatment of advanced liver cancer. Hebei Med J. 2005;07:542–3 (in Chinese).

    Google Scholar 

  34. Fan CX. The observation of the effects of disodium norcantharidate in the treatment of advanced primary liver cancer. J Basic Clin Oncol. 2010;23(01):50–1.

    Google Scholar 

  35. Jiang F, Cai RC, Xin Y. Clinical study of high-dose epimedium combined with norcantharidin in the treatment of primary liver cancer. Pract Clin J Integ Tradit Chin West Med. 2011;11(6):49 (in Chinese).

    CAS  Google Scholar 

  36. Chu YP, Shen L, Bai Y. Efficacy observation of liver cancer at the later stage for the elderly treated with integration of norcantharidin and Chinese medicine. World J Integ Tradit West Med. 2012;258(1):224–9.

    Google Scholar 

  37. Zhou WL, Kao J, Fan QL. Clinical observation of norcantharidin tablets in the treatment of 30 cases of liver metastasis from gastrointestinal cancer. Shandong Med J. 2005;20:32–3 (in Chinese).

    Google Scholar 

  38. Chen SH, Wang JH, Tan QH, Tian SY. Sodium norcantharidin combined with DF regimen in the treatment of advanced gastric carcinoma. J Basic Clin Oncol. 2013;26(04):311–3.

    Google Scholar 

  39. Zhang LT, Xiang H. Clinical efficacy of norcantharidin combined with conventional chemotherapy treating postoperative gastric cancer. Med Recapit. 2013;19(11):2087–8.

    Google Scholar 

  40. Zhao PZ. Go to a sodium cantharidate clinical observation of treatment of esophageal cancer with radiotherapy. Med J Chin People’s Health. 2010;22(13):1648–50.

    Google Scholar 

  41. Feng BH. Therapeutic effect of radiotherapy combined with sodium norcantharidin for stage III cervical cancer. Chin Foreign Med Treat. 2010;29(03):40–1 (in Chinese).

    Google Scholar 

  42. Wu GX, Yang ZH, Chen EB. Clinical observation of chemotherapy combined with compound cantharidin injection in the treatment of malignant lymphoma. J Chin Physician. 2005;10:1425–6 (in Chinese).

    Google Scholar 

  43. Chen WM, Zeng GY. Clinical research on norcantharidin injection combined with CTOP scheme for treating non-Hodgkin’s lymphoma. Fujian Med J. 2012;34(01):85–7.

    Google Scholar 

  44. Zhi XJ, Li GL. Clinical observation on 30 cases of advanced lung cancer treated with norcantharidin sodium. J Hebei North Univ. 2008;03:60.

    Google Scholar 

  45. Guan ZF. Clinical observation on 50 cases of advanced non-small cell lung cancer treated with sodium norcantharidin. J Qiqihar Med Univ. 2010;31(17):2727 (in Chinese).

    Google Scholar 

  46. Gong D, Wang MH, Zhang S, Wang XY, Chen MY, Fang F, et al. Aidi injection combined with paclitaxel-containing chemotherapy protocol in treatment of 42 patients with non-small-cell lung carcinoma in stage III–IV. Med Pharm J Chin PLA. 2014;26(08):81–4.

    Google Scholar 

  47. Li ZY, Ma QT, Zhang Y, Wang XC, Liu YL. Gemcitabine and ciplatin combined with norcantharidin sodium for the treatment of advanced NSCLC. Chin J Cancer Prev Treat. 2014;21(04):293–5.

    CAS  Google Scholar 

  48. Li YG, Wu JZ, Liu HM, Zhang LX. Effect of sodium cantharidate injection on immune function in patients with non-small cell lung cancer. Chin J Gerontol. 2015;35(06):1538–40 (in Chinese).

    Google Scholar 

  49. Ma Q, Feng Y, Deng K, Shao H, Sui T, Zhang X, et al. Unique responses of hepatocellular carcinoma and cholangiocarcinoma cell lines toward cantharidin and norcantharidin. J Cancer. 2018;9:2183–90.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Yang EB, Tang WY, Zhang K, Cheng LY, Mack PO. Norcantharidin inhibits growth of human HepG2 cell-transplanted tumor in nude mice and prolongs host survival. Cancer Lett. 1997;117:93–8.

    Article  CAS  PubMed  Google Scholar 

  51. Liao HF, Su SL, Chen YJ, Chou CH, Kuo CD. Norcantharidin preferentially induces apoptosis in human leukemic Jurkat cells without affecting viability of normal blood mononuclear cells. Food Chem Toxicol. 2007;45:1678–87.

    Article  CAS  PubMed  Google Scholar 

  52. Liao HF, Chen YJ, Chou CH, Wang FW, Kuo CD. Norcantharidin induces cell cycle arrest and inhibits progression of human leukemic Jurkat T cells through mitogen-activated protein kinase-mediated regulation of interleukin-2 production. Toxicol In Vitro. 2011;25(1):206–12.

    Article  CAS  PubMed  Google Scholar 

  53. Li KY, Shi CX, Huang JZ, Tang KL. Cisplatin plus norcantharidin alter the expression of TGF-β1/Smads signaling pathway in hepatocellular carcinoma. Bratisl Lek Listy. 2017;118:85–8.

    CAS  PubMed  Google Scholar 

  54. Lee YC, Lee LM, Yang CH, Lin AM, Huang YC, Hsu CC, et al. Norcantharidin suppresses cell growth and migration with enhanced anticancer activity of gefitinib and cisplatin in human non-small cell lung cancer cells. Oncol Rep. 2013;29:237–43.

    Article  CAS  PubMed  Google Scholar 

  55. Jiang YM, Meng ZZ, Yue GX, Chen JX. Norcantharidin induces HL-60 cells apoptosis in vitro. Evid Based Complement Altern Med. 2012;2012:154271.

    Google Scholar 

  56. Liu Y, Zahng J, You Z, Liao H. Inhibitory effect of norcantharidin combined with evodiamine on the growth of human hepatic carcinoma cell line HepG2 in vitro. Chin J Cell Mol Immunol. 2014;30(8):824–8.

    CAS  Google Scholar 

  57. Zhang QY, Yue XQ, Jiang YP, Han T, Xin HL. Author correction: FAM46C is critical for the anti-proliferation and pro-apoptotic effects of norcantharidin in hepatocellular carcinoma cells. Sci Rep. 2017;7(1):17576.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Sun ZX, Ma QW, Zhao TD, Wei YL, Wang GS, Li JS. Apoptosis induced by norcantharidin in human tumor cells. World J Gastroenterol. 2000;6(2):263–5.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Peng F, Wei YQ, Tian L, Yang L, Zhao X, Lu Y, et al. Induction of apoptosis by norcantharidin in human colorectal carcinoma cell lines: involvement of the CD95 receptor/ligand. J Cancer Res Clin Oncol. 2002;128(4):223–30.

    Article  CAS  PubMed  Google Scholar 

  60. Chen YJ, Kuo CD, Tsai YM, Yu CC, Wang GS, Liao HF. Norcantharidin induces anoikis through Jun-N-terminal kinase activation in CT26 colorectal cancer cells. Anticancer Drugs. 2008;19(1):55–64.

    Article  PubMed  CAS  Google Scholar 

  61. Mumby M. PP2A: unveiling a reluctant tumor suppressor. Cell. 2007;130(1):21–4.

    Article  CAS  PubMed  Google Scholar 

  62. Hart ME, Chamberlin AR, Walkom C, Sakoff JA, McCluskey A. Modified norcantharidins; synthesis, protein phosphatases 1 and 2A inhibition, and anticancer activity. Bioorg Med Chem Lett. 2004;14:1969–73.

    Article  CAS  PubMed  Google Scholar 

  63. Shou LM, Zhang QY, Li W, Xie X, Chen K, Lian L, et al. Cantharidin and norcantharidin inhibit the ability of MCF-7 cells to adhere to platelets via protein kinase C pathway-dependent downregulation of α2 integrin. Oncol Rep. 2013;30:1059–66.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Ayad Nagi G. CDKs give Cdc6 a license to drive into S phase. Cell. 2005;122(6):825–7.

    Article  CAS  PubMed  Google Scholar 

  65. Li JL, Cai YC, Hu ZM, Gao JM. Norcantharidin inhibits DNA replication initiation protein Cdc6 in cancer cells. Nan Fang Yi Ke Da Xue Xue Bao. 2010;30:1851–3.

    CAS  PubMed  Google Scholar 

  66. Chen S, Qu X, Wan P, Li QW, Wang Z, Guo F, et al. Norcantharidin inhibits pre-replicative complexes assembly of HepG2 cells. Am J Chin Med. 2013;41:665–82.

    Article  CAS  PubMed  Google Scholar 

  67. Fan YZ, Fu JY, Zhao ZM, Chen CQ. Influence of norcantharidin on proliferation, proliferation-related gene proteins proliferating cell nuclear antigen and Ki-67 of human gallbladder carcinoma GBC-SD cells. HBPD INT. 2004;3:603–7.

    CAS  PubMed  Google Scholar 

  68. Yang PY, Chen MF, Tsai CH, Hu DN, Chang FR, Wu YC. Involvement of caspase and MAPK activities in norcantharidin-induced colorectal cancer cell apoptosis. Toxicol In Vitro. 2010;24(3):766–75.

    Article  CAS  PubMed  Google Scholar 

  69. Janssens N, Janicot M, Perera T. The Wnt-dependent signaling pathways as target in oncology drug discovery. Invest New Drugs. 2006;24:263–80.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Chuang KA, Lieu CH, Tsai WJ, Wu MH, Chen YC, Liao JF, et al. Evaluation of anti-Wnt/β-catenin signaling agents by pGL4-TOP transfected stable cells with a luciferase reporter system. Braz J Med Biol Res. 2010;43:931–41.

    Article  CAS  PubMed  Google Scholar 

  71. Cimmino F, Scoppettuolo MN, Carotenuto M, De Antonellis P, Dato VD, De Vita G, et al. Norcantharidin impairs medulloblastoma growth by inhibition of Wnt/β-catenin signaling. J Neurooncol. 2012;106(1):59–70.

    Article  CAS  PubMed  Google Scholar 

  72. Lv H, Li Y, Du H, Fang J, Song X, Zhang J. The synthetic compound norcantharidin induced apoptosis in mantle cell lymphoma in vivo and in vitro through the PI3K-Akt-NF-κB signaling pathway. Evid Based Complement Altern Med. 2013;2013:461487.

    Google Scholar 

  73. Huang Y, Liu Q, Liu K, Yagasaki K, Zhang G. Suppression of growth of highly-metastatic human breast cancer cells by norcantharidin and its mechanisms of action. Cytotechnology. 2009;59(3):201–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chen YJ, Shieh CJ, Tsai TH, Kuo CD, Ho LT, Liu TY, et al. Inhibitory effect of norcantharidin, a derivative compound from blister beetles, on tumor invasion and metastasis in CT26 colorectal adenocarcinoma cells. Anticancer Drugs. 2005;16(3):293–9.

    Article  CAS  PubMed  Google Scholar 

  75. Deryugina EI, Quigley JP. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006;25:9–34.

    Article  CAS  PubMed  Google Scholar 

  76. Makrilia N, Kollias A, Manolopoulos L, Syrigos K. Cell adhesion molecules: role and clinical significance in cancer. Cancer Invest. 2009;27:1023–37.

    Article  CAS  PubMed  Google Scholar 

  77. Chen YJ, Chang WM, Liu YW, Lee CY, Jang YH, Kuo CD, et al. A small-molecule metastasis inhibitor, norcantharidin, downregulates matrix metalloproteinase-9 expression by inhibiting Sp1 transcriptional activity in colorectal cancer cells. Chem Biol Interact. 2009;181(3):440–6.

    Article  CAS  PubMed  Google Scholar 

  78. Peng C, Li Z, Niu Z, Niu W, Xu Z, Gao H, et al. Norcantharidin suppresses colon cancer cell epithelial–mesenchymal transition by inhibiting the αvβ6-ERK-Ets1 signaling pathway. Sci Rep. 2016;6(1):20500.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Loboda A, Nebozhyn MV, Watters JW, Buser CA, Shaw PM, Huang PS, et al. EMT is the dominant program in human colon cancer. BMC Med Genomics. 2011;4:9.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Guo J, Wu Y, Yang L, Du J, Gong K, Chen W, et al. Repression of YAP by NCTD disrupts NSCLC progression. Oncotarget. 2017;8:2307–19.

    Article  PubMed  Google Scholar 

  81. Gao Y, Li W, Liu R, Guo Q, Li J, Bao Y, et al. Norcantharidin inhibits IL-6-induced epithelial–mesenchymal transition via the JAK2/STAT3/TWIST signaling pathway in hepatocellular carcinoma cells. Oncol Rep. 2017;38:1224–32.

    Article  CAS  PubMed  Google Scholar 

  82. Folkman Judah. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1995;1(1):27–30.

    Article  CAS  PubMed  Google Scholar 

  83. Fan YZ, Chen CQ, Zhao ZM, Sun W. Effects of norcantharidin on angiogenesis of human gallbladder carcinoma and its anti-angiogenic mechanisms. Nat Med J Chin. 2006;86:693–9.

    CAS  Google Scholar 

  84. Zhang JT, Fan YZ, Chen CQ, Zhao ZM, Sun W. Norcantharidin: a potential antiangiogenic agent for gallbladder cancers in vitro and in vivo. Int J Oncol. 2012;40:1501–14.

    CAS  PubMed  Google Scholar 

  85. Yu T, Hou F, Liu M, Zhou L, Li D, Liu J, et al. Norcantharidin anti-angiogenesis activity possibly through an endothelial cell pathway in human colorectal cancer. Asian Pac J Cancer Prev. 2012;13(2):499–503.

    Article  CAS  PubMed  Google Scholar 

  86. Zhang L, Ji Q, Liu X, Chen X, Chen Z, Qiu Y, et al. Norcantharidin inhibits tumor angiogenesis via blocking VEGFR2/MEK/ERK signaling pathways. Cancer Sci. 2013;104(5):604–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Maniotis AJ, Folberg R, Hess A, Seftor EA, Gardner LM, Pe’er J, et al. Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry. Am J Pathol. 1999;155:739–52.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Albini A, Melchiori A, Santi L, Liotta LA, Brown PD, Stetler-Stevenson WG. Tumor cell invasion inhibited by TIMP-2. J Natl Cancer Inst. 1991;83(11):775–9.

    Article  CAS  PubMed  Google Scholar 

  89. Fan YZ, Sun W. Molecular regulation of vasculogenic mimicry in tumors and potential tumor-target therapy. World J Gastrointest Surg. 2010;2:117–27.

    Article  PubMed  PubMed Central  Google Scholar 

  90. Seftor RE, Seftor EA, Koshikawa N, Meltzer PS, Gardner LM, Bilban M, et al. Cooperative interactions of laminin 5 gamma2 chain, matrix metalloproteinase-2, and membrane type-1-matrix/metalloproteinase are required for mimicry of embryonic vasculogenesis by aggressive melanoma. Cancer Res. 2001;61:6322–7.

    CAS  PubMed  Google Scholar 

  91. Zhang JT, Sun W, Zhang WZ, Ge CY, Liu ZY, Zhao ZM, et al. Norcantharidin inhibits tumor growth and vasculogenic mimicry of human gallbladder carcinomas by suppression of the PI3-K/MMPs/Ln-5γ2 signaling pathway. BMC Cancer. 2014;14:193.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Zhu W, Sun W, Zhang JT, Liu ZY, Li XP, Fan YZ. Norcantharidin enhances TIMP-2 anti-vasculogenic mimicry activity for human gallbladder cancers through downregulating MMP-2 and MT1-MMP. Int J Oncol. 2015;46:627–40.

    Article  CAS  PubMed  Google Scholar 

  93. Wang Z, You D, Lu M, He Y, Yan S. Inhibitory effect of norcantharidin on melanoma tumor growth and vasculogenic mimicry by suppressing MMP-2 expression. Oncol Lett. 2017;13:1660–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Achen MG, Stacker SA. Tumor lymphangiogenesis and metastatic spread-new players begin to emerge. Int J Cancer. 2006;119:1755–60.

    Article  CAS  PubMed  Google Scholar 

  95. Veikkola T, Jussila L, Makinen T, Karpanen T, Jeltsch M, Petrova TV, et al. Signalling via vascular endothelial growth factor receptor-3 is sufficient for lymphangiogenesis in transgenic mice. EMBO J. 2001;20:1223–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Siegfried G, Basak A, Cromlish JA, Benjannet S, Marcinkiewicz J, Chrétien M, et al. The secretory proprotein convertases furin, PC5, and PC7 activate VEGF-C to induce tumorigenesis. J Clin Invest. 2003;111:1723–32.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Joukov V, Sorsa T, Kumar V, Jeltsch M, Claesson-Welsh L, Cao Y, et al. Proteolytic processing regulates receptor specificity and activity of VEGF-C. EMBO J. 1997;16:3898–911.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Liu ZY, Qiu HO, Yuan XJ, Ni YY, Sun JJ, Jing W, et al. Suppression of lymphangiogenesis in human lymphatic endothelial cells by simultaneously blocking VEGF-C and VEGF-D/VEGFR-3 with norcantharidin. Int J Oncol. 2012;41:1762–72.

    Article  CAS  PubMed  Google Scholar 

  99. Yuan X, Chen Y, Li X, Zhang G, Jin D, Zhao H, et al. Norcantharidin inhibits lymphangiogenesis by downregulating the expression of VEGF-C and VEGF-D in human dermal lymphatic endothelial cells in vitro. Pharmacology. 2015;95:1–9.

    Article  CAS  PubMed  Google Scholar 

  100. Li XP, Jing W, Sun JJ, Liu ZY, Zhang JT, Sun W, et al. A potential small-molecule synthetic antilymphangiogenic agent norcantharidin inhibits tumor growth and lymphangiogenesis of human colonic adenocarcinomas through blocking VEGF-A,-C,-D/VEGFR-2,-3 “multi-points priming” mechanisms in vitro and in vivo. BMC Cancer. 2015;15(1):527.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  101. Gottesman MM, Fojo T, Bates SE. Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2002;2(1):48–58.

    Article  CAS  PubMed  Google Scholar 

  102. Li Y, Gao X, Yu Z, Liu B, Pan W, Li N, et al. Reversing multidrug resistance by multiplexed gene silencing for enhanced breast cancer chemotherapy. ACS Appl Mater Interfaces. 2018;10:15461–6.

    Article  CAS  PubMed  Google Scholar 

  103. Chen YJ, Kuo CD, Chen SH, Chen WJ, Huang WC, Chao KS, et al. Small-molecule synthetic compound norcantharidin reverses multi-drug resistance by regulating Sonic hedgehog signaling in human breast cancer cells. PLoS ONE. 2012;7:e37006.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Sims-Mourtada J, Izzo JG, Ajani J, Chao KS. Sonic Hedgehog promotes multiple drug resistance by regulation of drug transport. Oncogene. 2007;26:5674–9.

    Article  CAS  PubMed  Google Scholar 

  105. Miyashita T, Reed JC. Bcl-2 gene transfer increases relative resistance of S49.1 and WEHI7.2 lymphoid cells to cell death and DNA fragmentation induced by glucocorticoids and multiple chemotherapeutic drugs. Cancer Res. 1992;52:5407–11.

    CAS  PubMed  Google Scholar 

  106. Simonian PL, Grillot DA, Nuñez G. Bcl-2 and Bcl-XL can differentially block chemotherapy-induced cell death. Blood. 1997;90:1208–16.

    Article  CAS  PubMed  Google Scholar 

  107. Noutomi T, Chiba H, Itoh M, Toyota H, Mizuguchi J. Bcl-xL confers multi-drug resistance in several squamous cell carcinoma cell lines. Oral Oncol. 2002;38(1):41–8.

    Article  CAS  PubMed  Google Scholar 

  108. Kok SH, Cheng SJ, Hong CY, Lee JJ, Lin SK, Kuo YS, et al. Norcantharidin-induced apoptosis in oral cancer cells is associated with an increase of proapoptotic to antiapoptotic protein ratio. Cancer Lett. 2005;217(1):1–52.

    Article  CAS  Google Scholar 

  109. Oltersdorf T, Elmore SW, Shoemaker AR, Armstrong RC, Augeri DJ, Belli BA, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature. 2005;435:677–81.

    Article  CAS  PubMed  Google Scholar 

  110. Tse C, Shoemaker AR, Adickes J, Anderson MG, Chen J, Jin S, et al. ABT-263: a potent and orally bioavailable Bcl-2 family inhibitor. Cancer Res. 2008;68:3421–8.

    Article  CAS  PubMed  Google Scholar 

  111. Zhang S, Li G, Ma X, Wang Y, Liu G, Feng L, et al. Norcantharidin enhances ABT-737-induced apoptosis in hepatocellular carcinoma cells by transcriptional repression of Mcl-1. Cell Signal. 2012;24:1803–9.

    Article  CAS  PubMed  Google Scholar 

  112. Ren J, Li G, Zhao W, Lin L, Ye T. Norcantharidin combined with ABT-737 for hepatocellular carcinoma: therapeutic effects and molecular mechanisms. World J Gastroenterol. 2016;22:3962–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Wang X, Gu Z, Li G, Zhang S, Cao Z, Yang Z, et al. Norcantharidin enhances ABT-263-mediated anticancer activity in neuroblastoma cells by upregulation of Noxa. Oncol Rep. 2014;32:716–22.

    Article  PubMed  Google Scholar 

  114. Jackman D, Pao W, Riely GJ, Engelman JA, Kris MG, Jänne PA, et al. Clinical definition of acquired resistance to epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. J Clin Oncol. 2010;28:357–60.

    Article  CAS  PubMed  Google Scholar 

  115. Yano S, Yamada T, Takeuchi S, Tachibana K, Minami Y, Yatabe Y, et al. Hepatocyte growth factor expression in EGFR mutant lung cancer with intrinsic and acquired resistance to tyrosine kinase inhibitors in a Japanese cohort. J Thorac Oncol. 2011;6(12):2011–7.

    Article  PubMed  Google Scholar 

  116. Wu H, Fan F, Liu Z, Shen C, Wang A, Lu Y. Norcantharidin combined with EGFR-TKIs overcomes HGF-induced resistance to EGFR-TKIs in EGFR mutant lung cancer cells via inhibition of Met/PI3k/Akt pathway. Cancer Chemother Pharmacol. 2015;76:307–15.

    Article  CAS  PubMed  Google Scholar 

  117. Varley KE, Gertz J, Bowling KM, Parker SL, Reddy TE, Pauli-Behn F, et al. Dynamic DNA methylation across diverse human cell lines and tissues. Genome Res. 2013;23:555–67.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Lund AH, Lohuizen MV. Epigenetics and cancer. Gene Dev. 2004;18(19):2315–35.

    Article  CAS  PubMed  Google Scholar 

  119. Yang X, Gao L, Zhang S. Comparative pan-cancer DNA methylation analysis reveals cancer common and specific patterns. Brief Bioinform. 2017;18:761–73.

    CAS  PubMed  Google Scholar 

  120. Hesson LB, Cooper WN, Latif F. The role of RASSF1A methylation in cancer. Dis Markers. 2013;23(1–2):73.

    Google Scholar 

  121. Wang Y, Xu M, Di ZH, Zhang J, Mao XQ, Sun HB. Regulation of demethylation and re-expression of RASSF1A gene in hepatocellular carcinoma cell lines treated with NCTD in vitro. J Cancer Res Ther. 2015;11:818–22.

    Article  CAS  PubMed  Google Scholar 

  122. Mazieres J, He B, You L, Xu Z, Jablons DM. Wnt signaling in lung cancer. Cancer Lett. 2005;222(1):1–10.

    Article  CAS  PubMed  Google Scholar 

  123. Polakis P. Wnt signaling in cancer. Cold Spring Harb Perspect Biol. 2012;4(5):1–10.

    Article  CAS  Google Scholar 

  124. Kim SA, Kwak J, Nam HY, Chun SM, Lee BW, Lee HJ, et al. Promoter methylation of WNT inhibitory factor-1 and expression pattern of WNT/β-catenin pathway in human astrocytoma: pathologic and prognostic correlations. Mod Pathol. 2013;26:626–39.

    Article  CAS  PubMed  Google Scholar 

  125. Xie J, Zhang Y, Hu X, Lv R, Xiao D, Jiang L, et al. Norcantharidin inhibits Wnt signal pathway via promoter demethylation of WIF-1 in human non-small cell lung cancer. Med Oncol. 2015;32:145.

    Article  PubMed  CAS  Google Scholar 

  126. Xie D, Xie J, Wan Y, Ma L, Qi X, Wang K, et al. Norcantharidin blocks Wnt/β-catenin signaling via promoter demethylation of WIF-1 in glioma. Oncol Rep. 2016;35:2191–7.

    Article  CAS  PubMed  Google Scholar 

  127. Zhao Q, Qian Y, Li R, Tan B, Han H, Liu M, et al. Norcantharidin facilitates LPS-mediated immune responses by up-regulation of AKT/NF-κB signaling in macrophages. PLoS ONE. 2012;7:e44956.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Chen YC, Chang SC, Wu MH, Chuang KA, Wu JY, Tsai WJ, et al. Norcantharidin reduced cyclins and cytokines production in human peripheral blood mononuclear cells. Life Sci. 2009;84:218–26.

    Article  CAS  PubMed  Google Scholar 

  129. Bergmann C, Strauss L, Wang Y, Szczepanski MJ, Lang S, Johnson JT, et al. T regulatory type 1 cells in squamous cell carcinoma of the head and neck: mechanisms of suppression and expansion in advanced disease. Clin Cancer Res. 2008;14(12):3706–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  130. Mack P, Ha XF, Cheng LY. Efficacy of intra-arterial norcantharidin in suppressing tumour 14C-labelled glucose oxidative metabolism in rat Morris hepatoma. HPB Surg. 1996;10:65–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Wu LT, Chung JG, Chen JC, Tsauer W. Effect of norcantharidin on N-acetyltransferase activity in HepG2 cells. Am J Chin Med. 2001;29:161–72.

    Article  CAS  PubMed  Google Scholar 

  132. Zhang H, Jiang Y, Ni X, Chen L, Wu M, Liu J, et al. Glycyrrhetinic acid- modified norcantharidin nanoparticles for active targeted therapy of hepatocellular carcinoma. J Biomed Nanotechnol. 2018;14:114–26.

    Article  CAS  PubMed  Google Scholar 

  133. Li JL, Cai YC, Liu XH, Xian LJ. Norcantharidin inhibits DNA replication and induces apoptosis with the cleavage of initiation protein Cdc6 in HL-60 cells. Anticancer Drugs. 2006;17(3):307.

    Article  CAS  PubMed  Google Scholar 

  134. Li XQ, Shao SH, Fu GL, Han SH, Gao H. Study on norcantharidin-induced apoptosis in SMMC-7721 cells through mitochondrial pathways. Chin J Integr Med. 2010;16(5):448–52.

    Article  CAS  PubMed  Google Scholar 

  135. Li XQ, Shao SH, Han XH, Fan ZZ, Sun J, Yin PH, et al. Norcantharidin induces apoptosis in SMMC-7721 cells via the c-Jun-NH2-terminal kinase signaling pathways. Chin J Hepatol. 2010;18(2):146–7.

    CAS  Google Scholar 

  136. Chang C, Zhu Y, Tang X, Tao W. The anti-proliferative effects of norcantharidin on human HepG2 cells in cell culture. Mol Biol Rep. 2011;38:163–9.

    Article  CAS  PubMed  Google Scholar 

  137. Yeh CH, Yang YY, Huang YF, Chow KC, Chen MF. Induction of apoptosis in human Hep3B hepatoma cells by norcantharidin through a p53 independent pathway via TRAIL/DR5 signal transduction. Chin J Integr Med. 2012;18:676–82.

    Article  CAS  PubMed  Google Scholar 

  138. Xiong X, Wu M, Zhang H, Li J, Lu B, Guo Y, et al. Atg5 siRNA inhibits autophagy and enhances norcantharidin-induced apoptosis in hepatocellular carcinoma. Int J Oncol. 2015;47:1321–8.

    Article  CAS  PubMed  Google Scholar 

  139. Ren J, Li G, Zhao W, Lin L, Ye T. Norcantharidin combined with ABT-737 for hepatocellular carcinoma: therapeutic effects and molecular mechanisms. World J Gastroenterol. 2016;22(15):3962–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Chen YL, Hung MH, Chu PY, Chao TI, Tsai MH, Chen LJ, et al. Protein phosphatase 5 promotes hepatocarcinogenesis through interaction with AMP-activated protein kinase. Biochem Pharmacol. 2017;138:49–60.

    Article  CAS  PubMed  Google Scholar 

  141. Wang D, Yang C, Wang Z, Yang Y, Li D, Ding X, et al. Norcantharidin combined with Coix seed oil synergistically induces apoptosis and inhibits hepatocellular carcinoma growth by downregulating regulatory T cells accumulation. Sci Rep. 2017;7:9373.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  142. Fan YZ, Fu JY, Zhao ZM, Chen CQ. The in vitro effect of norcantharidin on proliferation and invasion of human gallbladder carcinoma GBC-SD cells and its mechanism. Zhonghua Zhong Liu Za Zhi. 2004;26:271–4.

    CAS  PubMed  Google Scholar 

  143. Fan YZ, Fu JY, Zhao ZM, Chen CQ. Inhibitory effect of norcantharidin on the growth of human gallbladder carcinoma GBC-SD cells in vitro. HBPD INT. 2007;6(1):72–80.

    CAS  PubMed  Google Scholar 

  144. Fan YZ, Zhao ZM, Fu JY, Chen CQ. Anti-tumor mechanism of norcantharidin for the implanted tumors of human gallbladder carcinoma in nude mice in vivo. Chin J Surg. 2006;44(9):618–22.

    PubMed  Google Scholar 

  145. Fan YZ, Zhao ZM, Fu JY, Chen CQ, Sun W. Norcantharidin inhibits growth of human gallbladder carcinoma xenografted tumors in nude mice by inducing apoptosis and blocking the cell cycle in vivo. HBPD INT. 2010;9(4):414–22.

    CAS  PubMed  Google Scholar 

  146. Peng C, Liu X, Liu E, Xu K, Niu W, Chen R, et al. Norcantharidin induces HT-29 colon cancer cell apoptosis through the alphavbeta6-extracellular signal-related kinase signaling pathway. Cancer Sci. 2010;100(12):2302–8.

    Article  CAS  Google Scholar 

  147. Qiu P, Wang S, Liu M, Ma H, Zeng X, Zhang M, et al. Norcantharidin inhibits cell growth by suppressing the expression and phosphorylation of both EGFR and c-Met in human colon cancer cells. BMC Cancer. 2017;17(1):55.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  148. Williams LA, Möller W, Merisor E, Kraus W, Rösner H. In vitro anti- proliferation/cytotoxic activity of cantharidin (Spanish Fly) and related derivatives. West Indian Med J. 2003;52(1):10–3.

    CAS  PubMed  Google Scholar 

  149. Yang PY, Chen MF, Kao YH, Hu DN, Chang FR, Wu YC. Norcantharidin induces apoptosis of breast cancer cells: involvement of activities of mitogen activated protein kinases and signal transducers and activators of transcription. Toxicol In Vitro. 2011;25(3):699–707.

    Article  PubMed  CAS  Google Scholar 

  150. Liu D, Shi P, Yin X, Chen Z, Zhang X. Effect of norcantharidin on the human breast cancer Bcap-37 cells. Connect Tissue Res. 2012;53(6):5.

    Article  CAS  Google Scholar 

  151. Zheng LC, Yang MD, Kuo CL, Lin CH, Fan MJ, Chou YC, et al. Norcantharidin-induced apoptosis of AGS human gastric cancer cells through reactive oxygen species production, and caspase- and mitochondria-dependent signaling pathways. Anticancer Res. 2016;36(11):6031–42.

    Article  CAS  PubMed  Google Scholar 

  152. An WW, Wang MW, Tashiro S, Onodera S, Ikejima T. Norcantharidin induces human melanoma A375-S2 cell apoptosis through mitochondrial and caspase pathways. J Korean Med Sci. 2004;19(4):560–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. An WW, Wang MW, Tashiro S, Onodera S, Ikejima T. Mitogen-activated protein kinase-dependent apoptosis in norcan-tharidin-treated A375-S2 cells is proceeded by the activation of protein kinase C. Chin Med J. 2005;118:198–203.

    CAS  PubMed  Google Scholar 

  154. Song XN, Du HF, Yu LJ, Meng YF, Lü HY, Sun LX, et al. Norcantharidin potentialize the chemosensitivity of adriamycin through the NF-κB/IκBα signaling pathway. Zhonghua Xue Ye Xue Za Zhi. 2011;32(12):809–13.

    CAS  PubMed  Google Scholar 

  155. He Y, Yan D, Zheng D, Hu Z, Li H, Li J. Cell division cycle 6 promotes mitotic slippage and contributes to drug resistance in paclitaxel-treated cancer cells. PLoS ONE. 2016;11:e0162633.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  156. Luan J, Duan H, Liu Q, Yagasaki K, Zhang G. Inhibitory effects of norcantharidin against human lung cancer cell growth and migration. Cytotechnology. 2010;62(4):349–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Zheng J, Du W, Song LJ, Zhang R, Sun LG, Chen FG, et al. Norcantharidin induces growth inhibition and apoptosis of glioma cells by blocking the Raf/MEK/ERK pathway. World J Surg Oncol. 2014;12(1):207.

    Article  PubMed  PubMed Central  Google Scholar 

  158. Han Z, Li B, Wang J, Zhang X, Li Z, Dai L, et al. Norcantharidin inhibits SK-N-SH neuroblastoma cell growth by induction of autophagy and apoptosis. Technol Cancer Res Treat. 2017;16:33–44.

    Article  CAS  PubMed  Google Scholar 

  159. An WW, Gong XF, Wang MW, Tashiro S, Onodera S, Ikejima T. Norcantharidin induces apoptosis in HeLa cells through caspase, MAPK, and mitochondrial pathways. Acta Pharmacol Sin. 2004;25:1502–8.

    CAS  PubMed  Google Scholar 

  160. Dong X, Li JC, Jiang YY, Xia MY, Tashiro S, Onodera S, et al. P38-NF-κB-promoted mitochondria-associated apoptosis and G2/M cell cycle arrest in norcantharidin-treated HeLa cells. J Asian Nat Prod Res. 2012;14:1008–19.

    Article  CAS  PubMed  Google Scholar 

  161. Yu CC, Ko FY, Yu CS, Lin CC, Huang YP, Yang JS, et al. Norcantharidin triggers cell death and DNA damage through S-phase arrest and ROS-modulated apoptotic pathways in TSGH 8301 human urinary bladder carcinoma cells. Int J Oncol. 2012;41:1050–60.

    Article  CAS  PubMed  Google Scholar 

  162. Chen S, Wan P, Ding W, Li F, He C, Chen P, et al. Norcantharidin inhibits DNA replication and induces mitotic catastrophe by degrading initiation protein Cdc6. Int J Mol Med. 2013;32:43–50.

    Article  PubMed  CAS  Google Scholar 

  163. Shen B, He PJ, Shao CL. Norcantharidin induced DU145 cell apoptosis through ROS-mediated mitochondrial dysfunction and energy depletion. PLoS ONE. 2013;8:e84610.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  164. Xiao W, Dai B, Zhu Y, Ye D. Norcantharidin induces autophagy-related prostate cancer cell death through Beclin-1 upregulation by miR-129-5p suppression. Tumor Biol. 2016;37(12):15643–8.

    Article  CAS  Google Scholar 

  165. Yang PY, Hu DN, Kao YH, Lin IC, Chou CY, Wu YC. Norcantharidin induces apoptosis in human prostate cancer cells through both intrinsic and extrinsic pathways. Pharmacol Rep. 2016;68:874–80.

    Article  CAS  PubMed  Google Scholar 

  166. Lin CL, Chen CM, Lin CL, Cheng CW, Lee CH, Hsieh YH. Norcantharidin induces mitochondrial-dependent apoptosis through Mcl-1 inhibition in human prostate cancer cells. Biochim Biophys Acta Mol Cell Res. 2017;1864(10):1867.

    Article  CAS  PubMed  Google Scholar 

  167. Zhu Y, Mi Y, Wang Z, Jia X, Jin Z. Norcantharidin inhibits viability and induces cell cycle arrest and apoptosis in osteosarcoma. Oncol Lett. 2019;17:456–61.

    CAS  PubMed  Google Scholar 

  168. Hong CY, Huang SC, Lin SK, Lee JJ, Chueh LL, Lee CH, et al. Norcantharidin-induced post-G2/M apoptosis is dependent on wild-type p53 gene. Biochem Biophys Res Commun. 2000;276:278–85.

    Article  CAS  PubMed  Google Scholar 

  169. Chen F, Wang S, Wei Y, Wu J, Huang G, Chen J, et al. Norcantharidin modulates the miR-30a/Metadherin/AKT signaling axis to suppress proliferation and metastasis of stromal tumor cells in giant cell tumor of bone. Biomed Pharmacother. 2018;103:1092–100.

    Article  CAS  PubMed  Google Scholar 

  170. Yeh CB, Hsieh MJ, Hsieh YH, Chien MH, Chiou HL, Yang SF. Antimetastatic effects of norcantharidin on hepatocellular carcinoma by transcriptional inhibition of MMP-9 through modulation of NF-kB activity. PLoS ONE. 2012;7:e31055.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  171. Yue M, Pacheco G, Cheng T, Li J, Wang Y, Henske EP, et al. Evidence supporting a lymphatic endothelium origin for angiomyolipoma, a TSC2(−) tumor related to lymphangioleiomyomatosis. Am J Pathol. 2016;186:1825–36.

    Article  PubMed  PubMed Central  Google Scholar 

  172. Zhao L, Yang G, Bai H, Zhang M, Mou D. NCTD promotes Birinapant-mediated anticancer activity in breast cancer cells by downregulation of c-FLIP. Oncotarget. 2017;8:26886–95.

    Article  PubMed  PubMed Central  Google Scholar 

  173. Wang WJ, Wu MY, Shen M, Zhi Q, Liu ZY, Gong FR, et al. Cantharidin and norcantharidin impair stemness of pancreatic cancer cells by repressing the β-catenin pathway and strengthen the cytotoxicity of gemcitabine and erlotinib. Int J Oncol. 2015;47:1912–22.

    Article  CAS  PubMed  Google Scholar 

  174. Du HF, Yu LJ, Meng YF, Lv HY, Meng J, Song XN, et al. Norcantharidin enhances bortezomib-antimyeloma activity in multiple myeloma cells in vitro and in nude mouse xenografts. Leuk Lymphoma. 2013;54(3):607–18.

    Article  CAS  PubMed  Google Scholar 

  175. Lu S, Gao Y, Huang X, Wang X. Cantharidin exerts anti-hepatocellular carcinoma by miR-214 modulating macrophage polarization. Int J Biol Sci. 2014;10(4):415–25.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  176. Zhu J, Zhang W, Wang DD, Li SZ, Wu W. Preparation and characterization of norcantharidin liposomes modified with stearyl glycyrrhetinate. Exp Ther Med. 2018;16:1639–46.

    PubMed  PubMed Central  Google Scholar 

  177. Chi JH, Jiang ZW, Chen XT, Peng YF, Liu WH, Han BS, et al. Studies on anti-hepatocarcinoma effect, pharmacokinetics and tissue distribution of carboxymethyl chitosan based norcantharidin conjugates. Carbohydr Polym. 2019;226:115297.

    Article  CAS  PubMed  Google Scholar 

  178. Chi JH, Jiang ZW, Qiao J, Peng YF, Liu WS, Han BQ. Synthesis and anti-metastasis activities of norcantharidin-conjugated carboxymethyl chitosan as a novel drug delivery system. Carbohydr Polym. 2019;214:80–9.

    Article  CAS  PubMed  Google Scholar 

  179. Chang MC, Wu JY, Liao HF, Chen YJ, Guo CD. N-Farnesyloxy-norcantharimide inhibits progression of human leukemic Jurkat T cells through regulation of mitogen-activated protein kinase and interleukin-2 production. Anticancer Drugs. 2015;26:1034–42.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by funds from the National Nature Science Foundation of China (Nos. 30672073, 81072004 and 81372614), the Natural Science Foundation Project in Shanghai (No. 13ZR1432300) and the Science and Technology Commission Foundation in Shanghai (Nos. 19411966300 and 19140902302).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed in the preparation of this manuscript. MSP and JC made contributions to acquisition, compiling and analysis of the data, writing this manuscript. YZF was responsible for design and revising of this manuscript. YZF is the corresponding author and the guarantor. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Yue-Zu Fan.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

The authors consent for the publication of this review.

Competing interests

The authors declare that they have no potential conflicts of interest.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Pan, MS., Cao, J. & Fan, YZ. Insight into norcantharidin, a small-molecule synthetic compound with potential multi-target anticancer activities. Chin Med 15, 55 (2020). https://doi.org/10.1186/s13020-020-00338-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13020-020-00338-6

Keywords