Abbreviation
ACE2:
angiotensin-converting enzyme 2
CSCs:
cancer stem cells
ECM:
extracellular matrix
EMT:
epithelial-mesenchymal transition
G6PD:
glucose-6-phosphate dehydrogenase
H. pylori:
Helicobacter pylori
HIV:
human immunodeficiency virus
IRAK3:
interleukin-1 receptor-associated kinase 3
MMP-9:
matrix metalloproteinase 9
mTOR:
mammalian target of rapamycin
NF-κB:
nuclear transcription factor κB
NMP-diepoxyovatodiolide:
N-methylpiperazine-diepoxyovatodiolide
PPP:
pentose phosphate pathway
SARS-CoV-2:
severe acute respiratory syndrome coronavirus 2
STAT:
signal transducer and activator of transcription
T1/2
:
half-life period
TCM:
traditional Chinese medicine
TGF-β
:
transforming growth factor-β
TMPRSS2:
transmembrane protease serine 2
TNFRSF12A:
TNF receptor superfamily member 12A
Introduction
Ovatodiolide ([Fig. 1]) is a macrocyclic diterpenoid that is extracted from Anisomeles indica (L.) Kuntze (Labiatae). The plant is a traditional Chinese medicine (TCM) and was
mainly used for the treatment of allergy, dermatoses, and gastrointestinal disease
[1]. Ovatodiolide is the major bioactive component of Anisomeles indica, which was first reported in 1969 [2]. After that, researchers endeavored to isolate, purify, and elucidate the stereo-structure
of ovatodiolide while also conducting preliminary investigations into its biological
activity [3], [4]. Ovatodiolide is a cembrane-type diterpenoid, which has a distinctive ring system
consisting of 5/14/5 rings, featuring a butenolide moiety and a trans α-methylene-γ-lactone ([Fig. 1]). In 2019, Xiang and colleagues [5] reported that they successfully achieved the efficient chemical synthesis of the
ovatodiolide skeleton for the first time, thereby confirming the absolute stereo configuration
of compounds including ovatodiolide, ent-ovatodiolide, and 4,5-epoxy-ovatodiolide.
They developed tandem reactions consist of six steps for ovatodiolide synthesis, including
a series of ring-opening metathesis and ring-closing metathesis. This method possessed
stereoselectivity and allowed for further structural modifications of ovatodiolide.
Fig. 1 Chemical structure of ovatodiolide.
The half-life period (T1/2) of ovatodiolide incubated with human liver microsomes was determined to be only
0.24 h [6]. This may be attributed to the presence of multiple allylic sites in the ovatodiolide
structure that are readily oxidized under the catalysis of cytochrome P450 enzymes
in human liver microsomes, leading to the rapid metabolism. To enhance metabolic stability,
two double bonds were epoxidized to shield these allylic sites, forming a prodrug
N-methylpiperazine-diepoxyovatodiolide (NMP-diepoxyovatodiolide). This modification
significantly extended the T1/2 to 5.12 h [6]. Furthermore, in vivo studies demonstrated liver accumulation of NMP-diepoxyovatodiolide, suggesting its
suitability for further investigation in liver diseases [6].
Regarding the toxicity of ovatodiolide, a study in rats demonstrated no adverse reactions
after 28 days of once daily gastric gavage administration at doses of 10, 25, and
50 mg/kg or even after a single acute dose of up to 1000 mg/kg [7]. In vitro studies comparing ovatodiolideʼs effects on multiple cervical cancer cell lines versus
normal cervical cells found its growth inhibitory effect on normal cells to be significantly
less pronounced than on cancer cells [8]. A similar selective cytotoxicity was observed in liver cancer models, where ovatodiolide
exhibited markedly reduced toxicity toward the normal human liver cell line THLE-2
[9]. These findings indicate a high safety profile for ovatodiolide and demonstrate
its potential for drug development with a wide therapeutic window.
The objective of this review is to critically evaluate the pharmacological activities
of ovatodiolide and elucidate its underlying mechanisms of action, thereby providing
robust evidence to support its potential clinical applications. Relevant literature
was gathered through online scientific databases such as PubMed and Chinese CNKI databases.
The following search terms were used: ovatodiolide, Anisomeles indica, Labiatae, antitumor, anti-fibrosis, antimicrobial, anti-inflammation, and the Boolean
operators “AND” and “PLUS”.
The Pharmacological Activities of Ovatodiolide
Antitumor effect
Extensive in vitro and in vivo studies have demonstrated that ovatodiolide exhibits broad-spectrum antitumor effects
across diverse cancer types, including hepatocellular carcinoma, colon cancer, nasopharyngeal
cancer, bladder cancer, glioblastoma, and breast cancer [9], [10], [11], [12], [13], [14], [15]. The compound exerts its anticancer activity by suppressing cellular proliferation,
invasion, and migration, while simultaneously inducing apoptosis [9], [10]. Furthermore, ovatodiolide sensitizes cancer cells to a variety of chemotherapeutic
agents as well as radiotherapy [9], [16]. In vitro studies demonstrated that ovatodiolide exhibits
significant cytotoxicity against most tumor cell lines at concentrations ranging from
approximately 2.5 to 20 µM, with treatment durations of 24 – 48 h [9], [10], [11], [12], [13], [14], [15]. This cytotoxic activity exhibited both concentration- and time-dependent effects.
The antitumor mechanisms of ovatodiolide are multifaceted, as illustrated in [Fig. 2].
Fig. 2 Signaling pathways modulated by ovatodiolide in cancer therapy. Created in BioRender.
Li, J. (2025) https://BioRender.com/d73f065. [rerif]
Notably, ovatodiolide exhibits selective inhibitory effects against cancer stem cells
(CSCs). CSCs–a minor subpopulation within tumors–possess unlimited self-renewal capacity
and typically remain quiescent until activated by specific stimuli [17]. Critically, CSCs demonstrate resistance to radiotherapy and chemotherapy, drive
metastasis via epithelial-mesenchymal transition (EMT) and immune evasion, and constitute
a primary factor in tumor initiation, therapeutic resistance, recurrence, and treatment
failure [18], [19]. Studies have confirmed that low concentrations of ovatodiolide exert potent cytotoxicity
against CSCs in diverse malignancies, including endometrial cancer, hepatocellular
carcinoma, glioblastoma, breast cancer, oral cancer, nasopharyngeal carcinoma, and
colorectal cancer [11], [20], [21], [22], [23]. This evidence suggests that ovatodiolide may target and eliminate CSCs, thereby
addressing a fundamental mechanism of cancer persistence.
Ovatodiolide may suppress cancer cell metastasis [9], [11], [14], [24]. Matrix metalloproteinase 9 (MMP-9), a critical mediator of extracellular matrix
(ECM) degradation and tumor cell migration, appears to be suppressed by ovatodiolide
[24]. Research suggests this occurs through inhibition of nuclear factor κB (NF-κB) signaling, thereby reducing MMP-9 expression and subsequent ECM degradation [24], [25]. Furthermore, studies demonstrate that ovatodiolide effectively inhibits the expression
and phosphorylation of β-catenin in multiple cancer types [11], [22], [26], [27]. It promotes β-catenin destabilization and disrupts its
interaction with transcription factor 4 [28]. Consequently, this dual action impedes downstream signal transduction, thereby
inhibiting cancer cell viability and suppressing migration and invasion. However,
in vivo evidence supporting the inhibitory effect of ovatodiolide on tumor metastasis is
currently lacking.
The signal transducer and activator of transcription (STAT) protein family is a vital
group of signal transduction factors, and their aberrant activation is closely associated
with tumor development. Among the STAT family pathways, the JAK2/STAT3 signaling pathway
is particularly well-studied [29]. STAT3 serves as a pharmacological target for diverse small molecule anticancer
agents, including ovatodiolide. Ovatodiolide can effectively inhibit the phosphorylation
of STAT3, ERK1/2, p38, and AKT and down-regulate the presence of exosomes containing
oncomiR-1246 and oncomiR-21, thereby dampening the downstream signaling pathways and
contributing to its anticancer effects [12], [21], [22], [25]. In CSCs of chronic myeloid leukemia, ovatodiolide can suppress the expression of
STAT5, concomitant with inhibition of the PI3K/AKT/mammalian
target of the rapamycin (mTOR) signaling pathway [30].
mTOR is a serine/threonine kinase that actively participates in crucial biological
processes, including gene transcription, protein translation, and ribosome synthesis,
and exerts significant influence on cellular growth, apoptosis, autophagy, and metabolism
[31]. In human liver cancer cell lines (Huh7 and Mahlavu), ovatodiolide suppresses colony
formation and proliferation through inhibition of both the ERK1/2 and Akt/mTOR signaling
pathways [9]. Furthermore, by targeting mTOR–a well-established autophagy suppressor–ovatodiolide
activates autophagy and triggers autophagy-mediated cell death [32].
Substantial evidence has demonstrated that ovatodiolide significantly enhances the
anticancer efficacy of multiple chemotherapeutic agents, including cisplatin, 5-fluorouracil,
temozolomide, sorafenib, sunitinib, and imatinib [11], [12], [13], [22], [30], [33]. Moreover, ovatodiolide decreases exosomal levels of miR-21 – 5 p, STAT3, and mTOR
in oral squamous cell carcinoma, thereby resensitizing CSCs to cisplatin and suppressing
tumorigenicity [26]. These synergistic interactions highlight ovatodiolideʼs potential to reduce therapeutic
toxicity and overcome drug resistance in oncology regimens.
Collectively, these findings position ovatodiolide as a promising candidate for development
into either an antitumor agent or a therapeutic adjuvant. Its multifaceted activity–simultaneously
targeting CSCs, suppressing critical signaling pathways governing cell proliferation,
survival, and metastasis, and potentiating conventional anticancer therapies–highlights
its unique mechanistic value in oncotherapy. Advancing translational research on ovatodiolide
could yield novel strategies to address the unmet clinical needs in cancer treatment.
Anti-fibrotic activity
Ovatodiolide and its semi-synthetic derivative NMP-diepoxyovatodiolide have demonstrated
significant inhibitory effects on renal, pulmonary, and peritoneal fibrosis ([Fig. 3]) [34], [35], [36]. Multiple studies indicate that the transforming growth factor-β (TGF-β) signaling pathway plays a pivotal role not only in tumor development but also in
fibrogenesis [37]. Overexpression of TGF-β induces EMT, ECM deposition, and the generation of cancer-associated fibroblasts,
thereby contributing to both fibrotic diseases and cancer [37], [38], [39]. In silico analysis demonstrated that ovatodiolide may be an inhibitor of TGF-βRI and TGF-βRII kinase [34]. Treatment with ovatodiolide
reduced TGF-β expression levels and attenuated TGF-β-induced migration of human lung fibroblasts and their transformation into myofibroblasts
[34]. Moreover, prodrug NMP-diepoxyovatodiolide prevents peritoneal fibrosis by inhibiting
the TGF-β1/Smad and JAK/STAT signaling pathway [35]. A recent study further identified the direct target of ovatodiolide in anti-renal
fibrosis [36]. Specifically, ovatodiolide binds to the Lys403 site of glucose-6-phosphate dehydrogenase
(G6PD), inhibiting its enzymatic activity. This suppresses pentose phosphate pathway
(PPP) overactivation and mitigates renal fibrosis [36]. This study represents the first identification of a direct molecular target for
ovatodiolide and raises the question of whether this mechanism extends to other fibrotic
diseases, such as liver fibrosis. However, further studies are needed
to confirm ovatodiolideʼs efficacy against fibrosis and to fully elucidate its underlying
molecular mechanisms.
Fig. 3 Mechanisms of ovatodiolide in fibrosis suppression. Ovatodiolide suppresses fibrosis
through inhibition of TGF-β/Smad signaling and/or by directly binding to G6PD’s Lys403 site. This binding inhibits
G6PD dimer formation and reduces its enzymatic activity, leading to PPP suppression
and cellular metabolic reprogramming, ultimately attenuating fibrosis. Created in
BioRender. Li, J. (2025) https://BioRender.com/ywtfjzy. [rerif]
Immune regulation effect
Previous research identified ovatodiolide as the most potent anti-inflammatory compound
among 14 tested Anisomeles indica extracts, significantly inhibiting lipopolysaccharide-induced inflammation [40]. Additionally, studies revealed that ovatodiolide downregulated expression of CD80,
CD86, and major histocompatibility complex class II on dendritic cells, impeding their
maturation and activation [41]. It also inhibited CD4+ T cell proliferation and reduced expression of IL-4, IL-5, and TNF-α. In a murine asthma model, ovatodiolide suppressed airway inflammation, mucus production,
and attenuated airway hyperresponsiveness by downregulating Th2 cell activation [42]. Crucially, ovatodiolide deactivates NF-κB signaling–the central inflammatory regulator–alleviating histamine-induced stress
and ischemia-reperfusion-induced microglial neuroinflammation [16], [43], [44]. This NF-κB inhibition may fundamentally underlie its anti-inflammatory efficacy, positioning
ovatodiolide as a promising therapeutic agent for inflammatory diseases, including
autoimmune disorders and allergies.
Bioinformatics analyses provide additional support for ovatodiolideʼs role in immune
regulation. Molecular docking studies showed that ovatodiolide targeted TNF receptor
superfamily member 12A (TNFRSF12A) and exhibited high binding affinity for interleukin-1
receptor-associated kinase 3 (IRAK3) [45], [46], both of which are pivotal regulators of immune responses. However, these computational
predictions require experimental validation.
Antiviral and antibacterial activity
Recent studies highlight ovatodiolideʼs potential against human immunodeficiency virus
(HIV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) [34], [47]. Against HIV-1, ovatodiolide inhibited cytopathic effects within a modest concentration
range (EC₅₀ = 0.3 µM, IC₅₀ = 3.7 µM) [47]. For SARS-CoV-2, it suppressed viral activity in a concentration-dependent manner
(1.56 – 100 µM) with an IC₅₀ of 5.09 ± 0.45 µM [34]. Recent evidence suggests that ovatodiolide targets the expression of angiotensin-converting
enzyme 2 (ACE2), transmembrane protease serine 2 (TMPRSS2), and neuropilin-1 to prevent
SARS-CoV-2 infections [48], [49]. However, the precise mechanisms underlying these antiviral effects remain to be
elucidated.
Beyond viral pathogens, ovatodiolide exhibits activity against Helicobacter pylori (H. pylori), a pathogen linked to peptic ulcers, gastric mucosa-associated lymphoid tissue lymphoma,
chronic inflammation, and gastric carcinogenesis [50], [51]. As antibiotic resistance compromises H. pylori eradication efficacy, plant-derived compounds such as ovatodiolide represent promising
non-antibiotic alternatives for chronic gastritis treatment [52]. It inhibits multidrug-resistant H. pylori strains and alleviates associated inflammation [53], [54], [55], [56]. Mechanistically, it may bind to the hydrophobic pocket of ribosomal protein RpsB,
reducing RpsB levels and thereby disrupting global protein synthesis to inhibit H. pylori growth [55]. Additionally, it protects against aspirin-induced gastric ulcers by attenuating
IL-1β secretion, reducing TNF-α production, and lowering gastric acid levels [56]. Collectively, these findings indicate that ovatodiolideʼs dual actions–inhibiting
H. pylori infection and preventing gastric ulcer formation–support its therapeutic potential
against H. pylori-associated gastrointestinal pathologies.
Discussion
Recent years have witnessed significant progress in medicinal chemistry research on
ovatodiolide [5], [6]. Successful chemical synthesis has enabled further biological activity studies,
facilitating advances in pharmacological research ([Table 1]).
Table 1 Pharmacological activities of ovatodiolide.
Pharmacological activity
|
Therapeutic effect
|
Signaling pathway/target involved
|
Reference
|
Antitumor effect
|
Inhibiting cell migration and invasion
|
NF-κB↓, MMP-9↓, p-AKT↓
|
[9], [11], [14], [24], [25]
|
Suppressing proliferation
|
mTOR↓, c-myc↓, β-catenin↓, PI3K↓, JAK2↓, STAT3↓, BCR-ABL↓
|
[12], [13], [22], [27], [30]
|
Inhibiting CSCs
|
p-JAK2↓, p-STAT3↓, β-catenin↓, mTOR↓
|
[11], [20], [21], [22], [23]
|
Anti-fibrotic activity
|
Suppressing EMT, ECM production; Metabolic reprogramming
|
TβRI/TβRII kinase↓, p-Smad2/3↓, p-JAK2↓, p-STAT3↓, G6PD↓
|
[27], [34], [35], [36]
|
Immune regulation effect
|
Suppressing inflammation
|
NO↓, TNF-α↓, IL-2↓, IL8↓,IL4↓, IL5↓, IL13↓
|
[40], [41], [42], [43], [53]
|
Antiviral and antibacterial activity
|
Anti-HIV
|
/
|
[47]
|
Anti-SARS-CoV-2
|
SARS-CoV-2 3clpro activity↓ ACE2↓ TMPRSS2↓ neuropilin-1↓
|
[34], [48], [49]
|
Anti-H. pylori
|
RpsB↓
|
[55]
|
A substantial body of research has focused on the potential antitumor activity of
ovatodiolide. Numerous studies indicate that ovatodiolide likely exerts its antitumor
effects by modulating key signaling pathways, such as the NF-κB/MMP-9, JAK2/STAT3, PI3K/AKT/mTOR, and Wnt/β-catenin signaling pathways [24], [25], [26], [27]. However, the underlying molecular pharmacological mechanisms remain incompletely
elucidated. For instance, the specific molecular targets of ovatodiolide have yet
to be definitively identified, and reported pathway changes may represent secondary
effects of the compoundʼs cytotoxicity. Notably, a key advantage of ovatodiolideʼs
antitumor activity is its targeting of CSCs, accounting for the significant research
interest in its oncotherapeutic potential.
Research on ovatodiolideʼs anti-fibrotic activity remains limited, yet it demonstrates
significant inhibitory effects against fibrosis in critical organs such as the kidneys
and lungs. Mechanistic studies preliminarily indicate that, beyond suppressing the
key fibrotic TGF-β pathway, ovatodiolide additionally targets G6PD [34], [36], thereby modulating cellular metabolic reprogramming to mitigate damage and suppress
fibrosis. Whether these effects extend to liver fibrosis–a severe consequence of chronic
liver disease–requires further investigation.
Research on ovatodiolideʼs antiviral activity is also limited, with only preliminary
in vitro data suggesting potential efficacy against HIV and SARS-CoV-2 [34], [47]; thus, its therapeutic potential for viral infections remains uncertain. In contrast,
ovatodiolide exhibits direct inhibitory activity against H. pylori–potentially underpinning Anisomeles indica’s traditional use in gastrointestinal disorders [53], [55]. Evidence further indicates ovatodiolide possesses anti-inflammatory properties,
suggesting therapeutic promise for autoimmune diseases (e.g., autoimmune hepatitis
and systemic lupus erythematosus) and allergic conditions. However, thorough investigation
is needed regarding its effects on lymphocyte development, differentiation, activation,
and macrophage polarization. Notably, potential modulation of the gut microbiome and
metabolome could represent an additional immunomodulatory mechanism, warranting focused
study.
A major limitation in ovatodiolideʼs pharmacokinetic profile is its extremely short
half-life [6]. Prodrug derivatization (e.g., NMP-diepoxyovatodiolide) offers a viable strategy
to overcome this issue, enhancing bioavailability and supporting clinical translation
[6]. However, the toxicity profiles of these derivatives require rigorous evaluation.
Addressing this critical knowledge gap is essential to advance its translational development.