6.1.1. Anti-Cancer Effects of Naturally Occurring Prenylated Chalcones
Licochalcone A (
2) is recognised for its potent anti-cancer properties across diverse cell types. It (
2) exerts its effects through multiple mechanisms, including inducing apoptosis, regulating the cell cycle to inhibit proliferation, suppressing invasion and metastasis, and inhibiting angiogenesis. It (
2) was shown to exert a cytostatic effect on HONE-1, NPC-39, and NPC-BM cells, promoting apoptosis via the mitochondrial pathway and the JNK/p38 signalling cascade at concentrations of 20–80 µM [
86]. Various mechanisms were postulated in the breast cancer cell line MCF-7, including the activation of LC3-II signalling, endogenous pathway-mediated apoptosis, and mitochondrial dysfunction, at concentrations between 10 and 50 µM [
87,
88,
89]. It (
2) also demonstrated strong activity on glioma cell lines, including U87, M059K, U-251, GBM8901, and GSC, at 5 to 40 µM. Mechanistically,
2 inhibited cell growth by inducing cell cycle arrest in the G0/G1 and G2/M phases, mitochondrial fragmentation and ADAM9 expression, and, at low concentrations (<12.5 µM), specifically induced cell death in glioma stem cells (GSCs), with almost all GSCs dying after 6 days of treatment, while both normal fibroblasts and neural stem cells were unaffected [
90,
91,
92]. Compound
2 demonstrated significant anti-lung cancer effects across various cell lines. It reversed lung injury induced by the tobacco carcinogen 4-(methyl-nitrosamino)-1-(3-pyridyl)-1-butanone (NNK) via modulation of the miR-144 and mitogen-activated protein kinase (MAPK) pathways at 10 µM, suggesting potential applications in both lung cancer and lung injury [
93]. It (
2) induced UPR, autophagy, and apoptosis in H292 lung cancer cells by upregulating miR-144-3p. At 10 μM,
2 increased miR-144-3p, which downregulated Nrf2 and promoted ER stress, enhancing apoptosis and growth inhibition. At 40 μM,
2 elevated the C/EBP homologous protein (CHOP) protein but failed to activate its pro-apoptotic targets, suggesting the inhibition of CHOP-dependent apoptosis [
94]. In A549 and H460 cells,
2 (20, 40, or 60 μM) blocked cell cycle progression at the G2/M phase and induced apoptosis [
95]. Similarly, in A549 and H1299 cells,
2 activated the CHOP pathway, contributing to its pro-apoptotic effects while not affecting human embryonic lung fibroblasts [
96]. It (
2) was investigated for its effects on malignant pleural mesothelioma, where it inhibited cell growth in MSTO-211H and H28 cells, with IC
50 values of ~26 and 30 µM, respectively, and suppressed Sp1 expression along with the downstream targets cyclin D1, Mcl-1, and survivin. Mechanistically,
2 activated the mitochondrial apoptotic pathway by altering the Bax/Bcl-xL ratio, inducing Bid-mediated mitochondrial membrane loss, caspase activation, and nuclear fragmentation. Flow cytometry confirmed annexin V/PI-positive apoptotic cells. These findings suggested that
2 induced apoptosis in MPM cells via Sp1 downregulation and mitochondrial pathway activation [
97]. In hepatoma (HepG2) cells, with IC
50 values of 65.96 μM (24 h) and 44.13 μM (48 h),
2 induced morphological changes, reactive oxygen species (ROS) generation, G2/M arrest, and apoptosis. It downregulated cell cycle genes (survivin, cyclin B1, and
CDK1) while upregulating Wee1, P21, cyclin D1, and JNK1 [
98,
99]. In HuH7 and HepG2 lines,
2 triggered autophagy through ULK1/Atg13 activation and ROS accumulation [
100]. Gastric cancer cells (MKN45, SGC7901, and GES-1) underwent growth inhibition via blockade of the Akt/HK2 axis [
101], while in BGC-823 cells, it paradoxically activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) [
102]. In squamous carcinoma (FaDu) cells, there was upregulation of the tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) through ERK [
103]. Compound
2 inhibited proliferation and induced apoptosis in T24 bladder cancer cells through mitochondrial and ER stress-related pathways. In the presence of
2, intracellular ROS and Ca
2+ levels increased, while the mitochondrial membrane potential was lowered, and Apaf-1, caspase-9, and caspase-3 were upregulated, indicating mitochondrial dysfunction. Calpain 2 and caspase-4 were also activated, linking apoptosis to ER stress. These results suggest that
2 triggers T24 cell death via combined mitochondrial and ER stress-mediated mechanisms [
104].
Recently,
2 exhibited anti-cancer activity against HCT-116, resulting in G0/G1 phase arrest, apoptosis, and high ROS generation, which were attenuated by the ROS inhibitor
N-acetyl-L-cysteine. The targeting of thioredoxin reductase 1 (TrxR1) in HCT-116 cells led to high ROS levels and apoptosis [
105]. Substrate
2 potently suppressed hypoxia-induced factor (HIF)-1α accumulation and the expression of HIF-1α target genes, including GLUT1 and PDK1 in HCT116 cells, and effectively inhibited ATP production [
106]. Substrate
2 inhibited non-small cell lung cancer (NSCLC) cell growth and induced apoptosis by destabilising proteins including survivin, XIAP, and RIP1 without altering their mRNA levels. It also activated ERK and p38 while suppressing JNK, leading to cytoprotective autophagy [
107].
It (
2) markedly decreased the viability and caused apoptosis in several endometrial cancer (EMC) cell lines (Hec1A (HTB-112), AN3CA (HTB-111), HEC59 (JCRB1120), and Ishikawa (JCRB1505)) and primary (EMC-7) cells. Additionally, it was discovered that
2 causes endoplasmic reticulum (ER) stress, which causes EMC lines to express more ER-related proteins (GRP78/PERK/IRE1α/CHOP). In human EMC cells treated with
2, suppression of GRP78 expression dramatically diminished the effects of
2, leading to the decreased production of proteins associated with ER and apoptosis, as well as ER stress-mediated cell death [
108]. Compound
2 also suppressed glioma growth, migration, and invasion by inducing mitochondrial dysfunction and ROS production, while ROS inhibition reversed these effects. In vivo, treatment with
2 suppressed glioma growth in nude mice in a dose-dependent manner: at 10 μM, the tumour volume and weight were reduced by about half compared to vehicle control, and at 20 μM, they were reduced by ~80%. Imaging and immunohistochemistry confirmed that
2 inhibited proliferation (Ki-67), reduced angiogenesis (CD-34), and promoted apoptosis (TUNEL), suggesting its anti-glioma activity involves apoptosis through ATM/ATR pathway activation and oxidative stress [
109]. In a 28-day study of uterine leiomyoma, BALB/c nude mice bearing ELT3 tumours were treated with
2 (10 or 20 mg/kg, orally). Treatment significantly suppressed tumour growth and weight in a dose-dependent manner without affecting body weight or causing toxicity to major organs. Histology and immunohistochemistry showed reduced Ki67 expression, while molecular analysis revealed the upregulation of p-JNK, p-NRF2, ER stress proteins (GRP78/CHOP), and apoptotic protein c-caspase-3. It activated the GRP78/IRE1α/ATF6/CHOP pathway, upregulated caspase-dependent proteins, and its effects were reversed by NAC or Z-VAD-FMK. Blood markers (GOT, GPT, BUN, and creatinine) remained unchanged, confirming safety. These results highlighted the potential as a therapeutic agent via the JNK/GRP78/NRF2 signalling axis [
110].
Various studies have explored the anti-tumour effects of xanthohumol (
3) against glioma. It (
3) upregulated miR-4725-3p in U87-MG and Hs-683 cells, leading to the downregulation of Stim1, a calcium sensor involved in invasion [
111]. In U87 cells,
3 reduced viability and induced apoptosis through caspase activation, mitochondrial dysfunction, ROS generation, and Bcl-2 family regulation. It (
3) upregulated miR-204-3p via the ERK/c-Fos pathway, and miR-204-3p overexpression enhanced glioma apoptosis. MiR-204-3p directly targets IGFBP2, inhibiting the IGFBP2/AKT/Bcl-2 pathway [
112]. In colorectal cancer cells, mechanistic experiments demonstrated that
3 triggered DNA damage, inducing the phosphorylation of ATM/ATR, γ-H2AX accumulation, and p53/p21 activation, leading to the S phase or G1 arrest. Synergistic potential may be indicated by the observation that sub-cytotoxic concentrations of
3 (2.5–10 µM) sensitised resistant SW480 cells to the active metabolite of irinotecan (SN38). The prenyl group of
3 was associated with the facilitation of nuclear uptake and DDR activation, allowing it to act as both a direct cytotoxic and chemosensitising adjuvant [
113]. In melanoma,
3 showed dose-dependent cytotoxicity and inhibited melanoma growth and metastasis. It reduced the proliferation, colony formation, and migration of Mel Ju and Mel Im cells at subtoxic concentrations (≤30 µM) but spared hepatocytes at up to 100 µM. In a murine model of hepatic metastasis, the continuous delivery of
3 at 10 mg/kg/day reduced hepatic B16-F10 metastases, with a lower Ki67 expression and greater necrosis within lesions. Again, lipophilicity and cell uptake was associated with prenylation. This observed selectivity of
3 in impairing proliferation and migration in melanoma cells led to
3 being touted as a promising candidate for targeting melanoma progression and liver metastasis [
114]. In canine haematological models,
3 and seven derivatives (natural and semisynthetic) were active against three cancer cell lines: CLBL-1 (B-cell lymphoma), CLB70 (B-cell leukaemia), and GL-1 (B-cell leukaemia). All compounds showed dose-dependent cytotoxicity (0.1–30 µM, 48 h), but the most potent were
3 (IC
50 = 1.3–6.3 µM), xanthohumol D (
148, IC
50 = 0.55–5.3 µM), and the 4′-
O-β-D-(4′′′-
O-methyl)-glucopyranoside of
3 (
149, IC
50 = 1.2–4.9 µM). All three compounds induced apoptosis, decreased mitochondrial potential, and increased ROS production (except in resistant GL-1 cells). Western blotting confirmed the downregulation of anti-apoptotic Bcl-2, particularly in CLBL-1 and CLB70. SAR analysis defined both the chalcone skeleton and uncyclised prenyl groups of
3,
148, and
149 as crucial for potency, that cyclisation of the prenyl unit reduced activity, and that glycosylation preserved cytotoxicity, while potentially improving solubility and bioavailability [
115].
![Ijms 26 09845 i001 Ijms 26 09845 i001]()
The cytotoxicity of
3 was also evaluated in human hepatocellular carcinoma (HepG2) cells. It (
3) inhibited HepG2 proliferation in a dose- and time-dependent manner, with IC
50 values of 68 µM (24 h) and 52 µM (48 h). Apoptosis assays showed that
3, at 25 µM, induced apoptosis in ~47% of HepG2 cells, increasing to >80% at 100 µM, mediated by caspase-3 activation and PARP cleavage. In contrast, primary rat hepatocytes were highly resistant: even at 100 µM
3, viability was >90% with no apoptosis. The chalcone scaffold and prenyl side chain were considered to enable selective apoptosis, likely through preferential uptake and mitochondrial disruption in HepG2 cells [
116].
In NSCLC cells,
3 was demonstrated to directly inhibit T-lymphokine-activated killer cell-originated protein kinase (TOPK), a serine/threonine kinase implicated in tumour proliferation and metastasis. At 20 µM,
3 reduced the viability of A549 and HCC827 cells by >80% after 72 h but not normal lung (MRC-5) fibroblasts. It induced G0/G1 cell cycle arrest, enhanced apoptosis, and suppressed migration and invasion. In an A549 xenograft model, a daily i.p. injection of
3 at 25 mg/kg reduced the tumour weight by 50% without causing hepatotoxicity or nephrotoxicity. Mechanistically,
3 was demonstrated to directly bind the ATP-binding pocket of TOPK (KD = 2.9 µM), inhibiting phosphorylation of histone H3 and Akt, both in vitro and in xenografts. The prenyl chain at C-3′ and the methoxy group at C-6′ were crucial for potency, with
3 (IC
50 = 11.6 µM) more active than xanthohumol D (
148) (28 µM) and desmethylxanthohumol (
62) (55 µM), correlating with stronger TOPK inhibition. Prenylation and methoxylation thus conferred superior TOPK binding and anti-cancer activity and supported
3 as a promising TOPK inhibitor [
117].
An optimised solid lipid nanoparticle formulation of
3 has been investigated to overcome its limited oral bioavailability and sub-optimal pharmacokinetics. Compared to
3, the formulation showed a 4.7-fold increase in systemic exposure, a 6.5-fold longer half-life, and nearly 4800% relative bioavailability in vivo. In PC-3 prostate cancer cells, encapsulated
3 produced sustained cytotoxicity, with enhanced inhibition compared to
3 and with effects comparable to the control, camptothecin. The results highlighted that while the prenyl side chain of
3 confers lipophilicity, it also limits solubility, and that formulation choice can overcome absorption barriers, enabling improved delivery to target cells and facilitating translational studies [
118].
Isobavachalcone (
80) was identified as a potent and non-toxic inducer of autophagic flux in human and murine cells. It (
80) inhibited AKT phosphorylation and downstream mTORC1 activity, leading to activation of the pro-autophagic transcription factors TFEB and TFE3. Also noted were the induction of ER stress (PERK/eIF2α phosphorylation, CHOP and ATF6 activation, and XBP1 splicing), showing crosstalk between the unfolded protein response and autophagy. In vivo,
80 injections reduced AKT/mTOR/S6K signalling and increased LC3-II, a marker of autophagy. It (
80) also enhanced immunogenic cell death (ICD): in combination with low-dose mitoxantrone or oxaliplatin,
80 increased the ATP release from cells, promoted dendritic/T-cell infiltration, decreased Tregs, increased the CD8
+/Treg ratio, and lowered the PD-1 expression on cytotoxic T-lymphocytes. These effects were lost in the presence of constitutively active AKT, Atg5 knockout, or PERK deficiency, confirming mechanistic dependence. Again, the prenylation of
80 and its impact on lipophilicity was held to enable the dual targeting of AKT/mTOR and ER stress pathways. Substrate
79 alone had little cytotoxicity but showed synergy with ICD-inducing chemotherapy, boosting anti-cancer immune responses [
119].
Substrate
80 was investigated for its dual role in modulating multidrug resistance (MDR). Substrate
80 was largely non-toxic to HT29 colorectal adenocarcinoma and the doxorubicin-resistant variant HT29/Dx up to 40 µM, though it showed moderate cytotoxicity to kidney (MDCK) cells (IC
50~26.6 µM). As
80 stimulated the growth of ABCB1-overexpressing MDCK-MDR1 cells at 10–20 µM, an effect abolished by verapamil,
80 was considered both a substrate and competitive inhibitor of ABCB1. Fluorescence assays confirmed that
80 increased doxorubicin and rhodamine 123 accumulation in resistant cells but did not fully reverse resistance. Differential scanning calorimetry showed that
80 intercalated into phosphatidylcholine bilayers, lowering transition temperatures and enthalpy, broadening phase transitions, and identifying it as an effective membrane perturbing agent. Molecular modelling indicated amphiphilicity (logP = 4.19) and favourable bilayer partitioning (ΔG = −4.81 kcal/mol), supporting both transporter binding and lipid interactions. Prenylation at C-6 was noted to affect the amphiphilic balance, enabling
80 to act on both membrane bilayers and ABCB1 transporters. Overall,
80 was considered to interfere with MDR through both membrane-disruptive and transporter-inhibitory mechanisms [
120].
Substrate
80 has also been evaluated against the CNS cancer, glioblastoma (GBM), using cell and xenograft models. Substrate
80 inhibited the proliferation, migration, and invasion of U87MG (p53 wild-type) and U251 (p53 mutant) cells, with IC
50 values of 4.4 and 1.9 µM at 48 h, respectively, compared to the reference therapy temozolomide (224–473 µM). In subcutaneous and orthotopic GBM xenografts, daily
80 (20–40mg/kg p.o.) suppressed tumour growth without causing systemic toxicity, decreased angiogenesis (CD31 ↓), and increased apoptosis (cleaved caspase-3 ↑). Substrate
80 was shown to bind directly to ERα (ESR1), to suppress NLRP3 inflammasome transcription, alleviate pyroptosis (NLRP3/ASC/GSDMD/IL-1β/IL-18 ↓), and promote mitochondria-dependent apoptosis via Bcl-2/Bax modulation and caspase-3 activation. Prenylation was associated with blood–brain barrier (BBB) permeability and engagement with ERα and to underpin both anti-inflammatory (pyroptosis inhibition) and pro-apoptotic activity. Prenylation of
80 was considered to optimise the compound for CNS druggability and to facilitate the dual modulation of pyroptosis/apoptosis in GBM [
121]. Both
80 and bavachalcone (
83) were also evaluated using the human leukaemia K562 cell line. SAR analysis indicated that prenylation at the 5 position markedly improved their cytotoxic effects. Substrate
83 exhibited stronger activity, with an IC
50 of 2.7 μM. Morphological observations and annexin V/PI staining revealed that
80 and
83 inhibited K562 cell proliferation, primarily by inducing apoptosis [
61].
Paratocarpin E (
108) has anti-cancer potential against various cancer cell lines, including breast, leukaemia, and kidney cancer cells. Substrate
108 exhibited significant cytotoxicity, notably against MCF-7 breast cancer cells, with an IC
50 of 19.6 μM. Treatment with
108 induced classical apoptotic features in MCF-7 cells by increasing the activation of caspase-8, caspase-9, and inducing PARP cleavage. Mechanistically, it induced apoptosis by activating the p38 and JNK MAPK signalling pathways and suppressing ERK signalling [
122].
Isocordoin (
150) has been evaluated for cytotoxic activity against colorectal (HT-29), breast (MCF-7), and prostate (PC-3) cancer cells, along with non-malignant colon cells (CCD841). Substrate
150 had the greatest toxicity against PC-3 cells (IC
50 = 15.2 µM, SI = 5.2), followed by MCF-7 (21.1 µM, SI = 3.7) and HT-29 (27.2 µM, SI = 2.9). Unlike daunorubicin, which lacked selectivity,
150 displayed moderate potency with improved safety. Mechanistic studies in HT-29 and MCF-7 cells revealed mitochondrial dysfunction (loss of ΔΨ m), ROS modulation, and caspase-3 activation, consistent with the induction of apoptosis. Docking studies showed that
150 bound to caspase-3 (ΔG = −6.13 kcal/mol, Ki = 32 µM), supported by H-bonding (Trp214, Phe250) and π-interactions (Phe256, Trp206, and Phe247). SAR analysis attributed the increased lipophilicity and caspase-3 binding affinity to the isoprenyl side chain, making
150 more active than its non-prenylated analogue (2′,4′-dihydroxychalcone, IC
50 > 100 µM). The α,β-unsaturated carbonyl and hydroxylation pattern were considered hallmarks of apoptotic activity. In silico pharmacokinetic predictions of
150 were favourable. Overall,
150 was held as a promising prenylated chalcone with selective cytotoxicity against prostate and breast cancer, mediated by caspase-dependent apoptosis and enhanced by prenylation-driven lipophilicity [
123].
![Ijms 26 09845 i002 Ijms 26 09845 i002]()
Kanzonol C (
151) and the related licoagrochalcone A 4′-methylether (
11) were evaluated for antiproliferative activity, with
151 more active against MDA-MB468 triple-negative breast cancer cells (IC
50 = 5.97 µM) and MCF-7 breast cancer cells (IC
50 = 10.10 µM), exceeding that of cisplatin in MCF-7 cells (cisplatin IC
50 = 21.54 µM). Synthetic derivatives of
151, namely hexahydro-kanzonol (
152) and triacetyl-kanzonol (
153), were less active with IC
50s > 25 µM, emphasising the importance of unsaturation within the prenyl groups and unmodified phenolic hydroxy groups for activity. Isobavachalcone (
80) and
11 were also weaker (IC
50s > 25–50 µM). As
151 was non-toxic to normal liver HepaRG cells up to 50 µM, this suggested a good safety margin [
36].
6.1.2. Anti-Cancer Effects of Synthesised Prenylated Chalcones
In addition to the native natural products discussed in
Section 5.1.1, many derivatives of these lead compounds have been synthesised and evaluated for anti-cancer activity. Imidazole-modified chalcones based on
2 were designed to improve potency and selectivity against prostate cancer cells. Substrate
2 itself inhibited proliferation across both androgen receptor (AR)-positive (LNCaP, 22Rv1) and AR-negative (PC-3, DU145) cell lines, with IC
50 values of 15.7–23.3 µM. Among the prepared derivatives, compounds
154,
155,
156, and
157 retained their potency, especially in AR-positive lines, suggesting an increased selectivity toward AR signalling-dependent cells. Imidazole-modified chalcones
158 and
159 were the most potent, with IC
50 = 9.4–9.8 µM in LNCaP and ~28 µM in 22Rv1 compared to the standard enzalutamide (IC
50 = 21.7 µM in LNCaP, 67.5 µM in 22Rv1). Bulky isobutyl or pentyl groups on the imidazole moiety led to enhanced AR-positive selectivity, while shorter alkyl chains reduced activity. Key concepts from the study were that the prenyl group was crucial for lipophilicity and uptake, that imidazole substitution enhanced AR-positive selectivity, and that bulky alkyl substitutions favoured potency against LNCaP/22Rv1 but not AR-null PC-3/DU145. The imidazole-modified analogues of
2 were considered to be selective anti-prostate cancer agents with potential advantages over existing therapies [
124].
![Ijms 26 09845 i004 Ijms 26 09845 i004]()
A comparison of
3 with its semisynthetic aurone derivative (
Z)-6,4′-dihydroxy-4-methoxy-7-prenylaurone (
160) was undertaken using ten human cancer cell lines (breast, colon, prostate, lung, and leukaemia) and two normal lines (human endothelial HLMEC, murine fibroblasts BALB/3T3). Both
3 and
160 exhibited potent-to-moderate antiproliferative activity (IC
50 ≈ 7–20 µM in breast cancers, leukaemia, and doxorubicin-resistant colon LoVo/Dx; weaker in HT-29 colon cells). Aurone
160 showed a higher selectivity than
3, with SI values up to 7.09 in LoVo/Dx and ~5.5 in breast cancer lines, while cisplatin displayed poor selectivity. Mechanistic interpretation highlighted that prenylation drives potency, while the aurone led to increased selectivity and reduced cytotoxicity 1.5–2.3-fold toward normal cells. Also, unlike
3,
160 cannot be metabolised into 8-prenylnaringenin, a potent phytoestrogen that may promote estrogen-dependent cancers, suggesting improved safety. Thus, while the prenyl group is essential for activity, aurone cyclization reduces the off-target estrogenic risk while maintaining antiproliferative potency, making aurone derivatives potentially more selective anti-cancer leads [
125].
![Ijms 26 09845 i005 Ijms 26 09845 i005]()
Many analogues of
3 have been synthesised to evaluate their anti-angiogenic activity in vitro. These compounds significantly inhibited several angiogenesis-related functions of human umbilical vein endothelial cells (HUVECs), including proliferation, adhesion, migration, invasion, and tube formation at 10 µM. Notably,
161, a fluorinated analogue with a
p-methoxy group on the B-ring, showed the strongest activity [
126].
![Ijms 26 09845 i006 Ijms 26 09845 i006]()
The novel 3′,5′-diprenylated pyridyl chalcone (
162) demonstrated potent anti-cancer activity against prostate cancer PC3 cells. In vitro,
162 inhibited proliferation, with IC
50 values of 4.56, 4.67, and 3.55 μM at 24, 48, and 72 h, respectively, showing a stronger selectivity for PC3 over DU145 and normal RWPE-1 cells. Substrate
162 caused cell cycle arrest at the sub-G1 phase (53.5% cells at 8 μM) and triggered programmed cell death via dual pathways: caspase-dependent apoptosis and gasdermin E-mediated pyroptosis. The upregulation of PKCδ and activation of the JNK pathway were observed, leading to Bax upregulation, caspase-3 cleavage, PARP cleavage, IL-6/IL-1β release, and gasdermin E-N formation. PKCδ knockdown or JNK inhibition suppressed both apoptosis and pyroptosis, confirming pathway dependence. In vivo,
162 (30–60 mg/kg i.p.) significantly suppressed PC3 xenograft growth without toxicity, upregulating PKCδ and IL-6 while reducing the proliferating cell nuclear antigen (PCNA) and Bcl-2 expression. The diprenylation pattern of
162 at the 3′ and 5′ positions was associated with enhanced potency, lipophilicity, and pathway activation compared to non-prenylated chalcones, enabling a dual apoptotic/pyroptotic mechanism. Overall,
162 was considered a promising lead compound, exploiting apoptosis–pyroptosis crosstalk to overcome resistance in prostate cancer [
127].
![Ijms 26 09845 i007 Ijms 26 09845 i007]()
The same group later prepared an additional series of thirty-seven derivatives. Among these,
163, bearing a diethylaminoethoxy side chain, showed the strongest activity. It inhibited the proliferation of both LNCaP prostate cancer cells (IC
50 = 2.33 µM) and K562 leukaemia cells (IC
50 = 2.38 µM) comparably to doxorubicin (1.36–2.17 µM). Also,
163 displayed a slightly better selectivity index (SI) of 1.97 than doxorubicin (SI = 1.74), indicating a relatively lower toxicity toward normal LX-2 liver cells. Mechanistic studies showed that
163 induced apoptosis and necrosis in both cell lines, caused cell cycle arrest (S phase in LNCaP, G2/M in K562), and directly targeted the PI3K/AKT pathway, downregulating pPI3K and pAKT (Ser473) levels. Docking and thermal shift assays confirmed AKT1 as a direct target, suggesting that prenylation coupled with the introduction of a diethylaminoethoxy chain at C-4 optimised the binding affinity and potency [
128].
![Ijms 26 09845 i008 Ijms 26 09845 i008]()
Seventeen isobavachalcone (
80) derivatives were synthesised and tested for cytotoxicity against three human lung cancer cell lines. Among them, compound
164 showed the strongest activity, particularly against H1975 and A549 cells, with IC
50 values of 4.35 and 14.21 μM, respectively. Compound
164 induced apoptosis through increasing the Bax/Bcl-2 ratio, elevating cytochrome c, downregulating Akt, and activating caspase-9 and -3. It triggered necroptosis by upregulating receptor-interacting protein kinase 3 (RIP3) and the mixed lineage kinase domain-like protein (MLKL). Additionally, it caused mitochondrial dysfunction, reduced ATP levels, and led to excessive ROS accumulation [
129].
![Ijms 26 09845 i009 Ijms 26 09845 i009]()
Nine novel prenylated chalcones were synthesised and screened against castration-resistant prostate cancer lines DU145 and PC3. The most active compounds were
165 and
166. Both displayed potent cytotoxic activity, with IC
50 values of 54.96 µM (PC3) and 88.73 µM (DU145) for
165 and 57.22 µM (PC3) and 61.71 µM (DU145) for
166, with favourable selectivity. Mechanistic studies showed that both induced apoptosis via mitochondrial membrane depolarization, phosphatidylserine externalisation, and cytoskeleton disruption. SAR analysis emphasised that halogen substitution (B-ring
meta-F in
165, A-ring
ortho-Cl in
166), combined with
O-prenylation, significantly improved potency compared to other analogues, highlighting the impact of both prenylation and halogen positioning. Both
165 and
166 were considered promising prenylated scaffolds against resistant prostate cancers [
130].
![Ijms 26 09845 i010 Ijms 26 09845 i010]()
O-prenylated chalcone
167 was found to arrest lung cancer (NCI-H460) cells during mitosis, causing spindle collapse and chromosome misalignment, which activated the spindle assembly checkpoint, ultimately leading to cell death [
131]. In an attempt to investigate the mechanism underlying the activity of
167, yeast growth-inhibitory and p53 MDM2 transactivation assays were used to evaluate the compound in HCT116 human colon adenocarcinoma cells. The potency of
167 was ascribed to activation of the p53 pathway, leading to cell cycle arrest and mitochondria-dependent apoptosis [
132]. Based on the promising results with
167, a library of related
O-and/or
C-prenylated chalcones was synthesised, alongside the evaluation of their cytotoxic effects on human colon cancer HCT116 cells. Among the compounds,
168,
O-prenylated and with a bromo at R
2, was the most potent, again highlighting the potential positive impact of halogenation and showing parallels with compound
165 [
133].
![Ijms 26 09845 i011 Ijms 26 09845 i011]()
Two synthetic prenylated chalcones,
169 and
170, were evaluated for their effects on prostate cancer (PC3) cells through the modulation of Bloom (BLM) helicase, a key enzyme in DNA repair and genomic stability. Both compounds suppressed cell proliferation, induced apoptosis, and caused cell cycle arrest at the G2/M phase, but
169 was more potent with an IC
50 of 0.375 µM, nearly ten times stronger than
170 (IC
50 = 3.86 µM). Mechanistic assays revealed that both compounds selectively decreased BLM helicase protein expression without altering its mRNA levels, suggesting post-transcriptional regulation at the translation level. In enzymatic assays,
169 and
170 inhibited the ATPase and DNA-unwinding activities of purified BLM helicase while sparing other helicases, demonstrating high target selectivity. In addition,
169 showed stronger inhibition of helicase ATPase activity than
170, correlating with its superior cytotoxicity. These results were considered to identify compounds with a novel anti-cancer mechanism distinct from traditional apoptotic or ROS-based pathways [
134].
![Ijms 26 09845 i012 Ijms 26 09845 i012]()
A series of eleven novel
O-prenylchalcones were tested against gastric cancer (AGS) cells, with most showing a dose-dependent reduction in viability and with IC
50s ranging from 26 to 60 µM. Among them,
171,
172, and
173 were the most potent, with IC
50 values of 49.15, 49.21, and 31.9 µM, respectively. In addition to suppressing proliferation, the compounds also triggered apoptosis, with late apoptosis reaching 28.4% for
171. All three compounds induced caspase-3/7 activation, loss of mitochondrial membrane potential, and ROS accumulation, implicating the intrinsic apoptotic pathway.
O-prenylation on ring B, combined with electron-withdrawing substituents (F, NO
2, and Cl) on ring A were noted to enhance cytotoxicity, with the
ortho (2-Cl, 2-NO
2) or
meta positions (3-F) being particularly important. In silico docking predicted that these chalcones interact strongly (−7 to −8.8 kcal/mol) with gastric cancer oncoproteins including MMP11, CDC6, and especially HOXA1, a transcription factor linked to poor prognosis, through H-bonding (TRP383, LYS392) and π–stacking interactions. These observations suggest HOXA1 as a plausible target of prenylated chalcones.
O-prenylchalcones featuring electron-withdrawing groups on the B-ring may thus act as mitochondrial disruptors and apoptosis inducers in gastric cancer cells [
135].
![Ijms 26 09845 i013 Ijms 26 09845 i013]()
Two series (either
C- or
O-prenylated) trimethoxychalcones featuring Mannich base modifications were based on
3. Screening against four cancer cell lines, Aspc-1 (pancreatic), SUN-5 (gastric), HepG2 (hepatocellular carcinoma), and HCT-116 (colon) showed that several outperformed
3. The most active,
174 and
175, pyrrolidinomethyl and piperidinomethyl
O-prenylated compounds, had IC
50 values of 2.52–5.37 μM across tested cell lines compared to
3 (IC
50~9–16 μM). Substrate
175 in particular was broadly active (IC
50 2.54–10.49 μM). Introducing polar aminomethyl groups at prenylated sites thus enhanced solubility and potency, overcoming some of the bioavailability limitations of
3.
O-prenylated chalcones bearing cyclic amine Mannich bases were more potent than their
C-prenylated analogues. The results confirmed the Mannich base approach as a viable strategy to optimise prenylated chalcones as anti-cancer lead compounds [
136].
![Ijms 26 09845 i014 Ijms 26 09845 i014]()
The following SAR map illustrates the structural determinants of prenylated chalcones influencing their cytotoxicity against various cancer cell lines, as described.