Abstract

Objectives

Cinobufagin is a natural active ingredient isolated from the traditional Chinese medicine Venenum Bufonis (Chinese: Chansu), which is the dried secretion of the postauricular gland or skin gland of the Bufo gargarizans Cantor or Bufo melanostictus Schneider. There is increasing evidence indicating that cinobufagin plays an important role in the treatment of cancer. This article is to review and discuss the antitumor pharmacological effects and mechanisms of cinobufagin, along with a description of its toxicity and pharmacokinetics.

Methods

The public databases including PubMed, China National Knowledge Infrastructure and Elsevier were referenced, and ‘cinobufagin’, ‘Chansu’, ‘Venenum Bufonis’, ‘anticancer’, ‘cancer’, ‘carcinoma’, and ‘apoptosis’ were used as keywords to summarize the comprehensive research and applications of cinobufagin published up to date.

Key findings

Cinobufagin can induce tumour cell apoptosis and cycle arrest, inhibit tumour cell proliferation, migration, invasion and autophagy, reduce angiogenesis and reverse tumour cell multidrug resistance, through triggering DNA damage and activating the mitochondrial pathway and the death receptor pathway.

Conclusions

Cinobufagin has the potential to be further developed as a new drug against cancer.

Introduction

Cancer is the second leading cause of death in the world.[1] Around 19.3 million new cases of malignant tumours and 10 million deaths were detected worldwide in 2020, which means that 1/5 of the world’s population will suffer from cancer in their lifetime.[2] Modern medicine treats cancer mainly by surgery, radiotherapy and chemotherapy, but single surgery is still limited due to postoperative recurrence and cancer cell metastasis, and so on.[3, 4] Presently, the effects of cancer treatment are not very ideal and there is a long way to go to achieve ideal therapy.

China has a long history of using Chinese medicine to treat cancer, with records of malignant tumours dating back 3500 years to the Yin and Zhou dynasties.[5] Based on the theory of traditional Chinese medicine, tumours are considered to be lumps in the abdomen causing distension and pain. Therefore, herbal medicines are often used to treat cancer by softening hard knots, activating blood circulation and removing blood stasis, and clearing heat and detoxification, such as Curcuma longa L.,[6]  Epimedium brevicornu Maxim.,[7]  Astragalus membranaceus (Fisch.) Bunge,[8]  Panax ginseng C. A. Meyer,[9]  Salvia miltiorrhiza Bge. [10] andVenenum Bufonis[11]. In addition to inhibiting tumour growth, Chinese medicine can also regulate immunity, enhance the effects of radiotherapy and chemotherapy, as well as reduce the side effects of radiotherapy and chemotherapy, and improve the quality of survival, taking advantage of the whole process of cancer rather than the terminal stage.[12] Therefore, Chinese medicine is of great importance in the treatment of malignant tumours.

Venenum Bufonis, also known as Chansu and Toad venom, is the dried secretion of the postauricular gland or skin gland of the Bufo gargarizans Cantor or Bufo melanostictus Schneider displaying functions of detoxification, pain relief and enlightenment, and has been used for hundreds of years to treat various diseases in China,[13, 14] although it is now commonly used for anaesthesia, pain relief, anticancer, etc.[15, 16] Chansu is a mixture that contains up to 96 types of bufadienolide monomers and 23 types of indole alkaloids, such as bufalin, cinobufagin, resibufogenin, telocinobufagin and arenobufagin.[17, 18] Cinobufagin is one of the main active ingredients, whose structure is shown in Figure 1. Numerous studies have displayed that cinobufagin has good anticancer activity.[19–21] It is widely used in the treatment of various malignant tumours such as liver cancer, gastric cancer, non-small cell lung cancer, colorectal cancer, rectal cancer, breast cancer, prostate cancer, cervical cancer, osteosarcoma, melanoma, nasopharyngeal cancer, oral squamous cell carcinoma, and glioblastoma. Its pharmacological effects include induction of tumour cell apoptosis and cell cycle arrest, inhibition of tumour cell proliferation, migration and invasion, reduction of cancer angiogenesis and autophagy, reversal of tumour cell multidrug resistance, triggering DNA damage and relief of cancer pain. This review describes in detail cinobufagin and its antitumour and other biomedical effects and mechanisms, and discusses its toxic effects and pharmacokinetics to explore its potential for development as a tumour suppressor, providing new ideas and insights into the current research of such compounds as shown in Figure 2.

The structure of cinobufagin.
Figure 1

The structure of cinobufagin.

Summary of this review. Source: By Figdraw.
Figure 2

Summary of this review. Source: By Figdraw.

Anticancer Effects

Cancer development is associated with anomalous proliferation of genetically damaged cells.[22] Drug treatments such as cinobufagin can alter the abnormal activity of the cells through specific mechanisms. Accumulating evidence has revealed the antitumour effects of cinobufagin and herein we summarize the reported mechanisms and presented them more intuitively as outlined in Figure 3. and Table 1.

Table 1

Pharmacological effects and mechanisms of cinobufagin in various cancers

Cancer typeCell line (in vitro)Animal model (in vivo)DoseEffectMechanismRefs.
Colorectal cancerHCT116, RKO, SW480HCT116(xenograft in mice)0.5 mg/kg, 1 mg/kgViability and proliferation↓; apoptosis↑; invasion and metastases↓STAT3↓; Bax↑; Bcl-2↓; caspase-3↑; EMT ↓; E-Cadherin↑; Vimentin↓[23]
HCT116, HT29HCT116(xenograft in mice)10 mg/kgProliferation↓; G2/M cell cycle arrest↑; apoptosis↑ER↑; mTORC1↓; caspase-12↑[24]
SW480, EOMA (xenograft in mice)2 mg/kg, 4 mg/kgAngiogenesis↓; apoptosis↑mOTRC1↓; HIF-1α↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[25]
SW480, SW1116Apoptosis↑; G2/M cell cycle arrest↑p53↓; p21↓; ROS↑[26]
Colon cancerLoVo/ADR, HCT116/L and Cao-2/ADRDrug resistance↓p-gp↓[27]
HCT116, HT29Invasion and metastases↓Cortactin↓[28]
HCT116, HT30HCT116(xenograft in mice)1.5 mg/kgApoptosis↑HIF-1α↓[29]
Oral squamous cell carcinomaCAL-27Invasion and metastases↓; apoptosis↑STAT3↓; ANO1↓; ANO1 mRNA↓; caspase-3↑; cleaved PARP-1↑[30]
OsteosarcomaU2OS, 143B143B (xenograft in mice)2 mg/kg, 6 mg/kgViability↓; apoptosis↑ROS↑; ΔΨm↓; GSH↓; GSH reductase↓; cytochrome C↑; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; pgH2AX↑; Apaf-1↑[31]
143B143B (xenograft in mice)5 mg/kgProliferation↓; apoptosis↑; S cell cycle arrest↑Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; Notch↓[32]
U2OS, 143B, MG-63143B (xenograft in mice)4 mg/kgApoptosis↑; S cell cycle arrest↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; Notch↓[33]
U2OS, MG-63, SaOS-2Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑xIAP↓; cIAP-1↓; Bcl-2↓; Bax↑; cleaved PARP↑; GSK-3β/NF-κB↓[34]
MNNG/HOS, U2OSViability↓; invasion and metastases↓FOXO1↑; EMT↓[35]
Autophagy↓ROS/JNK/p38↓[36]
Nasopharyngeal carcinomaHK-1S cell cycle arrest↑; apoptosis↑CDK2↓; Cyclin E↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[37]
Invasion and metastases↓ENKUR↑; p53↑; EMT↓[38]
MelanomaA375, A2058, G361Viability and proliferation↓LEF1↓; Wnt/β-catenin signalling pathway↓; c-Myc↓; cyclin D1↓; Axin-2↓[39]
A375Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑; PI3K/p-PI3K/AKT/p-AKT↓; ATM↑; Chk2↑; CDC25C↓; CDK1↓; cyclin B↓[40]
Uveal MelanomaOCM1OCM1(xenograft in mice)5 mg/kgViability and proliferation↓; G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[41]
CholangiocarcinomaQBC939, RBEQBC939(xenograft in mice)10 mg/kgProliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑; Notch↓[42]
Glioblastoma multiformeU87MG-EGFRU87MG-EGFR (xenograft in mice)1 mg/kg, 5mg/kgProliferation↓; apoptosis↑EGFR↓; ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[43]
GlioblastomaU87Proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; PI3K-AKT-4EBP1↓[42]
eOesophageal Squamous Cell CarcinomaEC-109, Kyse-150, Kyse-520Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; caspase-3↑; Puma↑; p73↑; B-CDC2↓; CDC2↓; p21↑; Wee1↑[44]
Hepatocellular carcinomaHepG2, SK-HEP-1Proliferation↓; G2/M cell cycle arrest↑TYMS↓; DDR↑[45]
Huh7Viability↓; G2/M cell cycle arrest↑; apoptosis↑;ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; p73↑[46]
DNA damage↑AUPKA↓; mTOR↓; p-mTOR↓; eIF4E↓; p-eIF4E↓;4E-BP1↑[47]
HepG2Apoptosis↑Fas↑; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓[48]
Apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; NF-κB↓[49]
Bel-7402G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[50]
HepG2, SMMC-7721Proliferation↓; apoptosis↑; S cell cycle arrest↑EPK↓; c-Myc↓[51]
Gastric cancerSGC-7901Autophagy↓; proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[52]
Acute myeloid leukaemiaHL60, Kasumi-1Proliferation↓c-Myc↓[53]
Non-small-cell lung cancerA549Proliferation↓; apoptosis↑; invasion and metastases↓Bax↑; Bcl-2↓; caspase-3↑; FOXO1↑; G9a↓[54]
A549, H1299, H460, SK-MES-1A549(xenograft in mice)1.5 mg/kg, 5 mg/kg, 10 mg/kgProliferation↓; apoptosis↑; G2/M cell cycle arrest↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; cleaved caspase-3↑; invasion and metastases ↓; AKT/mTOR↓; ROS↑[55]
Human multiple myelomaU266Apoptosis↑EPK/JNK/p38/MAPKs↑; ROS↑; cleaved caspase-3↑[56]
Prostate cancerLNCaP, DU145, PC3Apoptosis↑p53↑; caspase-9↑; caspase-3↑[57]
Breast cancerMCF-7G1 cell cycle arrest↑; apoptosis↑Bax/Bcl-2↑[58]
MDA-MB-231S cell cycle arrest↑[59]
Cervical carcinomaHeLaG2/M cell cycle arrest↑[60]
Cancer typeCell line (in vitro)Animal model (in vivo)DoseEffectMechanismRefs.
Colorectal cancerHCT116, RKO, SW480HCT116(xenograft in mice)0.5 mg/kg, 1 mg/kgViability and proliferation↓; apoptosis↑; invasion and metastases↓STAT3↓; Bax↑; Bcl-2↓; caspase-3↑; EMT ↓; E-Cadherin↑; Vimentin↓[23]
HCT116, HT29HCT116(xenograft in mice)10 mg/kgProliferation↓; G2/M cell cycle arrest↑; apoptosis↑ER↑; mTORC1↓; caspase-12↑[24]
SW480, EOMA (xenograft in mice)2 mg/kg, 4 mg/kgAngiogenesis↓; apoptosis↑mOTRC1↓; HIF-1α↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[25]
SW480, SW1116Apoptosis↑; G2/M cell cycle arrest↑p53↓; p21↓; ROS↑[26]
Colon cancerLoVo/ADR, HCT116/L and Cao-2/ADRDrug resistance↓p-gp↓[27]
HCT116, HT29Invasion and metastases↓Cortactin↓[28]
HCT116, HT30HCT116(xenograft in mice)1.5 mg/kgApoptosis↑HIF-1α↓[29]
Oral squamous cell carcinomaCAL-27Invasion and metastases↓; apoptosis↑STAT3↓; ANO1↓; ANO1 mRNA↓; caspase-3↑; cleaved PARP-1↑[30]
OsteosarcomaU2OS, 143B143B (xenograft in mice)2 mg/kg, 6 mg/kgViability↓; apoptosis↑ROS↑; ΔΨm↓; GSH↓; GSH reductase↓; cytochrome C↑; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; pgH2AX↑; Apaf-1↑[31]
143B143B (xenograft in mice)5 mg/kgProliferation↓; apoptosis↑; S cell cycle arrest↑Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; Notch↓[32]
U2OS, 143B, MG-63143B (xenograft in mice)4 mg/kgApoptosis↑; S cell cycle arrest↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; Notch↓[33]
U2OS, MG-63, SaOS-2Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑xIAP↓; cIAP-1↓; Bcl-2↓; Bax↑; cleaved PARP↑; GSK-3β/NF-κB↓[34]
MNNG/HOS, U2OSViability↓; invasion and metastases↓FOXO1↑; EMT↓[35]
Autophagy↓ROS/JNK/p38↓[36]
Nasopharyngeal carcinomaHK-1S cell cycle arrest↑; apoptosis↑CDK2↓; Cyclin E↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[37]
Invasion and metastases↓ENKUR↑; p53↑; EMT↓[38]
MelanomaA375, A2058, G361Viability and proliferation↓LEF1↓; Wnt/β-catenin signalling pathway↓; c-Myc↓; cyclin D1↓; Axin-2↓[39]
A375Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑; PI3K/p-PI3K/AKT/p-AKT↓; ATM↑; Chk2↑; CDC25C↓; CDK1↓; cyclin B↓[40]
Uveal MelanomaOCM1OCM1(xenograft in mice)5 mg/kgViability and proliferation↓; G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[41]
CholangiocarcinomaQBC939, RBEQBC939(xenograft in mice)10 mg/kgProliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑; Notch↓[42]
Glioblastoma multiformeU87MG-EGFRU87MG-EGFR (xenograft in mice)1 mg/kg, 5mg/kgProliferation↓; apoptosis↑EGFR↓; ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[43]
GlioblastomaU87Proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; PI3K-AKT-4EBP1↓[42]
eOesophageal Squamous Cell CarcinomaEC-109, Kyse-150, Kyse-520Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; caspase-3↑; Puma↑; p73↑; B-CDC2↓; CDC2↓; p21↑; Wee1↑[44]
Hepatocellular carcinomaHepG2, SK-HEP-1Proliferation↓; G2/M cell cycle arrest↑TYMS↓; DDR↑[45]
Huh7Viability↓; G2/M cell cycle arrest↑; apoptosis↑;ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; p73↑[46]
DNA damage↑AUPKA↓; mTOR↓; p-mTOR↓; eIF4E↓; p-eIF4E↓;4E-BP1↑[47]
HepG2Apoptosis↑Fas↑; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓[48]
Apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; NF-κB↓[49]
Bel-7402G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[50]
HepG2, SMMC-7721Proliferation↓; apoptosis↑; S cell cycle arrest↑EPK↓; c-Myc↓[51]
Gastric cancerSGC-7901Autophagy↓; proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[52]
Acute myeloid leukaemiaHL60, Kasumi-1Proliferation↓c-Myc↓[53]
Non-small-cell lung cancerA549Proliferation↓; apoptosis↑; invasion and metastases↓Bax↑; Bcl-2↓; caspase-3↑; FOXO1↑; G9a↓[54]
A549, H1299, H460, SK-MES-1A549(xenograft in mice)1.5 mg/kg, 5 mg/kg, 10 mg/kgProliferation↓; apoptosis↑; G2/M cell cycle arrest↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; cleaved caspase-3↑; invasion and metastases ↓; AKT/mTOR↓; ROS↑[55]
Human multiple myelomaU266Apoptosis↑EPK/JNK/p38/MAPKs↑; ROS↑; cleaved caspase-3↑[56]
Prostate cancerLNCaP, DU145, PC3Apoptosis↑p53↑; caspase-9↑; caspase-3↑[57]
Breast cancerMCF-7G1 cell cycle arrest↑; apoptosis↑Bax/Bcl-2↑[58]
MDA-MB-231S cell cycle arrest↑[59]
Cervical carcinomaHeLaG2/M cell cycle arrest↑[60]

↑Increase; ↓Decrease. JNK: c-Jun N-terminal kinase.

Table 1

Pharmacological effects and mechanisms of cinobufagin in various cancers

Cancer typeCell line (in vitro)Animal model (in vivo)DoseEffectMechanismRefs.
Colorectal cancerHCT116, RKO, SW480HCT116(xenograft in mice)0.5 mg/kg, 1 mg/kgViability and proliferation↓; apoptosis↑; invasion and metastases↓STAT3↓; Bax↑; Bcl-2↓; caspase-3↑; EMT ↓; E-Cadherin↑; Vimentin↓[23]
HCT116, HT29HCT116(xenograft in mice)10 mg/kgProliferation↓; G2/M cell cycle arrest↑; apoptosis↑ER↑; mTORC1↓; caspase-12↑[24]
SW480, EOMA (xenograft in mice)2 mg/kg, 4 mg/kgAngiogenesis↓; apoptosis↑mOTRC1↓; HIF-1α↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[25]
SW480, SW1116Apoptosis↑; G2/M cell cycle arrest↑p53↓; p21↓; ROS↑[26]
Colon cancerLoVo/ADR, HCT116/L and Cao-2/ADRDrug resistance↓p-gp↓[27]
HCT116, HT29Invasion and metastases↓Cortactin↓[28]
HCT116, HT30HCT116(xenograft in mice)1.5 mg/kgApoptosis↑HIF-1α↓[29]
Oral squamous cell carcinomaCAL-27Invasion and metastases↓; apoptosis↑STAT3↓; ANO1↓; ANO1 mRNA↓; caspase-3↑; cleaved PARP-1↑[30]
OsteosarcomaU2OS, 143B143B (xenograft in mice)2 mg/kg, 6 mg/kgViability↓; apoptosis↑ROS↑; ΔΨm↓; GSH↓; GSH reductase↓; cytochrome C↑; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; pgH2AX↑; Apaf-1↑[31]
143B143B (xenograft in mice)5 mg/kgProliferation↓; apoptosis↑; S cell cycle arrest↑Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; Notch↓[32]
U2OS, 143B, MG-63143B (xenograft in mice)4 mg/kgApoptosis↑; S cell cycle arrest↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; Notch↓[33]
U2OS, MG-63, SaOS-2Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑xIAP↓; cIAP-1↓; Bcl-2↓; Bax↑; cleaved PARP↑; GSK-3β/NF-κB↓[34]
MNNG/HOS, U2OSViability↓; invasion and metastases↓FOXO1↑; EMT↓[35]
Autophagy↓ROS/JNK/p38↓[36]
Nasopharyngeal carcinomaHK-1S cell cycle arrest↑; apoptosis↑CDK2↓; Cyclin E↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[37]
Invasion and metastases↓ENKUR↑; p53↑; EMT↓[38]
MelanomaA375, A2058, G361Viability and proliferation↓LEF1↓; Wnt/β-catenin signalling pathway↓; c-Myc↓; cyclin D1↓; Axin-2↓[39]
A375Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑; PI3K/p-PI3K/AKT/p-AKT↓; ATM↑; Chk2↑; CDC25C↓; CDK1↓; cyclin B↓[40]
Uveal MelanomaOCM1OCM1(xenograft in mice)5 mg/kgViability and proliferation↓; G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[41]
CholangiocarcinomaQBC939, RBEQBC939(xenograft in mice)10 mg/kgProliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑; Notch↓[42]
Glioblastoma multiformeU87MG-EGFRU87MG-EGFR (xenograft in mice)1 mg/kg, 5mg/kgProliferation↓; apoptosis↑EGFR↓; ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[43]
GlioblastomaU87Proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; PI3K-AKT-4EBP1↓[42]
eOesophageal Squamous Cell CarcinomaEC-109, Kyse-150, Kyse-520Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; caspase-3↑; Puma↑; p73↑; B-CDC2↓; CDC2↓; p21↑; Wee1↑[44]
Hepatocellular carcinomaHepG2, SK-HEP-1Proliferation↓; G2/M cell cycle arrest↑TYMS↓; DDR↑[45]
Huh7Viability↓; G2/M cell cycle arrest↑; apoptosis↑;ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; p73↑[46]
DNA damage↑AUPKA↓; mTOR↓; p-mTOR↓; eIF4E↓; p-eIF4E↓;4E-BP1↑[47]
HepG2Apoptosis↑Fas↑; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓[48]
Apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; NF-κB↓[49]
Bel-7402G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[50]
HepG2, SMMC-7721Proliferation↓; apoptosis↑; S cell cycle arrest↑EPK↓; c-Myc↓[51]
Gastric cancerSGC-7901Autophagy↓; proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[52]
Acute myeloid leukaemiaHL60, Kasumi-1Proliferation↓c-Myc↓[53]
Non-small-cell lung cancerA549Proliferation↓; apoptosis↑; invasion and metastases↓Bax↑; Bcl-2↓; caspase-3↑; FOXO1↑; G9a↓[54]
A549, H1299, H460, SK-MES-1A549(xenograft in mice)1.5 mg/kg, 5 mg/kg, 10 mg/kgProliferation↓; apoptosis↑; G2/M cell cycle arrest↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; cleaved caspase-3↑; invasion and metastases ↓; AKT/mTOR↓; ROS↑[55]
Human multiple myelomaU266Apoptosis↑EPK/JNK/p38/MAPKs↑; ROS↑; cleaved caspase-3↑[56]
Prostate cancerLNCaP, DU145, PC3Apoptosis↑p53↑; caspase-9↑; caspase-3↑[57]
Breast cancerMCF-7G1 cell cycle arrest↑; apoptosis↑Bax/Bcl-2↑[58]
MDA-MB-231S cell cycle arrest↑[59]
Cervical carcinomaHeLaG2/M cell cycle arrest↑[60]
Cancer typeCell line (in vitro)Animal model (in vivo)DoseEffectMechanismRefs.
Colorectal cancerHCT116, RKO, SW480HCT116(xenograft in mice)0.5 mg/kg, 1 mg/kgViability and proliferation↓; apoptosis↑; invasion and metastases↓STAT3↓; Bax↑; Bcl-2↓; caspase-3↑; EMT ↓; E-Cadherin↑; Vimentin↓[23]
HCT116, HT29HCT116(xenograft in mice)10 mg/kgProliferation↓; G2/M cell cycle arrest↑; apoptosis↑ER↑; mTORC1↓; caspase-12↑[24]
SW480, EOMA (xenograft in mice)2 mg/kg, 4 mg/kgAngiogenesis↓; apoptosis↑mOTRC1↓; HIF-1α↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[25]
SW480, SW1116Apoptosis↑; G2/M cell cycle arrest↑p53↓; p21↓; ROS↑[26]
Colon cancerLoVo/ADR, HCT116/L and Cao-2/ADRDrug resistance↓p-gp↓[27]
HCT116, HT29Invasion and metastases↓Cortactin↓[28]
HCT116, HT30HCT116(xenograft in mice)1.5 mg/kgApoptosis↑HIF-1α↓[29]
Oral squamous cell carcinomaCAL-27Invasion and metastases↓; apoptosis↑STAT3↓; ANO1↓; ANO1 mRNA↓; caspase-3↑; cleaved PARP-1↑[30]
OsteosarcomaU2OS, 143B143B (xenograft in mice)2 mg/kg, 6 mg/kgViability↓; apoptosis↑ROS↑; ΔΨm↓; GSH↓; GSH reductase↓; cytochrome C↑; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; pgH2AX↑; Apaf-1↑[31]
143B143B (xenograft in mice)5 mg/kgProliferation↓; apoptosis↑; S cell cycle arrest↑Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; Notch↓[32]
U2OS, 143B, MG-63143B (xenograft in mice)4 mg/kgApoptosis↑; S cell cycle arrest↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; Notch↓[33]
U2OS, MG-63, SaOS-2Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑xIAP↓; cIAP-1↓; Bcl-2↓; Bax↑; cleaved PARP↑; GSK-3β/NF-κB↓[34]
MNNG/HOS, U2OSViability↓; invasion and metastases↓FOXO1↑; EMT↓[35]
Autophagy↓ROS/JNK/p38↓[36]
Nasopharyngeal carcinomaHK-1S cell cycle arrest↑; apoptosis↑CDK2↓; Cyclin E↓; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑[37]
Invasion and metastases↓ENKUR↑; p53↑; EMT↓[38]
MelanomaA375, A2058, G361Viability and proliferation↓LEF1↓; Wnt/β-catenin signalling pathway↓; c-Myc↓; cyclin D1↓; Axin-2↓[39]
A375Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; cytochrome C↑; caspase-9↑; caspase-3↑; cleaved PARP-1↑; Apaf-1↑; PI3K/p-PI3K/AKT/p-AKT↓; ATM↑; Chk2↑; CDC25C↓; CDK1↓; cyclin B↓[40]
Uveal MelanomaOCM1OCM1(xenograft in mice)5 mg/kgViability and proliferation↓; G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[41]
CholangiocarcinomaQBC939, RBEQBC939(xenograft in mice)10 mg/kgProliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑; Notch↓[42]
Glioblastoma multiformeU87MG-EGFRU87MG-EGFR (xenograft in mice)1 mg/kg, 5mg/kgProliferation↓; apoptosis↑EGFR↓; ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[43]
GlioblastomaU87Proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; caspase-9↑; caspase-3↑; cleaved PARP↑; PI3K-AKT-4EBP1↓[42]
eOesophageal Squamous Cell CarcinomaEC-109, Kyse-150, Kyse-520Proliferation↓; G2/M cell cycle arrest↑; apoptosis↑Bax↑; Bcl-2↓; caspase-3↑; Puma↑; p73↑; B-CDC2↓; CDC2↓; p21↑; Wee1↑[44]
Hepatocellular carcinomaHepG2, SK-HEP-1Proliferation↓; G2/M cell cycle arrest↑TYMS↓; DDR↑[45]
Huh7Viability↓; G2/M cell cycle arrest↑; apoptosis↑;ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; p73↑[46]
DNA damage↑AUPKA↓; mTOR↓; p-mTOR↓; eIF4E↓; p-eIF4E↓;4E-BP1↑[47]
HepG2Apoptosis↑Fas↑; Bax↑; Bcl-2↓; ROS↑; ΔΨm↓[48]
Apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓; NF-κB↓[49]
Bel-7402G2/M cell cycle arrest↑; apoptosis↑ROS↑; ΔΨm↓; Bax↑; Bcl-2↓[50]
HepG2, SMMC-7721Proliferation↓; apoptosis↑; S cell cycle arrest↑EPK↓; c-Myc↓[51]
Gastric cancerSGC-7901Autophagy↓; proliferation↓; apoptosis↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; caspase-9↑; caspase-3↑; cleaved PARP↑[52]
Acute myeloid leukaemiaHL60, Kasumi-1Proliferation↓c-Myc↓[53]
Non-small-cell lung cancerA549Proliferation↓; apoptosis↑; invasion and metastases↓Bax↑; Bcl-2↓; caspase-3↑; FOXO1↑; G9a↓[54]
A549, H1299, H460, SK-MES-1A549(xenograft in mice)1.5 mg/kg, 5 mg/kg, 10 mg/kgProliferation↓; apoptosis↑; G2/M cell cycle arrest↑ROS↑; ΔΨm↓; Bad, Bax↑; Bcl-2, Bcl-xl↓; cleaved caspase-3↑; invasion and metastases ↓; AKT/mTOR↓; ROS↑[55]
Human multiple myelomaU266Apoptosis↑EPK/JNK/p38/MAPKs↑; ROS↑; cleaved caspase-3↑[56]
Prostate cancerLNCaP, DU145, PC3Apoptosis↑p53↑; caspase-9↑; caspase-3↑[57]
Breast cancerMCF-7G1 cell cycle arrest↑; apoptosis↑Bax/Bcl-2↑[58]
MDA-MB-231S cell cycle arrest↑[59]
Cervical carcinomaHeLaG2/M cell cycle arrest↑[60]

↑Increase; ↓Decrease. JNK: c-Jun N-terminal kinase.

The main mechanisms of cinobufagin-induced apoptosis of cancer cells. ↑Increase; ↓Decrease. Source: By Figdraw.
Figure 3

The main mechanisms of cinobufagin-induced apoptosis of cancer cells. ↑Increase; ↓Decrease. Source: By Figdraw.

Induction of tumour cell apoptosis

Apoptosis is a process of programmed cell death characterized by chromatin condensation, DNA fragmentation and the shedding of small fragments in cells,[61, 62] which is beneficial for maintaining the stability of the body’s internal environment. However, apoptosis does not occur naturally due to mutation, hence inducing apoptosis in tumour cells becomes one of the important strategies for the treatment of many cancers.

Cinobufagin can induce apoptosis in tumour cells by activating caspases through the death receptor pathway and the mitochondrial apoptosis pathway. The one of cell surface death receptor (Fas)-mediated extrinsic pathway plays a key role in the apoptosis of hepatocellular carcinoma HepG2 cells[48] and prostate cancer DU145 and PC3 cells.[57] Cinobufagin promotes the upregulation of Fas, which then binds to specific death ligands to activate the caspase cascade reaction to induce apoptosis. In osteosarcoma U2OS and 143B cells,[63, 31] uveal melanoma OCM1 cells,[41] melanoma A375 cells,[40] prostate cancer LNCaP, DU145 and PC3 cells,[64] hepatocellular carcinoma HepG2 cells[65] and glioblastoma U87 cells,[66] cinobufagin exerts pro-apoptotic effect through activation of the mitochondrial pathway. The mitochondria-dependent endogenous apoptotic pathway is dependent on reactive oxygen species (ROS) generation and loss of ΔΨm, resulting in the release of cytochrome C from mitochondria into the cytoplasm, where it binds to Apaf-1 and further activates caspase-9, followed by downstream caspase-3 activation by cleaved caspase-9, resulting in the cleavage of multiple key cellular substrates, including PARP, which in turn induces apoptosis.

The ratio of Bcl-2-associated X (Bax) to Bcl-2 is critical for maintaining drug-induced apoptosis in the mitochondria-mediated apoptosis pathway. In one study mice were intraperitoneally injected with cinobufagin at a dose of 5 mg/kg d−1. After 30 consecutive days, the mRNA expressions of Bcl-2-asociated death promoter (Bad), Bax, caspase-3 and PARP increased, while B-cell lymphoma-2 (Bcl-2) and B-cell lymphoma-xl (Bcl-xl) decreased in uveal melanoma tissues, indicating that cinobufagin can promote apoptosis of cancer cells by regulating Bax/Bcl-2 ratio.[41] Notch signalling pathway also plays an essential role in the apoptosis of cancer cells.[67] For example, cinobufagin promotes apoptosis in cholangiocarcinoma QBC939 and RBE cells by inhibiting the Notch pathway.[42] Similar effects were observed in osteosarcoma U2OS, 143B and MG-63 cells.[33] In addition, p53 is also involved in the apoptosis of HepG2 cells.[68] p73 is a member of the p53 family, which acts as an equivalent to p53 and may be a potential therapeutic target for cancer cells lacking functional p53.[69] Cinobufagin activates apoptotic signalling pathway by upregulating p73 expression in oesophageal squamous carcinoma EC-109, Kyse-150 and Kyse-520 cells[44] and hepatocellular carcinoma Huh-7 cells.[46] In addition to p73, cinobufagin can also act by inhibiting phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT),[66] glycogen synthase kinase-3β (GSK-3β)/NF-κB[49, 34] and MAPKs signalling pathways.[56] Besides, cinobufagin can activate the tumour suppressor gene forkhead box O1 (FOXO1)[35, 54]and inhibit c-Myc-related genes[53] to promote tumour cells apoptosis. By inhibiting the β-catenin signalling pathway, cinobufagin degrades PML-RARA cells and induces apoptosis of NB4 and NB4-R1 cells in a caspase-dependent manner, suggesting a new therapeutic strategy for the treatment of acute promyelocytic leukemia which is a specific malignant haematologic neoplasm, but its exact mechanism needs to be further explored.[70]

Inhibition of cancer cell proliferation

One of the most important characteristics of cancer cells is that they can proliferate indefinitely and become ‘immortal cells’. Therefore, induction of cancer cell inactivation or inhibition of their proliferation is one of the ways of cancer treatment.

In different cancers, cinobufagin inhibits the activity of cancer cells and its antiproliferation targets are also different. Signal transducer and activator of transcription 3 (STAT3) is associated with promoting tumour cell proliferation and therefore has been used as a therapeutic target to inhibit tumour cell proliferation in many studies.[71] Cinobufagin inhibits the growth of colorectal cancer cells HCT116, RKO and SW480 by inhibiting STAT3 signalling pathway.[23] Epidermal growth factor receptor (EGFR) amplification and mutations are present in different types of cancers. In a tumour-bearing nude mouse model inoculated subcutaneously with U87-EGFR cells, intraperitoneal injections of 1 or 5 mg/kg d−1of cinobufagin significantly slowed tumour growth, which is related to inactivation of the EGFR and STAT3 signalling pathways. Cinobufagin also exhibits specific antiproliferative effects in EGFR-expressing specific cancer cells, including glioblastoma, lung cancer, colorectal cancer and hepatocellular carcinoma.[43]

Besides, Anoctamin1 (ANO1), a calcium-activated chloride channel is also a potential therapeutic target.[72] Cinobufagin can downregulate the high amplification and expression of ANO1 in the OSCC cell line CAL-27, thereby inhibiting tumour cell viability.[30] mTORC1 inactivation may also be important for the antiproliferative activity of cinobufagin in CRC cells.[51] It is well known that activation of RAS-RAF-MEK-ERK, PI3K-AKT and canonical Wnt/β-catenin signalling pathways can promote cancer cell growth. Cinobufagin inhibits hepatocellular carcinoma cell proliferation by attenuating extracellular regulated kinase (ERK) phosphorylation and downregulating c-Myc expression.[24] As a potent inhibitor of Wnt/β-catenin signalling, cinobufagin reduces lymphoid enhancer-binding factor 1 (LEF1) expression, thereby inhibiting melanoma growth.[39]

Suppression of invasion and metastasis

Abnormal differentiation, invasion and metastasis are typical biological characteristics of tumour cells.[73] In general, metastasis of cancer cells makes treatment more difficult and is a key factor in clinical death of oncology patients. Therefore, it is urgent to find ways to treat cancer cells that can inhibit metastasis. Epithelial mesenchymal transition (EMT) is the process by which epithelial cells with malignant phenotype are transformed into mesenchymal stem cells to promote metastasis.[74] Cinobufagin not only inhibits EMT to suppress the growth, invasion and metastasis of colorectal cancer cells but also affects the expression of EMT protein levels. It induces Enkurin (ENKUR) to inhibit β-linked protein/c-Jun/MYH9 signalling, thereby reducing UBE3A-mediated p53 ubiquitin degradation. As a result, inactivation of EMT signalling leads to metastasis inhibition in nasopharyngeal carcinoma.[38] Besides, cortactin is also an important factor involved in cancer cell progression and invasion.[75] Cinobufagin inhibits the expression of cortactin in colon cancer to suppress colon cancer cell invasion and metastasis in vitro and in vivo.[28] Na/K-ATPase is an integral heterodimeric plasma membrane protein that maintains cell membrane potential and osmotic homeostasis between intracellular and extracellular pathways.[76] Due to its regulation by several hormones, studies have shown its involvement in abnormal protein expression and in the progression of several cancers.[77] Bufadienolide is an inhibitor of Na/K-ATPase, is considered a promising source of Na/K-ATPase inhibitory anticancer prototype drugs.[78] Cinobufagin acts as one of the important compounds of bufadienolide, the other being bufalin. Recent studies suggest that they may have other targets of action. Porcine aminopeptidase N (pAPN) and porcine aminopeptidase A (pAPA), members of the M1 and M17 families, are upregulated on the surface expression of many cells and are positively correlated with cell invasion ability. The results of preliminary screening experiments suggest that cinobufagin non-competitively inhibits pAPN and competitively inhibits pAPA activity, acting as a dual inhibitor. At the cellular level, it was shown that cinobufagin promotes cell death in melanoma MeWo cells and increases DNA degradation of the cells by 35%. Although the link between cell death induction and pAPN is limited, it merits further exploration and provides new ideas for the treatment of cancers associated with pAPN upregulation.[55]

Cell cycle arrest

The cell cycle is the basis of cell proliferation and blocking the cell cycle also facilitates the antitumour efficacy of drugs. Cinobufagin significantly decreases the expression of CDK2 (cyclin-dependent kinases 2) and cyclin E, which are the regulatory proteins associated with S-phase in HK-1 cells of nasopharyngeal carcinoma, suggesting that cinobufagin blocked the cell cycle in S-phase.[37]

In melanoma A375 cells, cinobufagin inhibits the expression of cyclin-dependent kinase 1 (CDK1) and cyclin B, and upregulates the levels of ATM serine/threonine kinase, Chk2 (checkpoint kinase 2), p-CDC25C (phosphorylated cell division cycle 25C), thereby inducing G2/M cell cycle arrest.[40] In addition, Cinobufagin also induces a variety of other cancer cells G2/M-phase arrest, such as HeLa cervical cancer cells, SK-HEP-1 and HepG2 liver cancer cells and U2OS, MG-63 and SaOS-2 colon cancer cells.[34, 45, 60] However, in MCF-7 breast cancer cells, cinobufagin induces cell cycle arrest in G0/G1 phase.[58] Cinobufagin-induced cell cycle arrest is unrestricted to specific stages and varies in a variety of non-small cell lung cancer cell lines, suggesting that cinobufagin has different targets in different cells.[79]

Depression of angiogenesis

New regeneration of blood vessels is often a sign of tumour cell invasion and metastasis.[80] The generation of tumour blood vessels provides the nutrients needed for tumour growth. When the mass is small, tumour cells can obtain nutrients directly from the surrounding tissue. However, when the mass is larger than 2 mm, the growth of tumour metastasis depends on its own formation of neovascularization. Therefore, inhibition of tumour angiogenesis is one of the effective strategies for current tumour treatment. In a mouse model transplanted with colorectal cancer (CRC) SW480 cells in situ, Li et al. found that cinobufagin inhibited CRC angiogenesis by downregulating activation of the AKT/mTORC1/HIF-1α pathway and triggering MOMP-mediated apoptosis, thus achieving depression of tumour growth.[25]

Reduction of autophagy

Cellular autophagy can exert bidirectionally modulatory effects on tumour growth. On the one hand, autophagy induces the degradation of anti-apoptotic factors when tumour cells are under the environment of chemotherapy or lack of oxygen and nutrients, promoting cancer progression. But on the other hand, autophagy can stabilize the cell state, stop normal cells on the way to cancer in time, and inhibit tumour neovascularization, reducing tumour growth. Targeting autophagic process will undoubtedly become a new hot spot in antitumour therapy. Cinobufagin induces cellular autophagy by accumulation of GFP- light chain 3 (LC3) fluorescent particles and upregulation of LC3-II levels and the inhibition of autophagy reduces apoptosis of cells. Cinobufagin triggers apoptosis and autophagic cell death in human osteosarcoma U2OS cells through activation of the ROS/JNK/p38 axis.[36] It can also decrease autophagy to increase apoptosis of gastric cancer cell line SGC-7901, thus exerting the anticancer effects.[52]

Reversal of drug resistance

Cross-resistance of tumour cells to multiple antitumour drugs with different chemical structures and mechanisms of action is a common clinical phenomenon. It is one of the current research areas to investigate the mechanism of tumour multidrug resistance and discover new effective chemotherapy sensitizers. It is currently believed that the mechanism of multidrug resistance in tumour cells is closely related to the overexpression of ABC transporters, and P-glycoprotein (P-gp) is the most studied ABC transporter.[81] Cinobufagin can reverse P-gp-mediated multidrug resistance by non-competitively inhibiting P-gp ATPase activity and P-gp efflux function without affecting its expression, indicating that it is a safe and effective P-gp reversal agent.[27]

Others

Sub-lethal amounts of cinobufagin elicit ROS overload, leading to intense DNA damage and oxidative stress, which in turn induces apoptosis.[26] Cinobufagin causes DNA damage of oncogenes by reducing the expression of AURKA, mTOR, p-mTOR, p-ERK, eukaryotic translation initiation factor 4E and p-eIF4E.[47] In addition, cinobufagin has been shown to inhibit growth and trigger DNA damage in human hepatocellular carcinoma cells through proteasome-dependent degradation of thymidylate synthase.[82] Pain is one of the most common complications of malignant tumours and alleviating cancer pain is beneficial to improve patients’ quality of life. In a paw cancer pain model of mice, cinobufagin significantly increases the pain threshold to alleviate cancer pain mainly through upregulating the expression levels of β-END and μ-OR in hind paw tumours and adjacent tissues.[83] The analgesic effect of cinobufagin via β-END in bone cancer has also been confirmed.[84] In addition, M4mAChR is also involved in regulating the pain relief of cinobufagin in bone cancer pain rat model.[85] Cinobufagin-induced local analgesia may be associated with increased POMC/β-END/μ-OR pathway activity released by invaded CD3/4/8 lymphocytes in cancer tissues.[86]

As described above, cinobufagin has strong anticancer effects, but it also has other bioactive effects that have potential for further development. Acute lung injury (ALI) due to bacterial infection is still a serious clinical condition with high morbidity and mortality. Studies have shown that in a lipopolysaccharide-induced ALI model, cinobufagin treatment can effectively reverse lung tissue damage such as disrupted alveolar structure, thickened septa, infiltrative oedema and inflammatory cells in the alveoli and interstitium. This reversal was found to be caused by the activation of cellular autophagy in vivo due to reduced levels of inflammatory cytokines such as IL-1β, IL-6 and TNF-α. In addition to this, significant activation of the p53/mTOR pathway, a classical autophagy-associated pathway in vivo, was also observed for cinobufagin.[87] However, in another study, exposure of dendritic cells to cinobufagin led to the release of IL-1β, recruitment of inflammatory cells to the site of infection and induction of production of the antimicrobial peptides hBD-2 and hBD-3, which have been shown to help fight bacterial infections.[88] Transforming growth factor beta (TGF-β) is a major promoter of myofibroblast activation and plays a very important role in EMT. Emerging evidence suggests that cinobufagin inhibits the TGF-β1/Smad3 signalling pathway in a concentration-dependent manner, primarily inhibiting fibroblast differentiation without significant toxic effects. More importantly, in vivo experiments have found that cinobufagin ameliorates bleomycin-induced lung fibrosis in mice and reduces the number of inflammatory cells and inflammatory cytokines.[89] Cinobufagin can also reduce the expression of c-Myc gene to inhibit the growth of leukaemia cells and thus fight against acute myeloid leukaemia.[90] There is also a study describing the efficacy of cinobufagin as an adjuvant for inactivated Salmonella typhimurium vaccine, stimulating IFN-γ production to enhance Th1 immune responses in vivo.[91] Then for hepatitis B virus, cinobufagin also exhibited a slight inhibitory effect.[92]

Toxicity

Cinobufagin belongs to bufadienolide and is one of the active ingredients of Venenum Bufonis. Although cinobufagin has good antitumour efficacy, its toxicity and side effects should also address. High dose cinobufagin is toxic to the heart as it triggers cardiac arrhythmias and to the central nervous system by triggering seizures, muscle spasms or fatal paralysis.[93, 94] Cinobufagin induces acute local ischaemia to cause heart failure, with half maximal inhibitory concentration (IC50)=10−8 M.[95] It also induces ROS-related cytotoxicity in human astrocyte cells and promotes the development of brain-related diseases. Cinobufagin at the dose of around 20 nM and above may be toxic to GHA and HCN-2 cells, although it does not induce Ca2+ signalling or ROS responses in HCN-2 cells. It may interfere with Ca2+ signalling or ROS downstream sensitivity.[96] In an acute toxicity test, cinobufagin caused the death of Kunming mice (20 ± 2g) after 10–60 min of administration, with an LD50 of 4.78 mg/kg, and the poisoning symptoms include closed eyes, curled up, respiratory depression and reduced activity.[97] However, in the experiment conducted by our research group, the C57BL/6 mice showed no obvious abnormal symptoms after daily intragastric administration of 20 mg/kg, suggesting that there exists the heterogeneity of the effect of cinobufagin and further exploration is needed.

Pharmacokinetics

Pharmacokinetics is one of the most important reasons affecting the development and marketing of anticancer drugs. Because of the toxic effects of cinobufagin, it is necessary to study its pharmacokinetics and metabolism. The pharmacokinetics of cinobufagin was first described in 1987 by Toma et al.[98] After intravenous administration of 0.25 mg/kg of cinobufagin to rats, the pharmacokinetic parameters were obtained by HPLC quantitative analysis: elimination half-life (t1/2α) = 0.05 h, t1/2β = 0.3 h, maximum concentration (Cmax)=148 ng/ml. It was demonstrated by NMR and mass spectrometry that cinobufagin was metabolized to desacetylcinobufagin and 3-epidesacetylcinobufagin in rats after intravenous injection. However, only 3-epidesacetylcinobufagin was detected in the serum of rats after oral administration of cinobufagin, indicating that cinobufagin is well absorbed in the gastrointestinal tract and prone to first-pass elimination. In addition, cinobufagin deacetylation was shown to occur in plasma and the liver.

With the advancement of modern science and technology, many new techniques have emerged, such as liquid chromatography-MS,[99] UPLC-ESI-MS/MS,[100] SPE-HPLC and[101] high-speed counter-current chromatography.[102] Thus, a more realistic and effective analysis of the pharmacokinetics of Chinese medicines can be performed. Liang et al. for the first time, established a sensitive, rapid and specific solid phase extraction high-performance liquid phase method (SPE-HPLC) to determine the pharmacokinetics of cinobufagin in rats after oral administration and obtained the following parameters: Cmax = 0.77 ± 0.12 μg/ml, Tmax = 20 ± 12 min, t1/2 = 138 ± 30 min, apparent clearance (CL/F) = 0.093 ± 0.017 l/ (min kg), Vz/F = 18.045 ± 2.908 l/kg. More detailed data are presented in Table 2.

Table 2

The pharmacokinetic parameters of cinobufagin

ModelDoseRoute of administrationQuantitationParameterRefs.
Wistar rats0.25 mg/ kgIntravenousHPLCt1/2α = 3 min[98]
t1/2β = 18 min
Cmax = 148 ng/ml
Sprague–Dawley rats120 mg/kgOralSPE-HPLCCmax = 0.77 ± 0.12 ug/ml[101]
Tmax = 20 ± 12 min
t1/2 = 138 ± 30 min
CL/F = 0.093 ± 0.017 l (min kg)
Vz/F = 18.045 ± 2.908 l/kg
AUC0~t = 92.160 ± 13.767 min ug/ml
AUC0~∞ = 98.688 ± 17.700 min ug/ml
MRT = 227.863 ± 66.785 min
Wistar rats100 mg/kgIntragastricLC-MS/MSTmax = 0.33 ± 0.13 h[103]
Cmax = 41.0 ± 20.6 ng/ml
Ke = 0.20 ± 0.04 h-1
t1/2 = 3.54 ± 0.67 h
AUC0~t = 66.3 ± 7.3ng h/ml
AUC0~∞ = 78.2 ± 9.3ng h/ml
AUMC0~t = 361.0 ± 76.8 ng h2/ml
MRT = 4.59 ± 0.60 h
ModelDoseRoute of administrationQuantitationParameterRefs.
Wistar rats0.25 mg/ kgIntravenousHPLCt1/2α = 3 min[98]
t1/2β = 18 min
Cmax = 148 ng/ml
Sprague–Dawley rats120 mg/kgOralSPE-HPLCCmax = 0.77 ± 0.12 ug/ml[101]
Tmax = 20 ± 12 min
t1/2 = 138 ± 30 min
CL/F = 0.093 ± 0.017 l (min kg)
Vz/F = 18.045 ± 2.908 l/kg
AUC0~t = 92.160 ± 13.767 min ug/ml
AUC0~∞ = 98.688 ± 17.700 min ug/ml
MRT = 227.863 ± 66.785 min
Wistar rats100 mg/kgIntragastricLC-MS/MSTmax = 0.33 ± 0.13 h[103]
Cmax = 41.0 ± 20.6 ng/ml
Ke = 0.20 ± 0.04 h-1
t1/2 = 3.54 ± 0.67 h
AUC0~t = 66.3 ± 7.3ng h/ml
AUC0~∞ = 78.2 ± 9.3ng h/ml
AUMC0~t = 361.0 ± 76.8 ng h2/ml
MRT = 4.59 ± 0.60 h

AUC: area under the concentration-time curve; AUMC: area under the first moment curve; Ke: elimination rate constant; LC-MS/MS: liquid chromatography-tandem mass spectrometry; MRT: mean residence time.

Table 2

The pharmacokinetic parameters of cinobufagin

ModelDoseRoute of administrationQuantitationParameterRefs.
Wistar rats0.25 mg/ kgIntravenousHPLCt1/2α = 3 min[98]
t1/2β = 18 min
Cmax = 148 ng/ml
Sprague–Dawley rats120 mg/kgOralSPE-HPLCCmax = 0.77 ± 0.12 ug/ml[101]
Tmax = 20 ± 12 min
t1/2 = 138 ± 30 min
CL/F = 0.093 ± 0.017 l (min kg)
Vz/F = 18.045 ± 2.908 l/kg
AUC0~t = 92.160 ± 13.767 min ug/ml
AUC0~∞ = 98.688 ± 17.700 min ug/ml
MRT = 227.863 ± 66.785 min
Wistar rats100 mg/kgIntragastricLC-MS/MSTmax = 0.33 ± 0.13 h[103]
Cmax = 41.0 ± 20.6 ng/ml
Ke = 0.20 ± 0.04 h-1
t1/2 = 3.54 ± 0.67 h
AUC0~t = 66.3 ± 7.3ng h/ml
AUC0~∞ = 78.2 ± 9.3ng h/ml
AUMC0~t = 361.0 ± 76.8 ng h2/ml
MRT = 4.59 ± 0.60 h
ModelDoseRoute of administrationQuantitationParameterRefs.
Wistar rats0.25 mg/ kgIntravenousHPLCt1/2α = 3 min[98]
t1/2β = 18 min
Cmax = 148 ng/ml
Sprague–Dawley rats120 mg/kgOralSPE-HPLCCmax = 0.77 ± 0.12 ug/ml[101]
Tmax = 20 ± 12 min
t1/2 = 138 ± 30 min
CL/F = 0.093 ± 0.017 l (min kg)
Vz/F = 18.045 ± 2.908 l/kg
AUC0~t = 92.160 ± 13.767 min ug/ml
AUC0~∞ = 98.688 ± 17.700 min ug/ml
MRT = 227.863 ± 66.785 min
Wistar rats100 mg/kgIntragastricLC-MS/MSTmax = 0.33 ± 0.13 h[103]
Cmax = 41.0 ± 20.6 ng/ml
Ke = 0.20 ± 0.04 h-1
t1/2 = 3.54 ± 0.67 h
AUC0~t = 66.3 ± 7.3ng h/ml
AUC0~∞ = 78.2 ± 9.3ng h/ml
AUMC0~t = 361.0 ± 76.8 ng h2/ml
MRT = 4.59 ± 0.60 h

AUC: area under the concentration-time curve; AUMC: area under the first moment curve; Ke: elimination rate constant; LC-MS/MS: liquid chromatography-tandem mass spectrometry; MRT: mean residence time.

In 2010, Ning et al. used 2D-NMR as a basic method to isolate and identify nine metabolites of cinobufagin in rat bile and further demonstrated that hydroxylation is the main reaction involved in rat bile metabolism.[104] After oral administration of cinobufagin to rats, 11 metabolites were obtained by liquid–liquid extraction, open column chromatography, medium pressure liquid chromatography and semi-preparative high-performance liquid chromatography. The main metabolic pathways of cinobufagin are hydroxylation, deacetylation and isomerization.[105] Its metabolic pathways and metabolite structures are presented in Figure 4 and Table 3.

Table 3

Metabolic products of cinobufagin

C1C2C33-epiC4C5C7C11C12C13C16
CinobufaginHHOHHHHHHCH3OAc
DesacetylcinobufaginHHOHHHHHHCH3OH
3-oxo-CinobufaginHHHHHHHCH3OAc
3-oxo-desacetylcinobufaginHHC=OHHHHHCH3OH
3-oxo-12α hydroxyl-cinobufaginHHC=OHHHHαOHCH3OAc
3-EpidesacetylcinobufaginHHOHHHHHHCH3OH
3-epi-12β-hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
5β-Hydroxyl-cinobufaginHHOHHβOHHαOHHCH3OAc
5β-Hydroxyl desacetylcinobufaginHHOHHβOHHHHCH3OH
5β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHβOHHHHCH3OH
7β-Hydroxyl-cinobufaginHHOHHHβOHHHCH3OAc
11α-Hydroxyl-cinobufaginHHOHHHHHβOHCH3OAc
12α-Hydroxyl-cinobufaginHHOHHHHHHαOHOAc
12β-Hydroxyl-cinobufaginHHOHHHHHHCH3OAc
12β-Hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
12β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHHHHβOHCH3OH
1α-Hydroxyl-3-epi-desacetylcinobufaginαOHHOHHHHHHCH3OH
1β-Hydroxyl-3-epi-desacetylcinobufaginβOHHOHHHHHHCH3OH
1α,5α-Dihydroxyl-3-epi-desacetylcinobufaginαOHHOHHαOHHHHCH3OH
1β,12β-Dihydroxyl cinobufaginβOHHOHHHHHβOHCH3OAc
5β,12β-Dihydroxyl cinobufaginHHOHHβOHHHβOHCH3OAc
4β,11α-Dihydroxyl cinobufaginHHOHβOHHHαOHHCH3OAc
4β,12α-Dihydroxyl cinobufaginHHOHβOHHHHαOHCH3OAc
1β,12β-Dihydroxyl desacetylcinobufaginβOHHOHHHHHβOHCH3OH
2α,5β-Dihydroxyl-3-epi-desacetylcinobufaginHαOHOHHβOHHHHCH3OH
C1C2C33-epiC4C5C7C11C12C13C16
CinobufaginHHOHHHHHHCH3OAc
DesacetylcinobufaginHHOHHHHHHCH3OH
3-oxo-CinobufaginHHHHHHHCH3OAc
3-oxo-desacetylcinobufaginHHC=OHHHHHCH3OH
3-oxo-12α hydroxyl-cinobufaginHHC=OHHHHαOHCH3OAc
3-EpidesacetylcinobufaginHHOHHHHHHCH3OH
3-epi-12β-hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
5β-Hydroxyl-cinobufaginHHOHHβOHHαOHHCH3OAc
5β-Hydroxyl desacetylcinobufaginHHOHHβOHHHHCH3OH
5β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHβOHHHHCH3OH
7β-Hydroxyl-cinobufaginHHOHHHβOHHHCH3OAc
11α-Hydroxyl-cinobufaginHHOHHHHHβOHCH3OAc
12α-Hydroxyl-cinobufaginHHOHHHHHHαOHOAc
12β-Hydroxyl-cinobufaginHHOHHHHHHCH3OAc
12β-Hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
12β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHHHHβOHCH3OH
1α-Hydroxyl-3-epi-desacetylcinobufaginαOHHOHHHHHHCH3OH
1β-Hydroxyl-3-epi-desacetylcinobufaginβOHHOHHHHHHCH3OH
1α,5α-Dihydroxyl-3-epi-desacetylcinobufaginαOHHOHHαOHHHHCH3OH
1β,12β-Dihydroxyl cinobufaginβOHHOHHHHHβOHCH3OAc
5β,12β-Dihydroxyl cinobufaginHHOHHβOHHHβOHCH3OAc
4β,11α-Dihydroxyl cinobufaginHHOHβOHHHαOHHCH3OAc
4β,12α-Dihydroxyl cinobufaginHHOHβOHHHHαOHCH3OAc
1β,12β-Dihydroxyl desacetylcinobufaginβOHHOHHHHHβOHCH3OH
2α,5β-Dihydroxyl-3-epi-desacetylcinobufaginHαOHOHHβOHHHHCH3OH
Table 3

Metabolic products of cinobufagin

C1C2C33-epiC4C5C7C11C12C13C16
CinobufaginHHOHHHHHHCH3OAc
DesacetylcinobufaginHHOHHHHHHCH3OH
3-oxo-CinobufaginHHHHHHHCH3OAc
3-oxo-desacetylcinobufaginHHC=OHHHHHCH3OH
3-oxo-12α hydroxyl-cinobufaginHHC=OHHHHαOHCH3OAc
3-EpidesacetylcinobufaginHHOHHHHHHCH3OH
3-epi-12β-hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
5β-Hydroxyl-cinobufaginHHOHHβOHHαOHHCH3OAc
5β-Hydroxyl desacetylcinobufaginHHOHHβOHHHHCH3OH
5β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHβOHHHHCH3OH
7β-Hydroxyl-cinobufaginHHOHHHβOHHHCH3OAc
11α-Hydroxyl-cinobufaginHHOHHHHHβOHCH3OAc
12α-Hydroxyl-cinobufaginHHOHHHHHHαOHOAc
12β-Hydroxyl-cinobufaginHHOHHHHHHCH3OAc
12β-Hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
12β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHHHHβOHCH3OH
1α-Hydroxyl-3-epi-desacetylcinobufaginαOHHOHHHHHHCH3OH
1β-Hydroxyl-3-epi-desacetylcinobufaginβOHHOHHHHHHCH3OH
1α,5α-Dihydroxyl-3-epi-desacetylcinobufaginαOHHOHHαOHHHHCH3OH
1β,12β-Dihydroxyl cinobufaginβOHHOHHHHHβOHCH3OAc
5β,12β-Dihydroxyl cinobufaginHHOHHβOHHHβOHCH3OAc
4β,11α-Dihydroxyl cinobufaginHHOHβOHHHαOHHCH3OAc
4β,12α-Dihydroxyl cinobufaginHHOHβOHHHHαOHCH3OAc
1β,12β-Dihydroxyl desacetylcinobufaginβOHHOHHHHHβOHCH3OH
2α,5β-Dihydroxyl-3-epi-desacetylcinobufaginHαOHOHHβOHHHHCH3OH
C1C2C33-epiC4C5C7C11C12C13C16
CinobufaginHHOHHHHHHCH3OAc
DesacetylcinobufaginHHOHHHHHHCH3OH
3-oxo-CinobufaginHHHHHHHCH3OAc
3-oxo-desacetylcinobufaginHHC=OHHHHHCH3OH
3-oxo-12α hydroxyl-cinobufaginHHC=OHHHHαOHCH3OAc
3-EpidesacetylcinobufaginHHOHHHHHHCH3OH
3-epi-12β-hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
5β-Hydroxyl-cinobufaginHHOHHβOHHαOHHCH3OAc
5β-Hydroxyl desacetylcinobufaginHHOHHβOHHHHCH3OH
5β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHβOHHHHCH3OH
7β-Hydroxyl-cinobufaginHHOHHHβOHHHCH3OAc
11α-Hydroxyl-cinobufaginHHOHHHHHβOHCH3OAc
12α-Hydroxyl-cinobufaginHHOHHHHHHαOHOAc
12β-Hydroxyl-cinobufaginHHOHHHHHHCH3OAc
12β-Hydroxyl desacetylcinobufaginHHOHHHHHβOHCH3OH
12β-Hydroxyl-3-epi-desacetylcinobufaginHHOHHHHHβOHCH3OH
1α-Hydroxyl-3-epi-desacetylcinobufaginαOHHOHHHHHHCH3OH
1β-Hydroxyl-3-epi-desacetylcinobufaginβOHHOHHHHHHCH3OH
1α,5α-Dihydroxyl-3-epi-desacetylcinobufaginαOHHOHHαOHHHHCH3OH
1β,12β-Dihydroxyl cinobufaginβOHHOHHHHHβOHCH3OAc
5β,12β-Dihydroxyl cinobufaginHHOHHβOHHHβOHCH3OAc
4β,11α-Dihydroxyl cinobufaginHHOHβOHHHαOHHCH3OAc
4β,12α-Dihydroxyl cinobufaginHHOHβOHHHHαOHCH3OAc
1β,12β-Dihydroxyl desacetylcinobufaginβOHHOHHHHHβOHCH3OH
2α,5β-Dihydroxyl-3-epi-desacetylcinobufaginHαOHOHHβOHHHHCH3OH
Metabolic pathways of cinobufagin. The main metabolic pathways of cinobufagin are hydroxylation, deacetylation and isomerization.
Figure 4

Metabolic pathways of cinobufagin. The main metabolic pathways of cinobufagin are hydroxylation, deacetylation and isomerization.

Biotransformation of natural products has great potential to generate new drugs, and the bioconversion of cinobufagin is possible using fungi of Mucor spinosus.[106] Three biotransformation products were obtained and all showed cytotoxic activity in four tumour cell lines of Bel 7420, BGC 823, HeLa and HL 60.[107] Besides, cunninghamella elegans[108] and syncephalastrum racemosum[109] have also been shown to be used for the biotransformation of cinobufagin, with the resulting transformation products possessing some biological activity and serving as a reference for the identification of cinobufagin metabolites in humans.

Conclusions and Perspectives

The prominent feature of cancer is its uncontrollable proliferation, which is a serious health hazard. Cinobufagin is derived from the dried secretions of the postauricular or cutaneous glands of Chinese toad or black orbital toad, which is one of the main active ingredients. A number of studies have demonstrated the efficacy of cinobufagin in the treatment of many cancers in vivo and in vitro, such as rectal cancer, colorectal cancer, oral squamous cell carcinoma, osteosarcoma, nasopharyngeal cancer, melanoma, cholangiocarcinoma, glioblastoma multiforme, hepatocellular carcinoma, gastric cancer, oesophageal squamous cell carcinoma, prostate cancer, breast cancer and cervical cancer. The mechanisms of action involved are also described in this article, cinobufagin can induce apoptosis through activation of the death receptor pathway and mitochondrial pathway, inhibit cancer cell proliferation, and block cell cycle in S-phase or G2/M-phase. In addition, it can also exert anticancer effects by inhibiting cancer angiogenesis, triggering DNA damage and relieving cancer pain. To further explore its anticancer mechanism, many scholars have investigated the metabolic process and anticancer effects of cinobufagin in vivo through pharmacokinetic analysis and the establishment of xenograft models. In recent years, good progress has been made regarding tumour immunotherapy. Cinobufagin upregulates the gene expression of antibacterial peptides (AMPs) hBD-2 and hBD-3 in dendritic cells, induces neutrophils to secrete HNP1-3 and hCAP-18/LL-37, and enhances the antibacterial activity of neutrophils, indicating that cinobufagin has a wide range of functions and further supports its anticancer potential.[110]

The structure and hydrophobicity of cinobufagin and resibufogenin in crude extracts of toad venom are similar, so it is difficult to separate them. However, cyclodextrin as mobile phase additive could significantly improve the separation and purification of cinobufagin.[111, 112] In addition, silica gel column chromatography with cyclohexane–acetone (5 : 1) as the mobile phase and high-speed counter-current chromatography (a two-phase solvent system consisting of n-hexane: chloroform: methanol: water (4 : 1 : 2.5 : 5 and 4 : 1 : 4 : 5, v/v)) can be used for the efficient purification of cinobufagin.[113, 114] Given the limitations of natural resources, the artificial synthesis of cinobufagin needs to be considered, which is important for reducing the cost of new drug development and protecting natural resources. In addition, the target and anticancer mechanism of action for cinobufagin needs to be elucidated clearly. Although the toxic effects of cinobufagin in vivo are not obvious at pharmacological doses, the systematic safety evaluation is lacking such as acute toxicity, long-term toxicity and reproductive toxicity, and toxicokinetics should be further explored. Taking 120 patients with bone metastasis as the study object, the clinical efficacy of intravenous zoledronic acid combined with oral cinobufagin in the treatment of metastatic bone tumours was improved.[115] Although experimental data for direct clinical use are relatively scarce, cinobufagin has the potential to be further developed as a new drug against cancer.

Author Contributions

HZ: conceptualization. C-LD and R-JZ: writing-original. C-LD, KR and HZ: writing-review and editing. HZ, PA and Y-QD: verification and recommendation. All authors contributed to the article and approved the submitted version.

Funding

This work was supported by funds from the National Natural Science Foundation of China (No. 82174023).

Ethical Statement

There are no ethical issues related to this work.

Conflict of Interest

The Authors declare that they have no conflict of interests to disclose.

Data Availability Statement

No new data were generated or analysed in support of this research.

References

1.

Pérez-Herrero
 
E
,
Fernández-Medarde
 
A.
 
Advanced targeted therapies in cancer: drug nanocarriers, the future of chemotherapy
.
Eur J Pharm Biopharm
 
2015
;
93
:
52
79
. https://doi.org/10.1016/j.ejpb.2015.03.018

2.

Liu
 
Z
,
Li
 
Z
,
Zhang
 
Y
 et al. .
Interpretation on the report of Global Cancer Statistics 2020
.
J Multidisciplin Cancer Manage
 
2021
;
7
:
1
14
.

3.

Jung
 
JH
,
Hwang
 
J
,
Kim
 
JH
 et al. .
Phyotochemical candidates repurposing for cancer therapy and their molecular mechanisms
.
Semin Cancer Biol
 
2021
;
68
:
164
74
. https://doi.org/10.1016/j.semcancer.2019.12.009

4.

Shi
 
X
,
Cheng
 
Y
,
Wang
 
J
 et al. .
3D printed intelligent scaffold prevents recurrence and distal metastasis of breast cancer
.
Theranostics
 
2020
;
10
:
10652
64
. https://doi.org/10.7150/thno.47933

5.

Liu
 
J
,
Wang
 
S
,
Zhang
 
Y
 et al. .
Traditional Chinese medicine and cancer: history, present situation, and development
.
Thorac Cancer
 
2015
;
6
:
561
9
. https://doi.org/10.1111/1759-7714.12270

6.

Chen
 
Y
,
Zhu
 
Z
,
Chen
 
J
 et al. .
Terpenoids from Curcumae Rhizoma: their anticancer effects and clinical uses on combination and versus drug therapies
.
Biomed Pharmacother
 
2021
;
138
:
111350
. https://doi.org/10.1016/j.biopha.2021.111350

7.

Zhang
 
C
,
Sui
 
X
,
Jiang
 
Y
 et al. .
Antitumor effects of icaritin and the molecular mechanisms
.
Discov Med
 
2020
;
29
:
5
16
.

8.

Chen
 
T
,
Yang
 
P
,
Jia
 
Y.
 
Molecular mechanisms of astragaloside-IV in cancer therapy (Review)
.
Int J Mol Med
 
2021
;
47
:
13
. https://doi.org/10.3892/IJMM.2021.4846

9.

Zhao
 
L
,
Zhang
 
Y
,
Li
 
Y
 et al. .
Therapeutic effects of ginseng and ginsenosides on colorectal cancer
.
Food Funct
 
2022
;
13
:
6450
66
. https://doi.org/10.1039/d2fo00899h

10.

Shi
 
MJ
,
Dong
 
BS
,
Yang
 
WN
 et al. .
Preventive and therapeutic role of Tanshinone ⅡA in hepatology
.
Biomed Pharmacother
 
2019
;
112
:
108676
. https://doi.org/10.1016/j.biopha.2019.108676

11.

Wei
 
WL
,
Hou
 
JJ
,
Wang
 
X
 et al. .
Venenum bufonis: an overview of its traditional use, natural product chemistry, pharmacology, pharmacokinetics and toxicology
.
J Ethnopharmacol
 
2019
;
237
:
215
35
. https://doi.org/10.1016/j.jep.2019.03.042

12.

Qi
 
F
,
Zhao
 
L
,
Zhou
 
A
 et al. .
The advantages of using traditional Chinese medicine as an adjunctive therapy in the whole course of cancer treatment instead of only terminal stage of cancer
.
Biosci Trends
 
2015
;
9
:
16
34
. https://doi.org/10.5582/bst.2015.01019

13.

Qi
 
J
,
Tan
 
CK
,
Hashimi
 
SM
 et al. .
Toad glandular secretions and skin extractions as anti-inflammatory and anticancer agents
.
Evid Based Complement Alternat Med
 
2014
;
2014
:
312684
. https://doi.org/10.1155/2014/312684

14.

Kou
 
G
,
Qin
 
Z
,
Deng
 
Y
 et al. .
Reasearch progress on Bufoins Venenum
.
Chin Trad Herb Drugs
 
2014
;
45
:
3185
9
.

15.

Zhao
 
Q
,
Meng
 
F
,
Liu
 
A.
 
Advances in study on Venenum Bufonis
.
Chin Trad Herb Drugs
 
2004
;
35
:
126
9
.

16.

Zhou
 
Y
,
Ji
 
A
,
Song
 
S.
 
Study on application of Venenum Bufoins
.
Chin J Biochem Pharm
 
2009
;
30
:
203
6
.

17.

Qi
 
F
,
Li
 
A
,
Inagaki
 
Y
 et al. .
Antitumor activity of extracts and compounds from the skin of the toad Bufo bufo gargarizans Cantor
.
Int Immunopharmacol
 
2011
;
11
:
342
9
. https://doi.org/10.1016/j.intimp.2010.12.007

18.

Li
 
FJ
,
Hu
 
JH
,
Ren
 
X
 et al. .
Toad venom: a comprehensive review of chemical constituents, anticancer activities, and mechanisms
.
Arch Pharm (Weinheim)
 
2021
;
354
:
e2100060
. https://doi.org/10.1002/ardp.202100060

19.

Li
 
J
,
Zhang
 
Z
,
Deng
 
H
 et al. .
Cinobufagin-loaded and folic acid-modified polydopamine nanomedicine combined with photothermal therapy for the treatment of lung cancer
.
Front Chem
 
2021
;
9
:
637754
. https://doi.org/10.3389/fchem.2021.637754.

20.

Wang
 
L
,
Mi
 
R
,
Wei
 
X
 et al. .
Cinobufagin treatment of relapsed or refractory hematological malignant tumor: 8 cases report and literatures review
.
Chin J Hematol
 
2015
;
36
:
954
6
.

21.

Yin
 
PH
,
Liu
 
X
,
Qiu
 
YY
 et al. .
Anti-tumor activity and apoptosis-regulation mechanisms of bufalin in various cancers: new hope for cancer patients
.
Asian Pac J Cancer Prev
 
2012
;
13
:
5339
43
. https://doi.org/10.7314/apjcp.2012.13.11.5339

22.

Stratton
 
MR
,
Campbell
 
PJ
,
Futreal
 
PA.
 
The cancer genome
.
Nature
 
2009
;
458
:
719
24
. https://doi.org/10.1038/nature07943.

23.

Bai
 
Y
,
Wang
 
X
,
Cai
 
M
 et al. .
Cinobufagin suppresses colorectal cancer growth via STAT3 pathway inhibition
.
Am J Cancer Res
 
2021
;
11
:
200
14
.

24.

Xu
 
ZW
,
Wang
 
FM
,
Gao
 
MJ
 et al. .
Cardiotonic steroids attenuate ERK phosphorylation and generate cell cycle arrest to block human hepatoma cell growth
.
J Steroid Biochem Mol Biol
 
2011
;
125
:
181
91
. https://doi.org/10.1016/j.jsbmb.2010.12.016

25.

Li
 
X
,
Chen
 
C
,
Dai
 
Y
 et al. .
Cinobufagin suppresses colorectal cancer angiogenesis by disrupting the endothelial mammalian target of rapamycin/hypoxia-inducible factor 1α axis
.
Cancer Sci
 
2019
;
110
:
1724
34
. https://doi.org/10.1111/cas.13988

26.

Niu
 
J
,
Wang
 
J
,
Zhang
 
Q
 et al. .
Cinobufagin-induced DNA damage response activates G2/M checkpoint and apoptosis to cause selective cytotoxicity in cancer cells
.
Cancer Cell Int
 
2021
;
21
:
446
. https://doi.org/10.1186/s12935-021-02150-0

27.

Yuan
 
Z
,
Shi
 
X
,
Qiu
 
Y
 et al. .
Reversal of P-gp-mediated multidrug resistance in colon cancer by cinobufagin
.
Oncol Rep
 
2017
;
37
:
1815
25
. https://doi.org/10.3892/or.2017.5410

28.

Li
 
C
,
Hashimi
 
SM
,
Cao
 
S
 et al. .
Chansu inhibits the expression of cortactin in colon cancer cell lines in vitro and in vivo
.
BMC Complement Altern Med
 
2015
;
15
:
207
. https://doi.org/10.1186/s12906-015-0723-3

29.

Li
 
C
,
Hashimi
 
SM
,
Cao
 
S
 et al. .
The mechanisms of chansu in inducing efficient apoptosis in colon cancer cells
.
Evid Based Complement Alternat Med
 
2013
;
2013
:
849054
. https://doi.org/10.1155/2013/849054

30.

Jo
 
S
,
Yang
 
E
,
Lee
 
Y
 et al. .
Cinobufagin exerts anticancer activity in oral squamous cell carcinoma cells through downregulation of ANO1
.
Int J Mol Sci
 
2021
;
22
:
12037
. https://doi.org/10.3390/ijms222112037

31.

Dai
 
G
,
Zheng
 
D
,
Guo
 
W
 et al. .
Cinobufagin induces apoptosis in osteosarcoma cells via the mitochondria-mediated apoptotic pathway
.
Cell Physiol Biochem
 
2018
;
46
:
1134
47
. https://doi.org/10.1159/000488842

32.

Dai
 
G
,
Yu
 
L
,
Yang
 
J
 et al. .
The synergistic antitumor effect of cinobufagin and cisplatin in human osteosarcoma cell line in vitro and in vivo
.
Oncotarget
 
2017
;
8
:
85150
68
. https://doi.org/10.18632/oncotarget.19554

33.

Cao
 
Y
,
Yu
 
L
,
Dai
 
G
 et al. .
Cinobufagin induces apoptosis of osteosarcoma cells through inactivation of Notch signaling
.
Eur J Pharmacol
 
2017
;
794
:
77
84
. https://doi.org/10.1016/j.ejphar.2016.11.016

34.

Yin
 
JQ
,
Wen
 
L
,
Wu
 
LC
 et al. .
The glycogen synthase kinase-3β/nuclear factor-kappa B pathway is involved in cinobufagin-induced apoptosis in cultured osteosarcoma cells
.
Toxicol Lett
 
2013
;
218
:
129
36
. https://doi.org/10.1016/j.toxlet.2012.11.006

35.

Ma
 
X
,
Suo
 
Z
,
Ma
 
X
 et al. .
Cinobufagin inhibits tumor progression and reduces doxorubicin resistance by enhancing FOXO1-mediated transcription of FCGBP in osteosarcoma
.
J Ethnopharmacol
 
2022
;
296
:
115433
. https://doi.org/10.1016/j.jep.2022.115433

36.

Ma
 
K
,
Zhang
 
C
,
Huang
 
MY
 et al. .
Cinobufagin induces autophagy-mediated cell death in human osteosarcoma U2OS cells through the ROS/JNK/p38 signaling pathway
.
Oncol Rep
 
2016
;
36
:
90
8
. https://doi.org/10.3892/or.2016.4782

37.

Pan
 
Z
,
Luo
 
Y
,
Xia
 
Y
 et al. .
Cinobufagin induces cell cycle arrest at the S phase and promotes apoptosis in nasopharyngeal carcinoma cells
.
Biomed Pharmacother
 
2020
;
122
:
109763
. https://doi.org/10.1016/j.biopha.2019.109763

38.

Hou
 
R
,
Liu
 
X
,
Yang
 
H
 et al. .
Chemically synthesized cinobufagin suppresses nasopharyngeal carcinoma metastasis by inducing ENKUR to stabilize p53 expression
.
Cancer Lett
 
2022
;
531
:
57
70
. https://doi.org/10.1016/j.canlet.2022.01.025

39.

Kim
 
GH
,
Fang
 
XQ
,
Lim
 
WJ
 et al. .
Cinobufagin suppresses melanoma cell growth by inhibiting LEF1
.
Int J Mol Sci
 
2020
;
21
:
6706
. https://doi.org/10.3390/ijms21186706

40.

Pan
 
Z
,
Zhang
 
X
,
Yu
 
P
 et al. .
Cinobufagin induces cell cycle arrest at the G2/M phase and promotes apoptosis in malignant melanoma cells
.
Front Oncol
 
2019
;
9
:
853
. https://doi.org/10.3389/fonc.2019.00853

41.

Zhang
 
L
,
Huang
 
X
,
Guo
 
T
 et al. .
Study of cinobufagin as a promising anticancer agent in uveal melanoma through intrinsic apoptosis pathway
.
Front Oncol
 
2020
;
10
:
325
. https://doi.org/10.3389/fonc.2020.00325

42.

Ren
 
J
,
Wang
 
S
,
Jin
 
L
 et al. .
Cinobufagin inhibits tumor growth by inducing apoptosis through Notch signaling pathways in human cholangiocarcinoma
.
Transl Cancer Res
 
2019
;
8
:
2461
9
. https://doi.org/10.21037/tcr.2019.10.06

43.

He
 
K
,
Wang
 
GX
,
Zhao
 
LN
 et al. .
Cinobufagin is a selective anti-cancer agent against tumors with EGFR amplification and PTEN deletion
.
Front Pharmacol
 
2021
;
12
:
775602
. https://doi.org/10.3389/fphar.2021.775602

44.

Deng
 
X
,
Sheng
 
J
,
Liu
 
H
 et al. .
Cinobufagin promotes cell cycle arrest and apoptosis to block human esophageal squamous cell carcinoma cells growth via the p73 signalling pathway
.
Biol Pharm Bull
 
2019
;
42
:
1500
9
. https://doi.org/10.1248/bpb.b19-00174

45.

Yang
 
AL
,
Wu
 
Q
,
Hu
 
ZD
 et al. .
A network pharmacology approach to investigate the anticancer mechanism of cinobufagin against hepatocellular carcinoma via downregulation of EGFR-CDK2 signaling
.
Toxicol Appl Pharmacol
 
2021
;
431
:
115739
. https://doi.org/10.1016/j.taap.2021.115739

46.

Zhao
 
L
,
Fu
 
L
,
Xu
 
Z
 et al. .
The anticancer effects of cinobufagin on hepatocellular carcinoma Huh-7 cells are associated with activation of the p73 signaling pathway
.
Mol Med Rep
 
2019
;
19
:
4119
28
. https://doi.org/10.3892/mmr.2019.10108

47.

Jin
 
X
,
Wang
 
J
,
Zou
 
S
 et al. .
Cinobufagin triggers defects in spindle formation and cap-dependent translation in liver cancer cells by inhibiting the AURKA-mTOR-eIF4E Axis
.
Am J Chin Med
 
2020
;
48
:
651
78
. https://doi.org/10.1142/S0192415X20500330

48.

Qi
 
F
,
Inagaki
 
Y
,
Gao
 
B
 et al. .
Bufalin and cinobufagin induce apoptosis of human hepatocellular carcinoma cells via Fas- and mitochondria-mediated pathways
.
Cancer Sci
 
2011
;
102
:
951
8
. https://doi.org/10.1111/j.1349-7006.2011.01900.x.

49.

Dong
 
YQ
,
Ma
 
WL
,
Gu
 
JB
 et al. .
Effect of cinobufagin on nuclear factor-kappaB pathway in HepG2 cells
.
J South Med Univ
 
2010
;
30
:
137
9
.

50.

Niu
 
K
,
Cheng
 
Y.
 
Effect of cinobufacini injection on Bel-7402 cell proliferation and cycle of human hepatoma
.
Shanxi J TCM
 
2013
;
29
:
47
9
.

51.

Lu
 
XS
,
Qiao
 
YB
,
Li
 
Y
 et al. .
Preclinical study of cinobufagin as a promising anti-colorectal cancer agent
.
Oncotarget
 
2017
;
8
:
988
98
. https://doi.org/10.18632/oncotarget.13519

52.

Xiong
 
X
,
Lu
 
B
,
Tian
 
Q
 et al. .
Inhibition of autophagy enhances cinobufagin-induced apoptosis in gastric cancer
.
Oncol Rep
 
2019
;
41
:
492
500
. https://doi.org/10.3892/or.2018.6837

53.

Hirasaki
 
Y
,
Okabe
 
A
,
Fukuyo
 
M
 et al. .
Cinobufagin inhibits proliferation of acute myeloid leukaemia cells by repressing c-Myc pathway-associated genes
.
Chem Biol Interact
 
2022
;
360
:
109936
. https://doi.org/10.1016/j.cbi.2022.109936

54.

Zhang
 
L
,
Liang
 
B
,
Xu
 
H
 et al. .
Cinobufagin induces FOXO1-regulated apoptosis, proliferation, migration, and invasion by inhibiting G9a in non-small-cell lung cancer A549 cells
.
J Ethnopharmacol
 
2022
;
291
:
115095
. https://doi.org/10.1016/j.jep.2022.115095

55.

Pascual Alonso
 
I
,
Rivera Méndez
 
L
,
Almeida García
 
F
 et al. .
Bufadienolides preferentially inhibit aminopeptidase N among mammalian metallo-aminopeptidases; relationship with effects on human melanoma MeWo cells
.
Int J Biol Macromol
 
2023
;
229
:
825
37
. https://doi.org/10.1016/j.ijbiomac.2022.12.280

56.

Baek
 
SH
,
Kim
 
C
,
Lee
 
JH
 et al. .
Cinobufagin exerts anti-proliferative and pro-apoptotic effects through the modulation ROS-mediated MAPKs signaling pathway
.
Immunopharmacol Immunotoxicol
 
2015
;
37
:
265
73
. https://doi.org/10.3109/08923973.2015.1027916

57.

Yu
 
CH
,
Kan
 
SF
,
Pu
 
HF
 et al. .
Apoptotic signaling in bufalin- and cinobufagin-treated androgen-dependent and -independent human prostate cancer cells
.
Cancer Sci
 
2008
;
99
:
2467
76
. https://doi.org/10.1111/j.1349-7006.2008.00966.x

58.

Zhu
 
L
,
Chen
 
Y
,
Wei
 
C
 et al. .
Anti-proliferative and pro-apoptotic effects of cinobufagin on human breast cancer MCF-7 cells and its molecular mechanism
.
Nat Prod Res
 
2018
;
32
:
493
7
. https://doi.org/10.1080/14786419.2017.1315575

59.

Ma
 
L
,
Song
 
B
,
Jin
 
H
 et al. .
Cinobufacini induced MDA-MB-231 cell apoptosis-associated cell cycle arrest and cytoskeleton function
.
Bioorg Med Chem Lett
 
2012
;
22
:
1459
63
. https://doi.org/10.1016/j.bmcl.2011.11.095

60.

Yang
 
P
,
Yu
 
TX
,
Ma
 
RD
 et al. .
Cell cycle arrest and apoptosis induced by tubeimoside in HeLa cell
.
Chin J Cancer
 
2002
;
21
:
346
50
.

61.

Elmore
 
S.
 
Apoptosis: a review of programmed cell death
.
Toxicol Pathol
 
2007
;
35
:
495
516
. https://doi.org/10.1080/01926230701320337

62.

Fuchs
 
Y
,
Steller
 
H.
 
Live to die another way: modes of programmed cell death and the signals emanating from dying cells
.
Nat Rev Mol Cell Biol
 
2015
;
16
:
329
44
. https://doi.org/10.1038/nrm3999

63.

Cao
 
F
,
Kang
 
X
,
Wang
 
L.
 
Effects of cinobufagin on apoptosis in U-2OS osteosarcomas cells
.
Chin J Repar Reconstr Surg
 
2014
;
28
:
349
53
.

64.

Yeh
 
JY
,
Huang
 
WJ
,
Kan
 
SF
 et al. .
Effects of bufalin and cinobufagin on the proliferation of androgen dependent and independent prostate cancer cells
.
Prostate
 
2003
;
54
:
112
24
. https://doi.org/10.1002/pros.10172

65.

Zhang
 
J
,
Hong
 
Y
,
Jiang
 
L
 et al. .
Global metabolomic and lipidomic analysis reveal the synergistic effect of bufalin in combination with cinobufagin against HepG2 Cells
.
J Proteome Res
 
2020
;
19
:
873
83
. https://doi.org/10.1021/acs.jproteome.9b00681

66.

Kong
 
B
,
Zuo
 
MR
,
Liu
 
YH.
 
The research on the anti-glioma effect and mechanism of cinobufagin
.
J Sichuan Univ Med Sci Ed
 
2018
;
49
:
388
93
.

67.

Crabtree
 
JS
,
Singleton
 
CS
,
Miele
 
L.
 
Notch signaling in neuroendocrine tumors
.
Front Oncol
 
2016
;
6
:
94
. https://doi.org/10.3389/fonc.2016.00094

68.

Qi
 
F
,
Li
 
A
,
Zhao
 
L
 et al. .
Apoptosis-inducing effect of cinobufacini on human hepatoma cell line HepG2 and its mechanism of action
.
Acta Pharmacol Sin
 
2010
;
45
:
318
23
.

69.

Botchkarev
 
VA
,
Flores
 
ER.
 
p53/p63/p73 in the epidermis in health and disease
.
Cold Spring Harb Perspect Med
 
2014
;
4
. https://doi.org/10.1101/cshperspect.a015248

70.

Bian
 
Y
,
Xue
 
M
,
Guo
 
X
 et al. .
Cinobufagin induces acute promyelocytic leukaemia cell apoptosis and PML-RARA degradation in a caspase-dependent manner by inhibiting the β-catenin signalling pathway
.
Pharm Biol
 
2022
;
60
:
1801
11
. https://doi.org/10.1080/13880209.2022.2118792

71.

Wang
 
Y
,
Shen
 
Y
,
Wang
 
S
 et al. .
The role of STAT3 in leading the crosstalk between human cancers and the immune system
.
Cancer Lett
 
2018
;
415
:
117
28
. https://doi.org/10.1016/j.canlet.2017.12.003

72.

Guo
 
S
,
Zhang
 
L
,
Li
 
N.
 
ANO1: more than just calcium-activated chloride channel in cancer
.
Front Oncol
 
2022
;
12
:
922838
. https://doi.org/10.3389/fonc.2022.922838

73.

Zhang
 
T
,
Yin
 
P
,
Zhang
 
Z
 et al. .
Deficiency of pigment epithelium-derived factor in nasopharyngeal carcinoma cells triggers the epithelial-mesenchymal transition and metastasis
.
Cell Death Dis
 
2017
;
8
:
e2838
. https://doi.org/10.1038/cddis.2017.114

74.

Sikandar
 
SS
,
Kuo
 
AH
,
Kalisky
 
T
 et al. .
Role of epithelial to mesenchymal transition associated genes in mammary gland regeneration and breast tumorigenesis
.
Nat Commun
 
2017
;
8
:
1669
. https://doi.org/10.1038/s41467-017-01666-2

75.

MacGrath
 
SM
,
Koleske
 
AJ.
 
Cortactin in cell migration and cancer at a glance
.
J Cell Sci
 
2012
;
125
:
1621
6
. https://doi.org/10.1242/jcs.093781

76.

Reinhard
 
L
,
Tidow
 
H
,
Clausen
 
MJ
 et al. .
Na+, K+-ATPase as a docking station: protein-protein complexes of the Na+, K+-ATPase
.
Cell Mol Life Sci
 
2013
;
70
:
205
22
. https://doi.org/10.1007/s00018-012-1039-9

77.

Felippe Gonçalves-de-Albuquerque
 
C
,
Ribeiro Silva
 
A
,
Ignácio da Silva
 
C
 et al. .
Na/K pump and beyond: Na/K-ATPase as a modulator of apoptosis and autophagy
.
Molecules
 
2017
;
22
:
578
. https://doi.org/10.3390/molecules22040578

78.

Sousa
 
LQ
,
Machado
 
KD
,
Oliveira
 
SF
 et al. .
Bufadienolides from amphibians: a promising source of anticancer prototypes for radical innovation, apoptosis triggering and Na+/K+-ATPase inhibition
.
Toxicon
 
2017
;
127
:
63
76
. https://doi.org/10.1016/j.toxicon.2017.01.004.

79.

Zhang
 
G
,
Wang
 
C
,
Sun
 
M
 et al. .
Cinobufagin inhibits tumor growth by inducing intrinsic apoptosis through AKT signaling pathway in human nonsmall cell lung cancer cells
.
Oncotarget
 
2016
;
7
:
28935
46
. https://doi.org/10.18632/oncotarget.7898

80.

Hanahan
 
D
,
Weinberg
 
RA.
 
Hallmarks of cancer: the next generation
.
Cell
 
2011
;
144
:
646
74
. https://doi.org/10.1016/j.cell.2011.02.013

81.

Wu
 
Q
,
Yang
 
Z
,
Nie
 
Y
 et al. .
Multi-drug resistance in cancer chemotherapeutics: mechanisms and lab approaches
.
Cancer Lett
 
2014
;
347
:
159
66
. https://doi.org/10.1016/j.canlet.2014.03.013

82.

Yang
 
A
,
Wu
 
Q
,
Chen
 
Q
 et al. .
Cinobufagin restrains the growth and triggers DNA damage of human hepatocellular carcinoma cells via proteasome-dependent degradation of thymidylate synthase
.
Chem Biol Interact
 
2022
;
360
:
109938
. https://doi.org/10.1016/j.cbi.2022.109938

83.

Chen
 
T
,
Hu
 
W
,
He
 
H
 et al. .
A study on the mechanism of cinobufagin in the treatment of paw cancer pain by modulating local β -endorphin expression in vivo
.
Evid Based Complement Alternat Med
 
2013
;
2013
:
851256
. https://doi.org/10.1155/2013/851256

84.

Apryani
 
E
,
Ali
 
U
,
Wang
 
ZY
 et al. .
The spinal microglial IL-10/β-endorphin pathway accounts for cinobufagin-induced mechanical antiallodynia in bone cancer pain following activation of α7-nicotinic acetylcholine receptors
.
J Neuroinflammation
 
2020
;
17
:
75
. https://doi.org/10.1186/s12974-019-1616-z

85.

Xu
 
LS
,
Feng
 
QL
,
Zhang
 
XP
 et al. .
Study on analgesic effect and mechanism of cinobufagin on rats with bone cancer pain
.
Natl Med J Chin
 
2019
;
99
:
1307
11
. https://doi.org/10.3760/cma.j.issn.0376-2491.2019.17.007

86.

Chen
 
T
,
Yuan
 
S
,
Wan
 
XN
 et al. .
Chinese herb cinobufagin-reduced cancer pain is associated with increased peripheral opioids by invaded CD3/4/8 lymphocytes
.
Oncotarget
 
2017
;
8
:
11425
41
. https://doi.org/10.18632/oncotarget.14005

87.

Wang
 
C
,
Mei
 
X
,
Wu
 
Y
 et al. .
Cinobufagin alleviates lipopolysaccharide-induced acute lung injury by regulating autophagy through activation of the p53/mTOR pathway
.
Front Pharmacol
 
2022
;
13
:
994625
. https://doi.org/10.3389/fphar.2022.994625

88.

Xie
 
S
,
Spelmink
 
L
,
Codemo
 
M
 et al. .
Cinobufagin modulates human innate immune responses and triggers antibacterial activity
.
PLoS One
 
2016
;
11
:
e0160734
. https://doi.org/10.1371/journal.pone.0160734

89.

Li
 
X
,
Bi
 
Z
,
Liu
 
S
 et al. .
Antifibrotic mechanism of cinobufagin in bleomycin-induced pulmonary fibrosis in mice
.
Front Pharmacol
 
2019
;
10
:
1021
. https://doi.org/10.3389/fphar.2019.01021

90.

Hirasaki
 
Y
,
Okabe
 
A
,
Fukuyo
 
M
 et al. .
Cinobufagin inhibits proliferation of acute myeloid leukaemia cells by repressing c-Myc pathway-associated genes
.
Chem Biol Interact
 
2022
;
360
:
109936
. https://doi.org/10.1016/j.cbi.2022.109936

91.

Wu
 
SC
,
Yi
 
PF
,
Guo
 
X
 et al. .
Cinobufagin enhances the protective efficacy of formalin-inactivated Salmonella typhimurium vaccine through Th1 immune response
.
Microb Pathog
 
2016
;
99
:
264
70
. https://doi.org/10.1016/j.micpath.2016.08.036

92.

Cui
 
X
,
Inagaki
 
Y
,
Xu
 
H
 et al. .
Anti-hepatitis B virus activities of cinobufacini and its active components bufalin and cinobufagin in HepG2.2.15 cells
.
Biol Pharm Bull
 
2010
;
33
:
1728
32
. https://doi.org/10.1248/bpb.33.1728

93.

Morishita
 
S
,
Shoji
 
M
,
Oguni
 
Y
 et al. .
Pharmacological actions of ‘kyushin’, a drug containing toad venom: cardiotonic and arrhythmogenic effects, and excitatory effect on respiration
.
Am J Chin Med
 
1992
;
20
:
245
56
. https://doi.org/10.1142/S0192415X92000254

94.

Wang
 
ZJ
,
Sun
 
L
,
Heinbockel
 
T.
 
Resibufogenin and cinobufagin activate central neurons through an ouabain-like action
.
PLoS One
 
2014
;
9
:
e113272
. https://doi.org/10.1371/journal.pone.0113272

95.

Yamahara
 
J
,
Tanaka
 
S
,
Matsuda
 
H
 et al. .
The mode of cardiac action of cardiotonic steroids isolated from Toad Cake in perfused working guinea-pig heart and effect of cinobufagin on experimental heart failure
.
Nihon Yakurigaku Zasshi
 
1986
;
88
:
413
23
. https://doi.org/10.1254/fpj.88.413

96.

Hsu
 
SS
,
Lin
 
YS
,
Liang
 
WZ.
 
Investigation of cytotoxic effect of the bufanolide steroid compound cinobufagin and its related underlying mechanism in brain cell models
.
J Biochem Mol Toxicol
 
2021
;
35
:
e22862
. https://doi.org/10.1002/jbt.22862

97.

Xu
 
L
,
Wang
 
S
,
Shen
 
H
 et al. .
Analgesic and toxic effects of venenum bufonis and its constituent compound cinobufagin: a comparative study
.
Neurotoxicol Teratol
 
2019a
;
73
:
49
53
. https://doi.org/10.1016/j.ntt.2019.03.004

98.

Toma
 
S
,
Hirai
 
Y
,
Sugimoto
 
C
 et al. .
Metabolic fate of bufalin and cinobufagin
.
Yakugaku Zasshi
 
1991
;
111
:
687
94
. https://doi.org/10.1248/yakushi1947.111.11_687

99.

Zhang
 
LS
,
Yoshida
 
T
,
Aoki
 
K
 et al. .
Metabolism of cinobufagin in rat liver microsomes. Identification of epimerized and deacetylated metabolites by liquid chromatography/mass spectrometry
.
Drug Metab Dispos
 
1991
;
19
:
917
9
.

100.

Zhang
 
Y
,
Tang
 
X
,
Liu
 
X
 et al. .
Simultaneous determination of three bufadienolides in rat plasma after intravenous administration of bufadienolides extract by ultra performance liquid chromatography electrospray ionization tandem mass spectrometry
.
Anal Chim Acta
 
2008
;
610
:
224
31
. https://doi.org/10.1016/j.aca.2008.01.029

101.

Liang
 
Y
,
Liu
 
AH
,
Qin
 
S
 et al. .
Simultaneous determination and pharmacokinetics of five bufadienolides in rat plasma after oral administration of Chansu extract by SPE-HPLC method
.
J Pharm Biomed Anal
 
2008
;
46
:
442
8
. https://doi.org/10.1016/j.jpba.2007.11.001

102.

Ma
 
XC
,
Xin
 
X
,
Zhang
 
BJ
 et al. .
Efficient isolation and purification of five products from microbial biotransformation of cinobufagin by high-speed counter-current chromatography
.
J Sep Sci
 
2010
;
33
:
2272
7
. https://doi.org/10.1002/jssc.201000212

103.

Xu
 
W
,
Luo
 
H
,
Zhang
 
Y
 et al. .
Simultaneous determination of five main active bufadienolides of Chan Su in rat plasma by liquid chromatography tandem mass spectrometry
.
J Chromatogr B Analyt Technol Biomed Life Sci
 
2007
;
859
:
157
63
. https://doi.org/10.1016/j.jchromb.2007.09.026

104.

Ning
 
J
,
Wu
 
TH
,
Tian
 
Y
 et al. .
Identification of cinobufagin metabolites in the bile of rats
.
Xenobiotica
 
2010
;
40
:
48
54
. https://doi.org/10.3109/00498250903331049

105.

He
 
X
,
Hu
 
H
,
Wu
 
Y
 et al. .
Urinary metabolites of cinobufagin in rats and their antiproliferative activities
.
Nat Prod Res
 
2012
;
26
:
489
99
. https://doi.org/10.1080/14786419.2010.510798

106.

He
 
X
,
Tang
 
J
,
Qiao
 
A
 et al. .
Cytotoxic biotransformed products from cinobufagin by Mucor spinosus and Aspergillus Niger
.
Steroids
 
2006
;
71
:
392
402
. https://doi.org/10.1016/j.steroids.2005.12.003

107.

Zhang
 
W
,
Ye
 
M
,
Qu
 
GQ
 et al. .
Microbial hydroxylation of cinobufagin by Mucor spinosus
.
J Asian Nat Prod Res
 
2005
;
7
:
225
9
. https://doi.org/10.1080/10286020410001690136

108.

Qiao
 
L
,
Zhou
 
YZ
,
Qi
 
XL
 et al. .
Biotransformation of cinobufagin by Cunninghamella elegans
.
J Antibiot (Tokyo)
 
2007
;
60
:
264
4
. https://doi.org/10.1038/ja.2007.32

109.

Ma
 
XC
,
Xin
 
XL
,
Liu
 
KX
 et al. .
Microbial transformation of cinobufagin by Syncephalastrum racemosum
.
J Nat Prod
 
2008
;
71
:
1268
70
. https://doi.org/10.1021/np800210a

110.

Xie
 
S
,
Spelmink
 
L
,
Codemo
 
M
 et al. .
Cinobufagin modulates human innate immune responses and triggers antibacterial activity
.
PLoS One
 
2016
;
16
:
e0160734
. https://doi.org/10.1371/journal.pone.0160734

111.

Xing
 
J
,
Chen
 
L
,
Song
 
J
 et al. .
Separation and determination of resibufogenin and cinobufagin in Chansu using reversed-phase liquid chromatography with γ-cyclodextrin as mobile-phase modifier
.
J Sep Sci
 
2012
;
35
:
1884
92
. https://doi.org/10.1002/jssc.201200058

112.

Zhao
 
Y
,
Luo
 
X
,
Ming
 
Y
 et al. .
Rapid separation and determination of resibufogenin and cinobufagin in toad venom and Liushen tablet by beta-cyclodextrin modified micellar electrokinetic chromatography
.
J Pharm Biomed Anal
 
2006
;
41
:
124
8
. https://doi.org/10.1016/j.jpba.2005.10.046

113.

Yang
 
Z
,
Luo
 
H
,
Wang
 
H
 et al. .
Preparative isolation of bufalin and cinobufagin from Chinese traditional medicine ChanSu
.
J Chromatogr Sci
 
2008
;
46
:
81
5
. https://doi.org/10.1093/chromsci/46.1.81

114.

Xin
 
XL
,
Liu
 
J
,
Ma
 
XC
 et al. .
Preparative separation of four major bufadienolides from the Chinese traditional medicine, Chansu, using high-speed counter-current chromatography
.
Nat Prod Commun
 
2010
;
5
:
1031
4
.

115.

Wang
 
T
,
Zhang
 
L
,
Han
 
L
 et al. .
Clinical effect of intravenous infusion of zoledronic acid combined with oral medication of cinobufagin in the treatment of metastatic bone tumors
.
Pak J Pharm Sci
 
2018a
;
31
:
1609
12
.

Author notes

Chun-Lan Dai and Run-jing Zhang contributed equally to this work.

This article is published and distributed under the terms of the Oxford University Press, Standard Journals Publication Model (https://dbpia.nl.go.kr/pages/standard-publication-reuse-rights)