Copyright
©The Author(s) 2025.
World J Clin Oncol. Nov 24, 2025; 16(11): 110911
Published online Nov 24, 2025. doi: 10.5306/wjco.v16.i11.110911
Published online Nov 24, 2025. doi: 10.5306/wjco.v16.i11.110911
Table 1 The in vitro anticancer activity of cardamonin
| Cell lines | Effects | Molecular mechanism | Ref. |
| Human hepatocellular carcinoma (HepG2) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G1 phase; (3) Induced apoptosis by accumulating mitochondrial ROS, upregulating caspases expression (caspase 3/7, caspase-8 and caspase-9), upregulating expression of proapoptotic proteins (FADD, Fas, TRIAL, HIF-1, cleaved caspase-3) and downregulating expression of antiapoptotic proteins (HSP60, HSP27 and HSP70, XIAP, catalase, clusterin, and surviving); and (4) Inhibited cellular colonization | Deactivated NF-κB signalling pathway by blocking nuclear translocation of NF-κB and increasing phosphorylation of p53 | [61] |
| (1) Inhibited cellular viability; and (2) Induced apoptosis by cell shrinkage and chromatin condensation | Blocked TSP50-mediated NF-κB signaling pathway by downregulating protein and mRNA expressions of TSP50 | [35] | |
| Human mesenchymal triple-negative breast cancer cells (SUM190; TLR3 activation-enhanced CSC phenotypes) | Inhibited mammosphere formation by blocking TLR3 on the induced CSCs by downregulating protein expression of associated stem-cell genes (OCT4 and c-Myc) | Deactivated concurrently Wnt/β-catenin and NF-κB signalling pathways by concurrent inhibition of nuclear translocations of both β-catenin and NF-κB | [27] |
| Human mesenchymal triple-negative breast cancer (SUM190 and MDA-MB231), SUM190 (inflammatory breast cancer cell line, invasive ductal carcinoma, ER-PR-HER2-/+) and CAMA-1 cells (adenocarcinoma, ER+/PR-, oncogenic mutations in PTEN and p53, in-frame mutation in E-cadherin gene) and MCF-7 cells (tolerant to 5-FU, doxorubicin, or paclitaxel) | (1) Augmented antimetastatic effects of 5-FU, doxorubicin or paclitaxel; (2) Inhibited mammosphere formation through inhibiting cellular colonization and abolishing self-renewal capacity of CSCs; and (3) Downregulated mRNA expression of CSCs-associated genes (ALDH1, SOX2, c-Myc, OCT4, NANOG) and CSCs-associated histone modifier genes (EZH2, SETDB1, SMYD3) | Deactivated NF-κB and STAT3 signalling pathways by downregulating protein expressions of NF-κB/IKβα and STAT3, respectively | [38] |
| MDA-MB-231 cells (transfected with pGL3-TSP50 promoter plasmid) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G2/M-phase by upregulating expression of CDK inhibitor (p21) in TSP50-overexpressed cells; and (3) Induced mitochondrial-dependent apoptosis as appeared by cells shrinkage and chromatin condensation with DNA fragmentations, upregulating expression of pro-apoptotic proteins (BAX, activated caspase-9, cleaved caspase-3) and downregulating expression of the anti-apoptotic protein BCL-2 | Blocked TSP50-mediated NF-κB signaling pathway through downregulating expression of p65 nuclear with downregulating protein and mRNA expression of TSP50 | [35] |
| Triple negative breast cancer cells (BT-549, ER-negative) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G2/M phase; (3) Induced mitochondrial apoptosis by upregulating expression of cytochrome c, proapoptotic proteins (BAX, cleaved caspase 3 and cleaved PARP) and downregulating expression of the antiapoptotic protein BCL-2; (4) Inhibited cellular colonization; and (5) Retarded invasion by blocking EMT through upregulating protein expression of the epithelia marker E-cadherin and downregulating protein expression of the mesenchymal markers N-cadherin and vimentin | Deactivated Wnt/β-catenin signaling pathway by downregulating protein expression of β-catenin and β-catenin downstream targets (cyclin D1, c-Myc, VEGF, and CDK-4) | [37] |
| Human TNBC cell line (MDA-MB-231) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G2/M phase; (3) Induced mitochondrial apoptosis through accumulating ROS, downregulating expressions of the anti-apoptotic proteins BCL-2 and upregulating expressions of pro-apoptotic proteins (BAX, cleaved caspase-3 and PARP); and (4) Metabolic reprograming through inhibiting HIF-1α by downregulating protein and mRNA expressions of HIF-1α and HIF-1α target genes (PDHK1 and LDHA) | Deactivated mTOR signalling pathway by downregulating protein expressions of mTOR and S6K | [32] |
| (1) Inhibited cellular viability; (2) Arresting cell cycle at G2/M phase; (3) Induced mitochondrial apoptosis through accumulating ROS, upregulating expression of pro-apoptotic proteins (cleaved caspase-3, PARP, and BAX) and downregulating expression of the antiapoptotic protein BCL-2; and (4) Activated FOXO3a by upregulating protein expression of FOXO3a and its target genes (p21, p27, Bim), and upstreaming JNK | Activated JNK/mTOR signalling pathway | [33] | |
| Human ER positive breast cancer cells (MCF-7); resistant to Rapamycin and its analogues, mTOR-inhibitors resistant cells) | Inhibited cellular viability of mTOR resistant cells | Deactivated mTOR signaling pathway by downregulating protein expressions of mTOR, S6K1, and raptor | [36] |
| Human TNBC cells (MDA-231 and MDA-468) | (1) Inhibited cellular viability; and (2) Desensitized cells by blocking tumor resistant through downregulating expression of PD-L1, and CCL2 | Deactivated JAK/STAT axis signaling pathway by downregulating mRNA expression of MUC1, JAK1, and STAT3; deactivated NF-κB signaling pathway by downregulating protein expressions of NF-κB1 (p50), NF-κB2 (p52) and Nrf2 | [34] |
| Human colorectal cancer cells (HCT116, lack p53) | (1) Inhibited cellular viability; and (2) Induced apoptosis (early and late stages) by increasing ROS formation, and upregulating expression of antiapoptotic proteins (BID, BAX, cleaved caspase-8, cleaved caspase 9, and cleaved caspase 3 and cleaved PARP), and downregulating expression of antiapoptotic proteins (cIAP-1, cFLIP, XIAP, BCL-2 and surviving) | Activated CHOP and SP1-dependent TRAIL by upregulating expressions of proteins and mRNA of TRAIL-DR4 and TRAIL-DR5 (death receptors) and upregulating protein expressions of CHOP and SP1 (specificity protein 1) | [58] |
| Human colon adenocarcinoma cells (SW480) | (1) Inhibited cellular viability; and (2) Arrested cell cycle at G2/M phase | Deactivated Wnt/β-catenin signaling pathway by down-regulating protein expressions of β-catenin and expressions of β-catenin dependent genes (c-Myc, and cyclin D1) | [57] |
| Human colorectal cancer cells (HCT116) | (1) Inhibited cellular viability; (2) Arrested cell growth at G2/M phase; and (3) Induced autophagic cellular death by increasing level of LC3 proteins (LC3-II) and increasing autophagosomes | Activated JNK signaling pathway by increasing phosphorylation of JNK isomers (JNK1 and JNK2) and upregulating protein and mRNA expressions of p53 | [54] |
| Human colorectal cancer cells (SW620) | (1) Inhibited cellular viability; (2) Arrested cell cycle at S phase; and (3) Induced mitochondrial apoptosis by increasing number of apoptotic cells, and increasing ROS generation | Activated JNK signalling pathway by increasing phosphorylation of JNK and p38 | [53] |
| Human colorectal cancer cells (HCT116) (TSP50 expressing, 5-flourouracil- resistant) | Augmented the antiproliferative effect of 5-FU to inhibit cellular viability, induce apoptosis by increasing activity of caspase-3 and -9, upregulating expression of the proapoptotic protein BAX and downregulating expressions of stem-cell associated proteins (c-Myc and OCT4) | Blocked the activated TSP50-mediated by NF-κB signaling pathway through downregulation of protein expressions of cyclin E, TSP50 and NF-κB | [55] |
| Human colon cancer cells (HT-29 and SW-460) | Inhibited cellular viability through downregulating mRNA expression of the proliferative factor Ki67 | Deactivated STAT3 signaling pathway by downregulating protein expression of STAT3 and its upstream protein JAK2 | [60] |
| Human colorectal cancer cells (HCT116) | Inhibiting cellular viability | Reversed the inflammatory conditions by downregulating mRNA expression of IL-6 and TNF-α | [59] |
| Human colorectal adenocarcinoma cells (HT-29) and human colorectal carcinoma cells (HCT116) | (1) Inhibited cellular viability; and (2) Hindered cells migration and invasion | Blocked ADRβ2/EMT by downregulating mRNA expression of ADRβ2 and protein expression of matrix metalloproteases (MMP-2 and MMP-9 N-cadherin) | [56] |
| Lewis lung carcinoma cells (LLC) | (1) Inhibited cellular viability; and (2) Hindered cells invasion and migration by downregulating protein expression of snail and upregulating protein expression of E-cadherin | Deactivated mTOR signaling pathway by inhibiting phosphorylation of mTOR and S6K1 | [71] |
| Non-small-cell lung cancer (H460) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G2/M phase by downregulating protein expression of cyclin D1 and CDK4; (3) Induced apoptosis by upregulating proapoptotic proteins (BAX and cleaved caspase-3) and downregulating expression of the antiapoptotic proteins BCL-2; (4) Inhibited cellular colonization; and (5) Hindered cells migration and invasion | Deactivated PI3K/Akt/mTOR signaling pathway by downregulating protein expression of Akt and mTOR, modulating EMT by upregulating E-cadherin with N-cadherin) and downregulating EMT promotion transcription factor (ZEB1) | [64] |
| Human gastric cancer 5-FU resistant cells (BGC-823/5-FU) | (1) Restored inhibitory effect of 5-FU on cellular viability, overcoming P-glycoprotein-mediated resistance by downregulating protein expression of P-glycoprotein as well as blocking P-glycoprotein efflux pump through increasing levels of accumulated intracellular Rh-123; (2) Augmented antiproliferative effect of 5-FU through inducing apoptosis (more increase in number of apoptotic cells); and (3) Arrested cell cycle growth at G1 phase | Deactivated Wnt/β-catenin signaling pathway by downregulating protein expression of β-catenin, mRNA expression of stem cell-associated genes (MDR1, CD44, ALDH1, OCT4, c-Myc), Wnt target genes (β-catenin, TCF4 and cyclinD1) and blocking complex formation of β-catenin/TCF4 | [73] |
| Human multiple myeloma cells (U266 and ARH-77) | (1) Inhibited cellular viability; and (2) Induced apoptosis by upregulating expression of proapoptotic proteins (cleaved caspase-3 and PARP) and downregulating expression of antiapoptotic proteins (BCL-2, BCL-xL, survivin, XIAP, cIAP-1 and cIAP-2) | Deactivated NF-κB signaling pathway by blocking phosphorylation of NF-κB p65, downregulating protein expression of Iκβα and IKKβ and products of NF-κB regulating-genes (ICAM-1, COX-2, VEGF) | [67] |
| Mouse leukemia cells (WEHI-3) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G0/G1 phase; and (3) Induced mitochondria-dependent apoptosis (early and late stage) by increasing accumulation of mitochondrial ROS, upregulating expression of proapoptotic proteins (BAX, cytochrome c, AIF, Endo G, Apaf-1 and cleaved PARP), downregulating expression of antiapoptotic proteins (BCL-2), upregulating mRNA expressions of DAP, TMBIM4 transmembrane, ATG5, and downregulating mRNA expression of DDIT3, DDIT4, BAG6, BCL-2 L13 and BRAT1 | Deactivated mitochondria-dependent and ER stress signaling pathways | [66] |
| Lymphoma cells (SUDHL-4 and OCI-Ly7) | Inhibited cellular viability | Deactivated mTOR signaling pathway by inhibiting phosphorylation of mTORC1 and its downstream substrate S6K1 and downregulating expression of raptor | [65] |
| Human prostate tumor cell line (PC-3) | (1) Inhibited cellular viability; and (2) Induced apoptosis by increasing DNA fragmentation | Deactivated NF-κB signaling pathway by downregulating mRNA expression of NF-κB1 | [50] |
| Human androgen independent prostate cancer cells (DU145) | (1) Inhibited cellular viability by down regulating expression of complex cell cycle progression proteins (cyclin D1/CDK4, cyclin E/CDK2), and down regulating mRNA expression of cyclin D1; (2) Induced apoptosis by upregulating expression of proapoptotic proteins (cleaved caspase 3 and cleaved PARP), downregulating expression of the antiapoptotic protein BCL-2 and upregulating protein expression of caspase-8 and 9; and (3) Hindered cells invasion and migration | Deactivated STAT3 signaling pathway by decreasing phosphorylation of STAT3, suppressing activity of STAT3 to bind DNA, nuclear pool depletion of STAT3, while downregulating expression of upstream STAT3 kinase (JAK2) and downregulating protein expression of oncogenes products (BCL-xL, BCL-2, XIAP, VEGF, COX-2, and MMP-9) | [49] |
| Human melanoma cell (A375) | (1) Inhibited cellular viability; (2) Induced apoptosis by upregulating expression of proapoptotic proteins (cleaved caspase-3 and cleaved PARP); and (3) Hindered cells invasion | No signaling pathway was reported | [18] |
| (1) Inhibited cellular viability; (2) Induced apoptosis by upregulating pro-apoptotic proteins expression (BAX, cleaved PARP, cleaved caspase 9, cleaved caspase 8) and downregulating expression of the antiapoptotic proteins BCL-2; and (3) Inhibited cells migration and invasion | Deactivated NF-κB signaling pathway by downregulating protein expression of p65 NF-κB | [70] | |
| Human epithelial ovarian cancer cells (SKOV3 and CoCl2-mimicked hypoxic cells) | (1) Inhibited cellular viability; and (2) Anti-angiogenesis and inhibited HIF 1 by downregulating mRNA and protein expressions of VEGF of HIF-1α and HIF-2α, respectively | Deactivated mTOR by downregulating protein expression of mTOR and S6K1 | [47] |
| Human epithelial ovarian cancer cells (SKOV3, cisplatin sensitive) | (1) Augmented the antiproliferative effect of cisplatin through inhibiting cellular viability, arresting cells growth at G2/M phase, and inducing apoptosis by downregulating protein expression of anti-apoptotic proteins (BCL-2, XIAP and survivin); and (2) Augmented antimetastatic effect of cisplatin through inhibiting cellular colonization | Deactivated mTOR signaling pathway by inhibiting phosphorylation of mTOR and its downstream target S6K | [72] |
| (1) Inhibited cellular viability; (2) Induced autophagy by increasing autophagosomes and upregulating protein expression of an autophagy marker LC3-II; and (3) Induced apoptosis by upregulating expression of proapoptotic proteins (cleaved PARP and cleaved caspase-3) | Deactivated mTOR signaling pathway by downregulating protein expression of raptor and decreasing phosphorylation level of S6K1 | [44] | |
| (1) Induced autophagy by evidenced by upregulating protein expression of autophagy markers (LC3-II and LAMP1); and (2) Metabolic programming by decreasing lactate secretion and inhibiting activity of hexokinase and lactate dehydrogenase | Deactivated mTOR signaling pathway by deactivating phosphorylation of mTOR and S6K1 | [45] | |
| Inhibited cellular viability | Deactivated mTOR signaling pathway by inhibiting phosphorylation of mTOR and S6K1, while downregulating protein expression of raptor | [47] | |
| Human epithelial ovarian cancer cells (SKOV3) | (1) Inhibited cellular viability; and (2) Induced autophagy by increasing number of autophagosomes and upregulation protein expression of LC3II (an autophagy marker) | Deactivation mTOR signaling pathway by inhibiting phosphorylation of mTOR and S6K1 through upregulating mRNA expression of DAP1 that negatively regulated autophagy | [41] |
| (1) Inhibited cellular viability; (2) Arrested cell cycle at G2/M phase by upregulating expressions of cyclin D1, cyclin B1 and p-histone H3 with downregulating expressions of cyclin A, p-cdc2 and Myt1; (3) Induced apoptosis (early and late) by downregulating expression of antiapoptotic proteins (MCL-1, BCL-2) and upregulating expressions of proapoptotic proteins (BAX, and cleaved caspase 3); and (4) Inhibited cellular colonization | Deactivated mTOR signaling pathway by downregulating protein expressions of mTOR, PRAS40, raptor and RagC, in association with deactivating NF-κB signaling pathway by downregulating protein expression of NF-κB, IKKα and IKKβ | [43] | |
| Paclitaxel-resistant ovarian cancer cells (SKOV3-Taxol) | Enhanced the antiproliferative effect of paclitaxel through enhancing inhibiting cellular viability, arresting cell cycle at G2/M phase, and inducing apoptosis by downregulating mRNA expression of MDR1 and protein expression of P-gp | Deactivated NF-κB signaling pathway by downregulating protein expression of p65 NF-κB | [40] |
| Ovarian cancer cells (SKOV3) with induced TAMs | (1) Inhibited cellular viability by inhibiting M-2 polarization of TAMs through downregulating protein expressions of pro-tumorigenic factors (MMP2 and MMP9), and mRNA expressions of IL-6 and VEGF-α; and (2) Hindered cells migration and invasion | Deactivation mTOR signaling pathway by inhibiting phosphorylation of mTOR and S6K1 and downregulating expression of raptor; deactivated STAT3 signaling pathway by inhibiting phosphorylation of STAT3 | [39] |
| Human ovarian cancer SKOV3 and A2780 cells | (1) Inhibited cellular viability of both cell lines; (2) Inducing oxidative stress through increasing liberation of cellular ROS (little stronger in SKOV3 than that in A2780); and (3) Hindered SKOV3 cells migration through inducing ROS liberation by partial abolishing the reactive oxygen scavenger NAC | Deactivated mTOR signalling pathways pathway by inhibiting phosphorylation of S6K1 at Thr389 and mTORC1 at Ser2448 with downregulating expression of raptor and p-ERK1/2 | [48] |
| Human ovarian SKOV3 and A2780 cells | (1) Inhibited cellular viability; (2) Induced apoptosis by reducing MMP (inducing mitochondrial damage) and downregulating mRNA expression and proteins of SREBP1, FASN, ACC and ACLY; (3) Inhibited activity of CPT-1 (i.e., inhibited lipogenesis); and (4) Inhibited cellular colonization | Deactivated mTOR signalling pathway via raptor by inhibiting phosphorylation of mTOR, S6K1, and 4E-BP1 and expression of raptor | [42] |
| Pancreas cancer cells (PANC-1 and SW1990) | (1) Augmented the antiproliferative effect of gemcitabine through inhibiting cellular viability, inducing apoptosis by upregulating expression of pro-apoptotic proteins (BAX and cleaved caspase 3), while downregulating expression of the anti-apoptotic protein BCL-2; and (2) Augmented inhibitory effect of gemcitabine on cellular colonization | Deactivated mTOR signaling pathway by inhibiting the phosphorylation of PI3K, AKT, and mTOR; modulated FOXO3a-FOXM1 axis by upregulating expression of FOXO3a and downregulating expression of FOXM1 | [63] |
| Human pancreatic cancer cell line (BxPC3). | (1) Inhibited cellular viability; (2) Induced DNA damaged by enhancing chromatin condensation, nuclear shrinkage and fragmentation though increasing formation of γ-H2AX (one of the histone family member X); (3) Inducing apoptosis by upregulating expression of the pro-apoptotic protein Bax and downregulating anti-apoptotic protein BCL-2, involving iintrinsic or extrinsic pathways as evidenced by increasing expression of cleaved caspase-3 and cytochrome C; and (4) Inhibited cellular colonization | No signaling pathway was reported | [62] |
| Human esophageal cancer cells (EC-9706 and TE10) | (1) Inhibited cellular viability by downregulating protein expression of the proliferative marker PCNA; (2) Induced apoptosis by increasing dense chromatin-containing cells, proportion of apoptotic cells (at early and late stages), upregulating pro-apoptotic proteins expression (BAD, BAX, cleaved PARP, and cleaved caspase-3) and downregulating expression of anti-the apoptotic protein BCL-2; (3) Inhibited cellular colonization; and (4) Hindered cells invasion and migration through inversing EMT by upregulating protein expression of E-cadherin and downregulating protein expression of N-cadherin, vimentin, snail, MMP2, MMP7, and MMP9 | Deactivated PI3K/AKT/mTOR signaling pathway by inhibiting phosphorylation and downregulation of the expression of PI3K and its downstream effector AKT | [30] |
| Human osteosarcoma cells (143B and MG63) | (1) Inhibited cellular viability; (2) Arrested cell cycle at G2/M phase by downregulating expression of cell cycle-related proteins (cyclin D1, c-Myc, and PCNA); (3) Induced apoptosis by upregulating expression of proapoptotic proteins (BAX, BAD, cleaved caspase 3, and cleaved PARP) and downregulating expression of the antiapoptotic protein BCL-2; and (4) Hindered cells invasion and migration by reversing EMT process through downregulating protein expressions of MMP-2, N-cadherin, snail and vimentin | Activated JNK signaling pathway by increasing phosphorylation of p38 MPAK and JNK | [31] |
| Human glioblastoma stem cells (CD133+ GSCs) | (1) Inhibited cellular viability by downregulating cell cycle regulator protein (cyclin D1), and VEGF; and (2) Induced apoptosis by upregulating protein expression of intrinsic apoptosis pathway (caspases 3, caspases 9, and PARP), and downregulating expression of antiapoptotic proteins (BCL-2, BCL-xL, MCL-1, surviving) | Deactivated STAT3 signaling pathway by decreasing phosphorylation level of STAT3, and downregulating expressions of STAT3 downstream proteins (BCL-xL, BCL-2, MCL-1, survivin, and VEGF) | [69] |
| Human bladder cancer cells (T24 and UM-UC-3) | (1) Iinhibited cellular viability; (2) Inducing apoptosis (no evidence); (3) Blocked glycolysis by decreasing glucose uptake, ATP generation and lactate production; and (4) Induced oxidative stress by increasing liberation of cellular ROS through upregulating Nrf2 and NQO1genes | Deactivated PI3K/AKT/mTOR pathway by reducing phosphorylation of PI3K, AKT and mTOR | [51] |
| Human bladder cancer cells (HT1376 and HTB-9) | (1) Inhibited cellular viability; (2) Arrested cell cycle growth by blocking transition of G0/G1 phase to S phase; and (3) Induced apoptosis by increasing cellular apoptotic rate and upregulating protein expression of ESR1 | Deactivated PI3K/AKT/mTOR signaling pathway by downregulation expression of ESR1 | [52] |
Table 2 The in vivo animal-based anticancer activity of cardamonin
| Cancer type | Model | Cancer induction | Dosage | Mechanism | Ref. |
| Liver | Athymic nude mice | HepG2-xenograft | Single oral daily of 25 mg/kg or 50 mg/kg for 24 days | Deactivated NF-κB signalling pathway by downregulating protein expression of PCNA, Ki-67 NF-κB-p65 and Ikkβ | [29] |
| Breast | BALB/c female mic | 4T1-pcDNA3-TSP50-luc -xenograft | Single IP dose of 100 μg/50 μL every-three-days for 15 days | Inhibited overexpression of endogenous TSP50 | [35] |
| Athymic nude mice | SUM190-xenograft | Single IP dose of 25 mg/kg every-other-day for 17 days with doxorubicin | Abolished doxorubicin-enriched CSCs through downregulating mRNA expression of CSCs-associated genes (ALDH1, SOX2, OCT4, NANOG) | [38] | |
| Athymic nude mice | SUM190 and SUM149-xenograft | Single IP dose of 30 mg/kg every-other-day for 20 days | Blocked TLR3 on the induced-tumorigenic cancer stem cells | [27] | |
| Female nude mice | MDA-MB-231 cells xenograft | Single daily IP dose of 3 mg/kg for 28 days | Suppressed HIF-1α mediated-cell metabolism | [32] | |
| Female nude mice | MDA-MB-231 cells xenograft | Single IP dose of 30 mg/kg every-other-day for 28 days | Inducing apoptosis by upregulating expression of p21, p27, and Bim, and increasing the level of cleaved caspase 3, while cyclin D1 level decreased; activated FOXO3a-JNK signalling pathway by upregulating expressions of JNK and FOXO3a | [33] | |
| BALB/c mic | 4T1-xenograft | Single oral daily dose of 5 mg/kg for 30 days | Retarded tumour growth (no further mechanistic information) | [37] | |
| Colorectal | BALB/c nude mice | HT29-xenograft | Single IP dose of 25 mg/kg every-two-days per week for 28 days | Hindered metastasis to lungs by downregulating expression of ADRB2 and matrix metalloproteases (MMP-2 and MMP-9 N-cadherin) | [56] |
| C57BL/6 mice | Azoxymethane-induced | Single oral daily dose of 40 mg/kg every-other-day for 90 days | Downregulated expression Ki67; deactivated STAT signaling pathway by downregulating protein expression of JAK2, STAT1, STAT3 and STAT5 | [60] | |
| C57Bl/6 mice | Azoxymethane-induced | Single oral dose of 10 mg/kg five days per week for 30 weeks | Stopped colorectal cancer cells proliferation by downregulating Ki-67 index; deactivated Wnt/β-catenin and NF-κB signaling pathways; modulated expression of microRNAs | [53] | |
| C57Bl/6 mice | Azoxymethane-induced | Single oral dose of 10 mg/kg either from day 1 to day 140 or from day 50 to day 140 | Reduced risk of colitis-induced colon cancer by reducing tumors number, increasing colon shrinkage and alleviating inflammatory insult; blocked accumulation of immune cells; deactivated NF-κB and iNOS signalling pathways; protected mice against inflammation-associated colitis by modulating expression of microRNAs | [59] | |
| Lung (LLC cell) | C57BL/6 mice | LLC-xenograft | Single daily IP dose of 3.5 mg/kg, 7 mg/kg, or 10.5 mg/kg for 20 days | Reduced tumour size; hindered metastasis through inhibiting lung colonization of LLC cells | [71] |
| Lung (NSCLC) | BALB/c nude mice | H460-xenograft | Single daily IP dose of 5 mg/kg for 14 days | Downregulated expression of Ki-67 in lungs, and deactivated PI3K/Akt/mTOR signalling pathway by downregulating protein expression of Akt and mTOR | [64] |
| Gastric (5-FU-resistan) | BALB/c nude mice | BGC-823/5-FU xenograft | Single IP dose of 25 mg/kg twice per week for 30 days | Superior retarding tumour growth (no further mechanistic information) | [73] |
| Esophageal cancer | Male nude mice | EC9706 xenograft | Single oral dose of 5 mg/kg, 15 mg/kg, or 25 mg/kg every-three-days for 26 days | Downregulated protein expression of PCNA, BCL-2, vimentin, PI3K, and Akt; deactivated mTOR signalling pathway | [30] |
| Pancreas | Athymic BALB/c nude mice | PANC-1 and SW1990 xenograft | Single daily IP dose of 5 μg/kg for 42 days | Modulating FOXO3a/FOXM1 axis pathway through upregulating expression of FOXO3a; deactivated FOXM1/AKT/mTOR signalling pathway by lowering phosphorylation of PI3K, AKT, and mTOR | [63] |
| Ovarian | Female BALB/c nude mic | SKOV3 or a mix of SKOV3 and M2 macrophages xenograft | Single daily IP dose of 5 mg/kg or 25 mg/kg for 15 days | Downregulated protein expression of Ki67, CD163 and CD206; deactivated mTOR signalling pathway | [39] |
| BALB/c nude mice | SKOV3 and PDC xenograft | Single daily IP dose of 20 mg/kg for 13 days | Induced apoptosis; deactivated mTOR and NF-κB signaling pathways | [43] | |
| Female BALB/c nude mice | SKOV3 xenograft | Single oral daily dose of 15 mg/kg or 30 mg/kg for 20 days | Downregulated expression of Ki-67, raptor and FASN, activated mTOR signaling pathway | [42] | |
| Leukemia | Male BALB/c mice | WEHI-3 xenograft | 5 mg/kg, IP, once every two days for 14 days and 42 days | Improved animal survival after 42 days, decreased population of CD3 (T cells), CD11b (monocytes) and macrophages-3 after 14 days, increased population of CD19 (B cells), and enhanced macrophages phagocytic ability | [68] |
| Lymphoma | Female SCID mice | SUDHL-4 cells expressing mutant; RRAGC xenograft | Single oral daily dose of 15 mg/kg for 30 days | Sensitized RagCT90N-mutant highly to cardamonin | [65] |
| Osteosarcoma | Female BALB/c nude mice | 143B xenograft | Single oral dose of 5 mg/kg, 15 mg/kg, or 25 mg/kg every-two-days for 20 days | Deactivated mTOR signalling pathway, inhibited level of PCNA, BCL-2 and vimentin, and increased phosphorylation level of p38 MAPK and JNK | [31] |
Table 3 Comparative analysis of cardamonin’s in vitro and in vivo anticancer activities across cancer types
| Cancer type | Cell studies | Animal studies | Future prospective | ||||
| Cells line | IC50 (μM) | Ref. | Induced cancer | Inhibition | Ref. | ||
| Liver | HepG2 cells | 17.1 | [61] | HepG2-xenograft | 65.2% | [29] | Promising |
| 53 | [35] | ||||||
| Breast | 4T1 Luc cells | 85.96 | [35] | 4T1 Luc-xenograft | Substantial (graphic presentation) | [35] | Not promising in vitro |
| MDA-MB-231 cells | 45.6 | [35] | |||||
| MDA-MB-231 cells | 25.5 | [32] | MDA-MB-231 xenograft | Mild (≤ 50%; graphic presentation) | [32] | Promising | |
| Lung | LCC cells | NR | [71] | LLC-xenograft | 84.3% | [71] | Needs in vitro testing |
| Esophageal | EC9706 cells | 8.7 | [30] | EC9706-xenograft | Substantial (graphic presentation) | [30] | Promising |
| Ovary | SKOV3 cells | 8.04 | [43] | SKOV3 and PDC xenograft | Substantial (graphic presentation) | [43] | Promising |
| PDC cells | 14.9 | ||||||
| SKOV3 cells | 23.4 | [42] | SKOV3-xenograft | Substantial (graphic presentation) | [42] | Promising | |
| A2780 cells | 28.8 | ||||||
- Citation: Badroon NA, Alsalahi A, Aljaberi MA, Abdul Majid N, Alshawsh MA. Cardamonin as a potential anticancer agent: Preclinical insights and clinical implications. World J Clin Oncol 2025; 16(11): 110911
- URL: https://www.wjgnet.com/2218-4333/full/v16/i11/110911.htm
- DOI: https://dx.doi.org/10.5306/wjco.v16.i11.110911
