Next Article in Journal
Assessment of Psychological Burden in Individuals with Hereditary Risk of Pancreatic Cancer Under Surveillance: Evaluation of Distress 3 Years After Enrollment
Previous Article in Journal
The Cost of Breast Cancer: Economic and Social Perspective
Previous Article in Special Issue
New Promising Steroidal Aromatase Inhibitors with Multi-Target Action on Estrogen and Androgen Receptors for Breast Cancer Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanoformulated Terpenoids in Cancer: A Review of Therapeutic Applications, Mechanisms, and Challenges

by
Arunagiri Sharmila
1,
Priyanka Bhadra
2,
Chandra Kishore
3,4,
Chinnadurai Immanuel Selvaraj
5,*,
Joachim Kavalakatt
6 and
Anupam Bishayee
6,*
1
Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
2
Department of Biological Sciences, Bose Institute, Kolkata 700 091, West Bengal, India
3
Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
4
Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
5
Department of Genetics and Plant Breeding, VIT School of Agricultural Innovations and Advanced Learning, Vellore Institute of Technology, Vellore 632 014, Tamil Nadu, India
6
Department of Pharmacology, College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA
*
Authors to whom correspondence should be addressed.
Cancers 2025, 17(18), 3013; https://doi.org/10.3390/cancers17183013
Submission received: 2 July 2025 / Revised: 31 August 2025 / Accepted: 8 September 2025 / Published: 16 September 2025
(This article belongs to the Collection Innovations in Cancer Drug Development Research)

Simple Summary

Cancer remains a significant global health concern for which safe and effective treatment is needed. Terpenoids are natural substances derived from plants that possess the ability to prevent cancer by inhibiting cell growth, inducing cell death, and preventing metastasis. Nevertheless, the clinical applicability of terpenoids is restricted by their poor bioavailability and low water solubility. This review addresses the ways in which nanoformulation techniques can improve terpenoid distribution and efficacy. It discusses several nanocarriers that facilitate the regulated release of terpenoids as well as enhance their solubility, stability, and targeted delivery. The review also highlights current issues in clinical translation as well as emerging developments, such as the integration of artificial intelligence in nanomedicine. In summary, nanoformulated terpenoids represent a potential approach for cancer treatment by combining bioactive natural compounds with advanced drug delivery systems.

Abstract

Cancer remains a major global health concern, and thus, there is a growing demand for efficient and selective therapies with low systemic toxicity. Natural bioactive compounds have emerged as promising alternatives, and terpenoids have shown notable anticancer properties. They exert antiproliferative, proapoptotic, anti-invasive, and antimetastatic effects through the regulation of multiple molecular targets and signaling pathways, including modulation of apoptosis, suppression of angiogenesis, and inhibition of tumor-promoting inflammation. However, their clinical translation is constrained by poor aqueous solubility, low bioavailability, rapid systemic clearance, and inadequate tumor accumulation. Recent advances in nanotechnology offer strategies to overcome these limitations. Nanocarrier-based systems improve the solubility, stability, and pharmacokinetics of terpenoids, while enabling tumor-targeted delivery and controlled release. Various strategies, such as enhanced permeability and retention effect, ligand-mediated active targeting, and stimuli-responsive release have been used to achieve selective tumor accumulation and improved therapeutic outcomes. The purpose of this review is to provide a comprehensive evaluation of nanoformulated terpenoids in cancer with a special emphasis on their therapeutic applications and mechanisms of action. Preclinical studies demonstrate that nanocarrier-loaded terpenoids significantly increase bioavailability, enhance apoptosis, and suppress tumor angiogenesis compared with free terpenoids. The incorporation of artificial intelligence and machine learning further holds promise for the rational design of nanomedicines, accelerating their path toward clinical translation. Collectively, these developments position nanoformulated terpenoids as a powerful platform in precision oncology with strong potential for future application in cancer therapy.

1. Introduction

Cancer remains a critical health burden across the world, affecting global populations and healthcare systems with high incidence and fatality rates. The estimated 20 million new cases of cancer in 2022 are anticipated to rise by 77% to over 35 million cases in 2050 worldwide [1]. It remains the second-leading cause of mortality globally, accounting for around 8.97 million deaths per year, and is expected to increase to almost 18.63 million by 2060. Men are marginally more likely than women to develop cancer, with a lifetime risk of 20.2% up until the age of 74. Overall, stomach, liver, and lung cancers have the largest mortality rates, with breast cancer being more common in women and lung cancer in males [2]. It is the foremost cause of mortality among individuals under 85 in the United States. In 2025, the US is projected to report approximately 2,041,910 new cancer cases and 618,120 cancer fatalities [3]. India, the most populous country in the world, faces an increasing cancer prevalence with anticipated rises in both incidence and death rates. Cancer incidence is estimated to increase from 529.40 cases per 100,000 population in 2022 to 549.17 cases by 2031, while fatalities are anticipated to rise from 71.52 to 73.00 per 100,000 during the same period. Numerous causes, including fast urbanization, an aging population, more sedentary lifestyles, consumption of unhealthy food choices, and increase in air pollution, are some of the causes for this increase in cancer incidence in India [4]. These statistics underscore the urgent need for effective strategies in cancer detection, prevention, and treatment to address these escalating global health challenges.
Conventional cancer therapies include surgery, chemotherapy, radiation, targeted therapy, immunotherapy, and hormone therapy. Despite their effectiveness, radiation and chemotherapy have a significant risk of recurrence, severe side effects, drug-specific toxicities, and may lead to the progression of secondary cancers. Targeted therapies have improved precision but remain limited by multidrug resistance and dermatologic adverse effects. Immunotherapy has shown great promise in reducing recurrence and metastasis. It is less effective against solid tumors and entails the risk of autoimmune diseases. Solid tumors frequently produce a thick extracellular matrix, which hinders the ability of immune cells to infiltrate [5,6]. Surgery is frequently used when cancer is in its early stages and is usually not recommended in advanced stages [7]. These limitations underscore the urgent need for safer and more effective therapeutic strategies. It is essential to develop novel treatments that maintain efficacy comparable to other treatments while minimizing adverse effects and offering improved safety and tolerability profiles. The phytochemicals and plant metabolites offer potential strategies to increase therapeutic effectiveness and reduce adverse effects. Several reports have exhibited the anticancer efficacy of phytocompounds, such as phenols [8], alkaloids [9], glycosides [10], terpenoids [11], saponins [12], and sulfur-containing compounds [13]. Paclitaxel [14], vincristine [15], camptothecin [16], and etoposide [17] are some of the successfully plant-derived drugs that are popular anticancer agents in the treatment against various types of cancer, drawing attention in the research community toward further potential plant-based drug alternatives and therapies.
Plant-derived anticancer compounds can be a better alternative to conventional approaches, and terpenoids are promising choices. Terpenoids, derived from oxygenated terpenes, are crucial bioactives promoting the global use of natural remedies. The mevalonate pathway in plants plays a major role in terpenoid synthesis, while certain other terpenoids can also be derived from aquatic organisms [18]. They promote holistic, sustainable physiological benefits by providing antibacterial, anti-inflammatory, and therapeutic properties [19]. There are currently about 90,000 known terpenoids, with the majority of them obtained from plants, microbes, and marine sources [20]. The diverse range of intriguing terpenoid structures synthesized by marine species can be identified by their different structural characteristics. The metabolic products of aquatic organisms have a wide range of well-defined structural groups, including chamigrene, cembrane, and amphilectane, as well as unique functional categories, including isothiocyanate, isonitrile, isocyanate, dichloroimine, and halogen-containing groups [21].
Terpenoids are essential in a variety of sectors, including medicine [22], agriculture [23], cosmetics [24], food processing [25], and biofuels [26], owing to their diverse range of special physicochemical activities and biological roles. Terpenoids possess a significant role in different pharmacological applications (Figure 1). They exhibit antimicrobial effects on bacteria, facilitated by the terpenoids’ ability to induce cell lysis and prevent DNA and protein synthesis [27,28]. Terpenoids demonstrated excellent antioxidant activity, with the multiple mechanisms of action not limited to electron transfer, hydrogen transfer, and singlet oxygen quenching [29,30,31]. They have demonstrated significant anti-inflammatory [32,33,34], antidiabetic [35], antinociceptive [36], and anticancer properties [11,37,38]. Despite their promise of therapeutic qualities, terpenoids exhibit limited clinical applicability due to their non-specific distribution, low intestinal permeability, quick elimination, and poor water solubility. These restrictions make it more difficult to directly convert preclinical findings into clinical outcomes. Various advancements, such as nanocarrier-based drug delivery systems, present a promising solution and increase their total therapeutic potential by improving solubility, stability, pharmacokinetics, and biodistribution, while reducing toxicity [39]. In comparison with other plant phytochemicals, terpenoids further present distinct advantages as candidates for anticancer nanoformulations. Their structural diversity confers broad-spectrum anticancer activity, while their generally low toxicity profile enhances safety [40]. Moreover, the inherent lipophilicity of terpenoids facilitates efficient encapsulation within nanocarriers and promotes cellular uptake. Importantly, terpenoids have demonstrated potential in overcoming drug resistance and can synergize with other therapeutic agents, positioning them as highly promising core components for nanocarrier-based cancer therapies [41].
Nanomedicine is essential in the development of precise treatments and drug formulations, as well as in regulating the release of drug delivery systems. Drug nanocarriers offer a promising strategy in cancer therapy by improving circulation and therapeutic index while reducing toxicity to normal cells [42]. Nanotechnological approaches, especially nanoformulations, are an advanced method for enhancing the solubility and stability of phytoconstituents, enhancing absorption, safeguarding them from the body’s early deterioration, and extending their half-life. Additionally, these nanoparticles exhibit a higher penetration and absorption effect in disease tissues, hinder early contact with the biological environment, improve cellular uptake, and reduce toxicity by demonstrating a significant differential uptake capability in targeted cells [43]. The clinical use of phytocompounds is limited by several physicochemical and pharmacokinetic restrictions, including low water solubility, bioavailability, and poor selectivity. To improve their anticancer efficacy, numerous nanodrug delivery systems are being developed [44,45]. Nano-based delivery systems in cancer have the benefit of improved permeability and retention, lowering tumor hypoxia and resistance, and focusing on tumor-associated factors, such as fibroblasts, endothelial cells, and immune cells [46].
The therapeutic potential of terpenoids is limited by poor solubility, rapid metabolism, low bioavailability, and toxicity to normal cells, which hinder their effectiveness in vivo and restrict their medicinal use. Nanoformulations offer a promising solution by protecting terpenoids from degradation, enhancing pharmacokinetic properties, and improving oral absorption [47]. Through nanotechnology-based delivery systems, terpenoids achieve better systemic availability, targeted delivery, and enhanced therapeutic efficiency across various cancers. They enable targeted delivery to specific tissues, thereby reducing adverse effects. In addition, nanoencapsulation allows the co-delivery of multiple bioactive molecules to achieve synergistic anticancer effects [48]. Furthermore, terpenoid formulations based on nanotechnology have demonstrated encouraging outcomes in cancer therapeutics, with fewer side effects and enhanced effectiveness [49]. Nanotechnology provides innovative strategies to overcome the major limitations of terpenoids, including poor solubility, low bioavailability, and limited cellular uptake, by enhancing their target specificity and therapeutic efficacy. Although the synthesis and classification of terpenoids in anticancer research have been extensively studied, the application of advanced nanotechnology-based delivery systems to improve their dispersion, stability, and biological uptake remains relatively underexplored.
The aim of this review is to provide a comprehensive and critical analysis of terpenoid-loaded nanoformulations specifically developed for anticancer applications. The novelty of this review lies in presenting an integrative perspective on recent advancements and therapeutic significance in anticancer treatment. In contrast to other reviews that have primarily addressed terpenoids or nanocarriers separately [11,46], this work brings together both aspects to emphasize their combined potential in oncology, with particular focus on enhancing targeted drug delivery to tumor sites. Nanoformulated terpenoids enable selective tumor accumulation, reduce systemic toxicity, and maximize clinical efficacy, thereby underscoring their translational promise in cancer therapy. In addition, this review addresses translational challenges and evaluates diverse nanocarrier platforms. Together, these perspectives provide timely insights into the development of terpenoid-based anticancer nanomedicines by integrating natural product pharmacology, nanotechnology, and cancer therapeutics.

2. Classification and Biosynthesis of Terpenoids

Terpenes are simple hydrocarbons that include limonene, terpinene, pinene, p-cymene, and myrcene, while terpenoids are modified forms with oxygenated hydrocarbons or oxidized methyl groups [40]. They are classified by C5 units into hemiterpenes, monoterpenes, sesquiterpenes, diterpenes, sesterterpenes, triterpenes, tetraterpenes, and polyterpenes [50]. Hemiterpenes (C5H8) act as plant defense signals, while monoterpenes (C10H16) and sesquiterpenes (C15H24) show ecological roles and pharmacological effects, including antiviral, antibacterial, anti-inflammatory, and anticancer activities [51,52]. Diterpenes (C20H32), sesterterpenes (C25H40), and triterpenes (C30H48) are widely distributed in nature and exhibit antioxidant, antimicrobial, and anticancer properties [53,54]. Tetraterpenes (C40H64) such as carotenoids have immunomodulatory and cardioprotective effects [55], while polyterpenes (C40H64)n, including natural rubber, occur as plant latex [52]. Their classification by isoprene units, carbon number, and biological activity is presented in Table 1 and Figure 2.
Terpenoids are synthesized in vivo via two distinct pathways, namely the mevalonate pathway (MVA) and the 2-C-methyl-D-erythritol-4-phosphate (MEP)/1-deoxy-D-xylulose-5-phosphate pathway (DXP) (Figure 3) [56]. Both generate the C5 precursors IPP and DMAPP, which undergo rearrangements and cyclizations to form terpenoid backbones. The MVA pathway, used by eukaryotes, archaea, and various bacteria, provides precursors for triterpenes, sesquiterpenes, and phytosterols, while the MEP pathway in cyanobacteria, algae, and plant chloroplasts yields carotenoids, monoterpenes, and diterpenes [57,58]. Terpenoid biosynthesis involves three stages: (i) isopentenyl diphosphate (IPP)/dimethylallyl diphosphate (DMAPP) formation; (ii) precursor synthesis of geranyl diphosphate (GPP), farnesyl diphosphate (FPP), and geranylgeranyl diphosphate (GGPP); and (iii) terpene skeleton formation and modification [59]. GPP, FPP, and GGPP act as key intermediates for mono-, sesqui-, di-, and tetraterpenes, as well as sterols and carotenoids [60]. Further structural diversity arises from modifications by terpene synthases and enzymes such as cytochrome P450, leading to a wide range of chemical properties and biological activities [61].
Table 1. Terpenoid classification according to carbon atoms, isoprene unit, compound name, and biological activities.
Table 1. Terpenoid classification according to carbon atoms, isoprene unit, compound name, and biological activities.
ClassNo. of C AtomsNo. of Isoprene Units (C5H8)nName of CompoundsBiological ActivitiesRef.
HemiterpenesC51Isovaleric acid, senecioic acid, angelic acid, and tiglic acidConstituents in medicines, rubber, potential biofuels, and flavors[62]
MonoterpenesC102Geraniol, pinene, linalool, sabinene, borneol, menthol, myrcene, and limoneneConstituents in cosmetics, food, and pharmaceuticals. It shows antimicrobial, anti-inflammatory, antitumor, and cardioprotective properties[63]
SesquiterpenesC153Germacrane, eudesmane, β-cadinene, β-caryophyllene, artemisinin, farnesol, geosmin, and cadinene Possess antiviral, antibacterial, antidiabetic, antiobesity, and anti-inflammatory properties[55,64]
DiterpenesC204Andrographolide, sclareol, excavatolide B, kirenol, carnosic acid, phytol, retinoic acid, and oridoninPossess antioxidant, anti-inflammatory, immune-modulatory, and antirheumatoid arthritis action[53]
SesterterpenesC255Ophiobolin A, bilosespene A, merochlorin A, secoemestrin D, sterigmatocystin, leucosceptroid A, and colquhounoid AExhibits anticancer, antifeedant, and antifungal activities[51,65]
TriterpenesC306Squalene, lupeol, lanostane, oleanolic acid, betulinic acid, ursolic acid, dammarane, and ginsenosidePossess antibacterial, antifungal, anti-inflammatory, antioxidant, anticancer, antiviral, and chemopreventive properties[66]
TetraterpenesC408Carotenoid, lutein, astaxanthin, fucoxanthin, lycopene, phytoene, and β-caroteneImmunomodulatory, cardioprotective, and anticancer activities[67]
Polyterpenes>C40>8Natural rubber and resinsUsed as sealants, hot-melt adhesives, and pressure-sensitive adhesives, chewing gum contains certain polyterpene resins [68]

3. Mechanistic Insights into the Anticancer Properties of Terpenoids

Interest in terpenoids has grown because of their unique structural characteristics, strong anticancer activity, and potential to serve as key molecules in the advancement of effective antitumor medications [61]. Terpenoids display anticancer properties by targeting various molecular mechanisms involved in oncogenesis and cancer progression and by inhibiting cell growth mechanisms. They suppress the initial phases of carcinogenesis through cell cycle arrest induction, inhibition of cancer cell differentiation, and initiation of cancer cell death. By targeting various intracellular signaling mechanisms, some terpenoids can inhibit angiogenesis and metastasis in the advanced phases of cancer growth [11,69]. Terpenoids possess significant anticancer properties because they target multiple phases of cancer growth, including cellular invasion, cell death, autophagy, and proliferation (Figure 4). Terpenoids induce cell cycle arrest and apoptosis by activating p53/p21, inhibiting cyclin B/cdk1, and triggering mitochondrial pathway-related molecules, such as Bax/Bak, cytochrome c (cyt. c), caspase-9, caspase-3, and caspase-7. They also downregulate Bcl-2/Bcl-xL and suppress Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3), as well as nuclear factor-κβ (NF-κβ) signaling, reducing matrix metalloproteinase-9 (MMP-9) and cell invasion (Figure 4A). Terpenoids promote autophagy by activating AMP-activated protein kinase (AMPK), reactive oxygen species (ROS), and mitogen-activated protein kinase (MAPK) pathways, as well as upregulating Beclin 1, autophagy-related protein 5/7 (ATG5/7), and microtubule-associated protein 1A/1B-light chain 3 (LC3-II), while inhibiting phosphatidylinositol-3-kinase (PI3K)/protein kinase B (Akt)/mammalian target of rapamycin (mTOR), mesenchymal–epithelial transition factor (c-Met), and glucose transporter 1 (Glut-1) to reduce proliferation and glucose uptake. By suppressing heat shock factor 1 (HSF-1), heat shock protein 70 (HSP70), and NF-κβ, they inhibit tumor growth and show therapeutic potential against drug-resistant cancers (Figure 4B).
Terpenoids can prevent the erroneous processes implicated in cancer by regulating cell cycle arrest, apoptosis, and autophagy rather than activating through individual pathways in a coordinated and synergistic manner. These processes are connected to growth inhibition, cellular stress responses, and programmed cell death through similar signaling mediators such as p53, Bcl-2 family proteins, caspases, MAPK, and NF-κβ. Terpenoids eradicate cancerous cells by inducing mitochondrial and receptor-mediated apoptosis, restrict unchecked proliferation by enforcing cell cycle checkpoints, and improve cellular quality control and apoptosis sensitization in cancer cells by modifying autophagy. These pathways also interact to provide a cumulative inhibitory impact that increases cytotoxicity and gets past resistance mechanisms. Research demonstrates that terpenoids can increase tumor suppression by simultaneously inducing apoptosis, controlling autophagic flux, and stopping cell cycle progression in the same biological environment. Furthermore, their combination with conventional chemotherapeutics potentiates these synergistic actions, offering a promising strategy to improve overall treatment efficacy [11,70,71].

4. Challenges with Natural Terpenoids and Advantage of Nanoformulations

Terpenoids have shown significant anticancer activities via oxidative stress reduction, cell growth inhibition, apoptosis induction, angiogenesis suppression, metastasis suppression, and inflammation resolution. However, their clinical application is mostly hindered by limitations, including low aqueous solubility, low systemic availability, and quick elimination from the body. Recently, phyto-nanomedicine has received significant focus, which deals with phytochemical-loaded nanoformulations for human healthcare. Evidence indicates that by utilizing the benefits of drug delivery via nanoparticles, terpenoids can overcome their limitations and become viable anticancer treatment candidates through nanoformulation. This approach enhances their solubility and stability, extends systemic circulation, and enhances site-specific delivery to tumor sites. For instance, celastrol, a pentacyclic terpenoid with anticancer activity, suffers from poor aqueous solubility and low bioavailability. Incorporating celastrol into nanocarriers has been shown to increase its therapeutic efficacy by improving its delivery and accumulation in tumors [72,73,74]. Terpenoids’ hydrophobic characteristic restricts the drug’s therapeutic application, and increasing the production of hydrophilic terpenoids to supplement terpenoid solubility may improve their pharmacokinetic characteristics, including plasma content in the blood and oral bioavailability. Furthermore, structural changes, such as less epoxidation and allylic methyl group oxidation, might lessen the toxicity of terpenoids. As a result, solid lipid nanoparticles incorporated with terpenoids have improved stability and bioavailability while retaining their anticancer properties [11]. Moreover, nanoformulations can facilitate the co-delivery of terpenoids with other chemotherapeutic agents, with the potential to enhance synergy and combat multidrug resistance in cancer. This approach could improve the overall efficacy of cancer therapy [44].
Nanocarrier systems offer specific solutions to overcome the limitations of terpenoids, including poor solubility, instability, and low bioavailability. For instance, solid lipid nanoparticles loaded with thymoquinone improved oral bioavailability [75], while liposomes loaded with ursolic acid increased solubility and sustained release [76]. Betulinic acid encapsulated in poly(lactic-co-glycolic acid) (PLGA) nanoparticles achieved sustained release and greater tumor accumulation [77], and celastrol in polymeric micelles showed markedly improved solubility and therapeutic efficacy [78]. These examples demonstrate that nanoformulations not only resolve the inherent drawbacks of terpenoids but also maximize their anticancer potential through improved delivery and therapeutic outcomes. Numerous research organizations have shown interest in nanoformulations as possible drug delivery vehicles. Nanoformulations involve incorporating these terpenoids into nanoparticles, which can overcome the aforementioned limitations in bioavailability and solubility and enhance therapeutic potential. Nanoformulations offer key benefits in cancer therapeutics by enhancing the delivery of terpenoids. They improve water solubility and protect compounds from degradation, increasing stability in the body. Nanoparticles also enhance bioavailability by promoting better absorption and longer circulation time. Through surface modifications, they enable targeted distribution to cancer cells, reducing side effects on healthy tissue. Additionally, they provide controlled release, maintaining therapeutic levels over time. These features increase the efficacy and safety of terpenoids, enabling them to have more potential for cancer therapy [44,47,79].
The high surface-to-mass ratio of nanoparticles (between 10 and 100 nm) facilitates their easy absorption and passage through the cellular plasma membrane and permits them to attach, absorb, and deliver drugs, RNA, proteins, DNA, and identifying compounds into the cell [80]. Nanoparticles offer the benefits of target-specific drug administration, facilitating the route of water-insoluble drugs across cell membranes, and enabling the simultaneous administration of one or more therapeutic or diagnostic agents. Earlier studies have shown that terpenoids incorporated into nanoformulations have improved pharmacokinetics, biodistribution, and solubility, all of which contribute to their higher therapeutic index [81,82]. Nanoformulations promote efficient uptake by cancer cells due to their increased stability, targeted delivery, and therapeutic activity, and induce ROS, mitochondrial damage, and caspase-mediated apoptosis. The various advantages of nanoformulated terpenoids-derived anticancer medications are illustrated in Figure 5.

5. Various Processes of Preparing Nanoformulations Containing Terpenoids

Nanocarriers, such as liposomes, micelles, dendrimers, solid lipid nanoparticles, inorganic carriers, and carbon nanotubes, facilitate targeted delivery of bioactive compounds to tumors, mainly through the enhanced permeability and retention effect caused by poor lymphatic drainage [83,84,85]. Depending on synthesis methods, these can be structured as nanospheres or nanocapsules to control drug encapsulation and release, thereby improving drug accumulation at tumor sites, while reducing systemic toxicity. For terpenoids, nanoformulation plays a crucial role in overcoming hydrophobicity by enhancing solubility, stability, and bioavailability. A variety of nanocarrier approaches have been developed for terpenoid delivery, each offering unique benefits and limitations. The different types of nanocarriers for anticancer treatment are illustrated in Figure 6.
Several other nanocarrier systems have been employed to improve the delivery of terpenoids, each with distinct preparation processes and therapeutic implications. Self-emulsifying drug delivery systems (SEDDS) generate fine oil-in-water emulsions to enhance solubility and oral absorption. Polymeric nanoparticles are commonly prepared by emulsion–evaporation methods, allowing sustained and targeted release. Pickering emulsions use solid particles, such as silica nanoparticles, to stabilize emulsions without surfactants, forming core–shell structures. Liposomes, composed of phospholipid bilayers, encapsulate both hydrophilic and lipophilic terpenoids to improve systemic availability. Solid lipid nanoparticles (SLNs) and nanostructured lipid carriers (NLCs) utilize solid and mixed lipid matrices to provide controlled release and higher stability. Other advanced carriers, such as ethosomes, phytosomes, niosomes, and invasomes, employ phospholipid–ethanol or surfactant-based vesicles to enhance skin permeability, oral absorption, and transdermal delivery [86,87,88].
Nanoformulation of terpenoids through various nanodelivery systems significantly improves their solubility, stability, bioavailability, and targeted delivery. Liposomes and polymeric nanoparticles may serve as promising carriers for solid tumors, such as breast, lung, and ovarian cancers, due to their ability to enhance solubility and sustain release. SLNs and NLCs appear more suitable for highly lipophilic terpenoids requiring prolonged circulation, while SEDDS and Pickering emulsions show potential in gastrointestinal cancers. Ethosomes and phytosomes can be considered preferable for skin- and liver-targeted therapies, whereas inorganic nanoparticles may hold greater value for theranostic applications. These approaches may enhance the therapeutic efficacy of terpenoids in cancer therapeutics, offering controlled release and reduced toxicity. The comparative overview of nanocarriers and their route of administration, advantages, and disadvantages is presented in Table 2.

6. Mechanistic Insights and Anticancer Applications of Nanoformulated Terpenoids

Nanoformulated phytocompounds exert enhanced anticancer effects through multiple mechanisms driven by improved delivery, stability, and bioavailability. Terpenoid-loaded nanoformulations can stimulate natural immune system components (cytokines and antibodies), which work together to combat and defeat cancer at the molecular level [92]. This strategy increases the survival rate of cancer patients by offering appropriate chances for multimodal, site-specific drug delivery to the tumor locations. Terpenoids are encapsulated within nanoparticles whose surfaces are functionalized with cell-specific ligands that recognize and attach to receptors on cancer cells. This targeting facilitates internalization of nanoparticles by endocytosis, followed by endosomal escape, resulting in the controlled release of terpenoids inside the cancer cells [93]. Nanoformulations can improve terpenoid safety by lowering the toxicity and general intake of the compound in the body through the direct delivery of terpenoids to particular tissues or cells. This method of targeted distribution enhances terpenoids’ safety profile and reduces adverse effects [94].
The anticancer effects of nanoformulated terpenoids encompass multiple mechanisms, including inhibition of tumor cell growth, induction of apoptosis, DNA damage, and mitochondrial disruption. When encapsulated in nanosized carriers, such as liposomes, polymeric nanoparticles, or solid lipid nanoparticles, their therapeutic efficacy is further enhanced. Additionally, co-delivery systems combining terpenoids with conventional chemotherapeutics provide synergistic antitumor effects with minimized side effects [95]. These nanoformulations trigger cancer cell apoptosis through mitochondrial dysfunction and caspase activation, induce sustained cell cycle arrest, and modulate autophagy, thereby disrupting key survival processes. It promotes enhanced tumor targeting and accumulation via the enhanced permeability and retention effect (EPR) which reduces systemic toxicity while overcoming multidrug resistance by bypassing efflux pumps and regulating critical signaling pathways [96]. Overall, nanoformulated terpenoids represent a potent strategy to improve therapeutic specificity and efficacy across various types of cancer. The schematic representation of the anticancer mechanism of nanoformulated terpenoids is shown in Figure 7.
The anticancer potential of terpenoids, including their antiproliferative, proapoptotic, and immunomodulatory activities, is often limited by poor solubility and low systemic availability. Nanoformulations overcome these limitations by enabling targeted delivery with reduced side effects, making terpenoids promising therapeutic agents for various cancers. In the following sections, we discuss different anticancer properties and their mechanisms of action of various nanoformulated terpenoids. These studies are also summarized in Table 3 and Table 4 based on in vitro and in vivo results, respectively.

6.1. Monoterpenoids

Numerous monoterpenes are cytotoxic to a variety of tumor cell lines. Nevertheless, because of their lipophilic nature and requirement for high concentrations to produce antitumor effects, their application in human cancer research is restricted. Consequently, research has focused on enhancing the anticancer efficacy of monoterpenes compared to their free form by using drug delivery systems [97]. Poly(methyl methacrylate) nanoparticles containing α-terpineol (5 µg/mL) exhibited notable cytotoxic activity against human (SK-MEL-28) and mouse (B16-F10) melanoma cell lines, with inhibitory percentages of 43.8 ± 1.8% and 40.8 ± 7.7%, respectively, indicating the potential of these nanoparticles for melanoma treatment [98]. The increased potency can be attributed to improved cellular uptake and sustained release of α-terpineol from the nanoparticles. The carvacrol nanoemulsions (CEN) exhibited an increase in ROS production against A549 cells at 100 µg/mL, which in turn induced cytochrome c (cyt. c) release, caspase activation, and the regulation of critical factors, including p-JNK, Bax, and Bcl-2. This ultimately led to intrinsic apoptosis mediated by mitochondria [99]. CEN decreased MMP levels and suppressed tumor growth while inhibiting angiogenesis by downregulating cyclooxygenase-2 (COX-2), VEGF, CD31, and MAPK pathway proteins in both in vitro and in vivo models [100], highlighting how nanoencapsulation potentiates apoptotic and anti-angiogenic signaling. NLCs loaded with citral, a monoterpenoid, showed higher in vitro anti-metastatic efficacy than free citral against MDA-MB-231 breast cancer cells, and in vivo, they prevented metastasis in TNBC 4T1-challenged mice [101]; the enhanced effect is likely due to improved stability, cellular uptake, and controlled release.
Lipid-based nanocarriers loaded with perillyl alcohol demonstrated notable cytotoxicity against brain cancer cell lines, indicating that lipid encapsulation facilitates brain delivery across the blood–brain barrier [102]. The anticancer agent geraniol was conjugated with hyaluronic acid via a disulfide bond to produce a targeted delivery system, which significantly increased cytotoxicity in PC-3 cells and inhibited tumor growth in male nude mice, suggesting receptor-mediated targeting enhances efficacy [103]. Linalool nanoparticles (size = 93 nm), prepared via a self-nanoemulsifying drug delivery system and administered at 100 mg/kg in BALB/c mice, increased caspase-3 activity, ROS production, apoptosis, and decreased MMP potential and tumor growth in epithelial ovarian carcinoma [104]. Menthol nanoliposomes enhanced lymph node homing of dendritic cell-based anti-tumor vaccinations, decreasing tight junction protein expression and increasing CCR7-mediated CCL21 secretion, thereby improving immune cell targeting [105]. These studies collectively indicate that monoterpenoid-based nanoformulations enhance cytotoxicity, apoptosis, tumor suppression, antimetastatic activity, and immune targeting primarily by improving cellular uptake, stability, and controlled release, while also modulating key molecular pathways.

6.2. Sesquiterpenoids

Sesquiterpenes are potent compounds in the treatment of cancer. Despite these advantages, they have demonstrated several pharmacological limitations due to poor physicochemical properties and unfavorable pharmacokinetics. These challenges can be mitigated through nanoformulation strategies [106]. Nerolidol-loaded solid lipid nanoparticles (NR-LNPs) demonstrated half-maximal inhibitory concentration (IC50) values of 38.5 μg/mL against the Caco-2 cell line following 24 h incubation. By triggering apoptosis, NR-LNPs limited cancer cell growth to 4.2-fold more effectively than free nerolidol, highlighting how nanoparticle encapsulation improves cellular uptake and bioavailability [107]. Similarly, zerumbone-loaded nano-lipid carriers exhibited potent anticancer activity against acute human lymphoblastic leukemia cells through activation of the mitochondrial apoptotic pathway, suggesting enhanced drug delivery and mechanistic targeting [108]. Costunolide-loaded bilosome nanoparticles demonstrated an IC50 of 6.20 µM versus 15.78 µM for free costunolide, and effectively downregulated BCL2 while upregulating CASP3, TP53, and BAX, showing that nanocarriers can amplify apoptotic signaling [109]. PLGA-PEG-helenalin nanoencapsulation (size = 35–85 nm) reduced IC50 from 1.3 μM (free drug) to 0.14 μM against breast cancer cell lines, demonstrating the role of controlled release and improved stability [110].
Co-loaded niosomal nanoparticles with curcumin and artemisinin (size = 210.10 ± 13.04 nm) markedly enhanced colorectal cancer inhibition by modulating BCL2, RB, and CYCLIN D1, and upregulating BAX, FAS, and p53 [111]. Furthermore, a niosomal drug transport system loaded with farnesol and gingerol (size = 184.4–338.4 nm) showed a notable inhibitory effect against MCF-7 and SKBR3 cell lines through apoptosis induction and cell cycle regulation, emphasizing targeted intracellular delivery by upregulating BAX, CASP3, CASP9, and p21, and downregulating BCL2, CCNE, CDK4, CCND1, and HER2 [112]. Copper oxide (CuO)/titanium dioxide (TiO2)-chitosan-farnesol nanocomposites generated ROS, decreased MMP, and initiated apoptosis in SK-MEL-3 cells [113]. Similarly, cisplatin’s anticancer action against hepatocellular carcinoma (HepG2) cells was enhanced by the co-encapsulated PLGA nanoform of farnesol and cisplatin (size = 115 ± 2.5, ζ potential = −17 ± 1.5 mV) at 1.5 µg/mL [114]. In another study, human serum albumin was utilized to encapsulate artemether (a chemical derivative of artemisinin), resulting in albumin nanoparticles (size = 171.3 ± 5.88 nm) with a ζ potential of −19.1 ± 0.82 mV. It demonstrated decreased colon cancer growth in BALB/c mice by lowering interlukin-4 (IL-4) production and enhancing interferon γ (IFN-γ) cytokine release [115]. Collectively, these findings indicate that sesquiterpenoid-based nanoformulations enhance cytotoxicity, apoptosis, ROS generation, and immune modulation, providing more effective and targeted anticancer therapy through improved delivery, stability, and mechanistic engagement.

6.3. Diterpenoids

Advanced drug delivery systems play a crucial role in improving the therapeutic efficacy of diterpenes against cancer and overcoming challenges, such as selective toxicity, low bioavailability, and rapid metabolism [116]. Membrane protein-chimeric liposomes loaded with triptolide (size = 157.3 ± 1.31 nm) demonstrated IC50 values of 17.25 and 33.97 ng/mL against Hep3B and Huh7 cells, respectively, illustrating enhanced cellular uptake and antitumor efficacy in hepatocellular carcinoma [117]. Celastrol-loaded poly(ethylene glycol)-block-poly(ɛ-caprolactone) nanopolymeric micelles suppressed SO-Rb 50 cell proliferation in a concentration-dependent manner (IC50 = 17.733 μg/mL), promoting apoptosis and tumor growth inhibition in mice, highlighting controlled release and tumor-targeting benefits [118]. Paclitaxel-functionalized gold nanoparticles exhibited higher anticancer activity against HT-29 and SiHa cells than free paclitaxel, emphasizing the role of nanocarrier-mediated drug delivery in enhancing cytotoxicity [119]. Nanostructured lipid carriers loaded with sclareol (size = 127 ± 16 nm and ζ = −27 ± 2 mV) outperformed solid lipid nanoparticles (size = 115 ± 5 nm and ζ = −22 ± 7mV) in cytotoxicity against MDA-MB-231 and HCT-116 cells (IC50 = 42 ± 3 and 75 ± 8 μM, respectively), demonstrating improved encapsulation efficiency and cellular delivery [120].
Carnosic acid-loaded PLGA nanoparticles induced higher ROS-mediated apoptosis in MDA-MB-231 cells compared to the free compound [121]. Combinatorial therapy of phytol and α-bisabolol in PLGA nanoparticles (size = 268 ± 54 nm) enhanced efficacy against A549 cells (IC50 = 14 μg/mL) [122]. Retinoic acid-loaded lipid–PLGA nanoparticles with CD133 aptamers (size = 129.9 nm and ζ = −18 mV) exhibited anticancer activity against H446 (IC50 = 5.5 ± 2.1 μg/mL) and A549 (IC50 = 8.5 ± 3.5 μg/mL) cells, demonstrating targeted delivery [123]. Chitosan-coated PLGA andrographolide nanoparticles induced G1 cell cycle arrest, enhanced cytotoxicity in MCF-7 cells, extended lifespan in Ehrlich ascites carcinoma-bearing mice by 78.08%, and reduced tumor mass by 68.21% [124]. Similarly, cetuximab-conjugated andrographolide-loaded chitosan–pectin nanocomposites (5 mg/kg) showed anticancer effects against 1,2-dimethylhydrazine-induced colon cancer of Swiss albino mice [125] and oridonin-loaded anisamide–lipid calcium phosphate nanoparticles (129.5 ± 23.7 nm, ζ potential = 23.6 ± 3.4 mV) demonstrated quicker release in acidic environments and prolonged release in vivo, both of which are advantageous for drug release in lung cancer [126]. In summary, these diterpenoid-based nanoformulations enhance anticancer efficacy through mechanisms including ROS-mediated apoptosis, cell cycle arrest, targeted drug delivery, and inhibition of tumor cell growth in vitro as well as tumor progression in vivo models.

6.4. Triterpenoids

Triterpenoids possess strong anticancer activity; however, their low aqueous solubility limits systemic availability and therapeutic efficacy. These limitations can be overcome using nanocarrier-based delivery systems such as liposomes, colloids, micelles, and nanoparticles [127]. Luteolin-incorporated electrospun polycaprolactone/gelatin nanocomposites exhibited cytotoxicity against ACHN (IC50 = 52.57 μg/mL) and HSC-3 (IC50 = 66.10 μg/mL) cells, demonstrating enhanced cellular uptake and controlled drug release. Self-assembled human serum albumin nanoparticles containing oleanolic acid and doxorubicin (size = ~140 nm) showed increased cellular association and synergistic chemotherapeutic activity in B16F10 and FaDuHTB-43 cells [128]. Oleanolic acid-loaded mPEG–PLGA nanoparticles (size = 200–250 nm) induced apoptosis in A549 and HepG2 cells, highlighting improved intracellular delivery and cytotoxicity [129]. PLGA nanoparticles co-loaded with oleanolic and ursolic acids (size = 217.98 ± 2.74 nm, ζ = 26.85 ± 0.49 mV) demonstrated significant cytotoxicity against Y-79 cells (IC50 = 16.6 ± 1.3 μM) [130], whereas magnetic nanoparticles of oleanolic acid (size = 40 nm) enhanced uptake and cytotoxic effects in A549 cells [131].
Ursolic acid-loaded methoxy poly(ethylene glycol)–polycaprolactone nanoparticles (size = 144.0 ± 4.0 nm, ζ = −0.99 ± 0.3) exhibited cytotoxicity in SGC7901 cells (IC50 = 46.0 ± 2.8 μM) via cell death induction, COX-2 inhibition, and caspase-3 activation [132]. Similarly, ursolic acid and ursolic acid-polymer micelles strongly suppressed HepG2 cell proliferation (IC50 = 43.2 ± 5.01 and 37.28 ± 2.44 μM, respectively) through enhanced apoptosis [133]. Niosomal ginsenoside Rh2 nanoparticles (size = 93.5 ± 2.1 nm) improved cytotoxicity and cellular uptake in PC3 cells (IC50 = 31.24 ± 0.75 μg/mL) [134]. Another study utilizing solid lipid nanoparticles (size = 73 nm) loaded with ganoderic acid demonstrated notable cytotoxicity against HepG2 cell lines during a 72-h incubation period, with an IC50 value of 25.10 µg/mL [135]. Similar results were shown in the significant cytotoxicity exhibited by ganoderic acid and ganoderic acid-loaded nano-lipidic carriers (GA-NLCs) (size = 156 nm) over a 48-h incubation period, with IC50 values of 40.0 and 32.61 µg/mL against HepG2 cells, respectively. Additionally, GA-NLCs may improve their anticancer activity in vivo by balancing biochemicals, antioxidants, and indicators of liver damage [136]. Functionalized graphene oxide carriers loaded with ganoderenic acid D reduced subcutaneous tumor mass by 27.27 ± 1.23% via activation of the intrinsic mitochondrial apoptotic pathway [137]. Overall, triterpenoid-based nanoformulations enhance anticancer potential by improving cellular uptake, inducing apoptosis, and exerting strong cytotoxicity across various cancer types, including liver, lung, stomach, prostate, and melanoma.

6.5. Tetraterpenoids

Numerous nanoencapsulation strategies have been developed to preserve the inherent properties of tetraterpenoids and enhance their anticancer potential [138]. Encapsulation of carotenoids in nanocarriers improves solubility, cell membrane penetration, intracellular uptake, bioaccessibility, and stability, thereby enhancing their cancer-preventive and therapeutic effects [139]. In prior studies, lycopene was incorporated into silver, iron, and gold nanoparticles with sizes ranging from 50 to 100 nm and HeLa, HT29, and COLO320DM cells were exposed to all nanoparticles at a concentration of 100 ug/mL . Gold nanoparticles (AuNPs) demonstrated 40.9% inhibition against HeLa and 41.47% inhibition against HT29, but silver nanoparticles (AgNPs) inhibited COLO320DM by 41.41% in the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay (MTT) assay. AgNPs demonstrated 82.68% inhibition for COLO320DM while AuNPs demonstrated 91.21% inhibition for HT29 and 87.98% inhibition for HeLa in the SRB study, indicating enhanced cytotoxicity through nanoparticle-mediated delivery [140]. Biosynthesized and functionalized AuNPs with hydroxylated 2-ketodeinoxanthin modulated gene expression and induced apoptosis, increasing anticancer efficacy [141].
Lutein-loaded PLGA–PEG–folate nanoparticles (188.0 ± 4.06 nm) enhanced lutein uptake by ~1.6–2-fold compared to PLGA nanoparticles or free lutein in SK-N-BE(2) cells, improving intracellular delivery [142]. Similarly, capsanthin-loaded diosgenin polyethylene glycol succinate 1000 (size = 508 ± 3) micelles demonstrated a noteworthy cytotoxic effect on MDA-MB-231 breast cancer cells, with an IC50 value of 3.10 ± 1.09 μg/mL by inducing apoptosis [143]. Another group of researchers found that chitosan–glycolipid nanocarriers of fucoxanthin (10 µM) demonstrated apoptotic action against Caco-2 cells [144]. Furthermore, another study showed that PLGA microspheres infused with fucoxanthin at a 1482.50 µg/mL concentration caused H1299 lung cancer cells to shrink, exhibit blebbing of the cell membranes, and develop apoptotic bodies [145]. Polypyrrole nanoparticles loaded with astaxanthin and coupled with bovine serum albumin (10–50 μg/mL) generated ROS in MDA-MB-231 cells under 808 nm laser irradiation, demonstrating potential for photothermal and photodynamic therapy as well as photoacoustic imaging [146]. These studies indicate that the tetraterpenoid-based nanoformulations enhance anticancer efficacy by promoting apoptosis, improving cellular uptake, and increasing cancer cell inhibition, with additional promise for theranostic applications.
Table 3. In vitro anticancer properties and characteristics of nanomaterials loaded with different terpenoids.
Table 3. In vitro anticancer properties and characteristics of nanomaterials loaded with different terpenoids.
Type of
Terpenoid
CompoundType of NanocarriersTargeting
Mediator/
Ligand
(Size, Shape and ζ Potential)Cancer Type and CellsIC50Anticancer EffectsMechanismsRef.
MonoterpenoidsGeraniolNanostructured lipid nanocarriersNone110 nm, ζ potential = −10 mV, shape = NRLung cancer (A549)1.5 mM↓Cell viability↑MMP loss[147]
Hyaluronic acid-based polymeric nanoconjugateHyaluronic acid110 nm, Spherical, ζ potential = NRLiver cancer (HepG2 and Huh7)80 and 100 μM↓Cell growth; ↑apoptosis; ↓cell proliferation↑Bax; caspase-3; ↑caspase-9; ↓Bcl-2, ↓PARP[148]
Zinc–tin oxide/dextran/geraniol
nanocomposites
None197.40 nm, Agglomerated, ζ potential = NRColon cancer (HCT-116)10 μg/mL↑Cytotoxicity; ↑apoptosis↑Caspase-3, ↑caspase-8; ↑caspase-9[149]
α-PineneNanoemlusionNone190 ± 8 nm, ζ potential = −10.4 ± 8 mV, shape = NRMelanoma (A-375) and breast cancer (MCF-7)106.19 and
168.02 μg/mL
↑Cytotoxicity; ↑apoptosis↑Bax/Bcl-2 ratio[150]
Chitosan nanoparticlesNone102 ± 6 nm, ζ potential = 41.7 ± 1 mV, shape = NRMelanoma (A-375)76.4 µg/mL↑Cytotoxicity↑Bax/Bcl-2 ratio[151]
LinaloolGold nanoparticles CALNN peptide5–20 nm, Spherical, ζ potential = NRBreast cancer (MCF-7)10 μg/mL↑Cell growth; ↑apoptosis; ↓cell proliferation
↑ROS; ↓MMP; ↑caspase-8; ↑p53; ↓NF-κB[152]
Solid lipid nanoparticlesNone90–130 nm, Spherical, ζ potential = −4.0 mVLiver (HepG2) and lung cancer (A549)2 mM↑CytotoxicityNot reported[153]
Gold nanoparticles Glutathione +
CALNN peptide
13 nm, shape and ζ potential = NROvarian cancer (SKOV-3)10 µg/mL↑Cytotoxicity; ↑apoptosis; ↓cell proliferation↑ROS; ↓MMP; ↑caspase-8; ↑p53; ↓NF-κB [154]
MentholIron oxide nanoparticlesNone20–60 nm, shape and ζ potential = NRGastric cancer (AGS)252 μg/mL↑Cytotoxicity; ↑apoptosis↑Caspase-8[155]
Chitosan gumNone160 ± 15 nm, ζ potential = 43 ± 4 mV, shape = NRMelanoma (A-375)29 µg/mL↑CytotoxicityNot reported[156]
LimoneneChitosan nanoparticlesNone209 ± 13 nm, shape and ζ potential = NRMelanoma (A-375) and breast cancer (MDA-MB-468)30.24 and 650.70 µg/mL↓Cell viabilityNot reported[157]
Sesquiterpenoidsβ-caryophylleneSilver nanoparticlesNone3.2 nm, Agglomerated, ζ potential = NRLiver cancer (HepG2)51.71 µg/mL↓Cell growthNot reported[158]
 Silver nanoparticlesNone29.42 nm, Spherical, ζ potential = NRLung cancer (A549)9.39 ± 0.08 g/mL↓Cell proliferationNot reported[159]
ArtemisininChitosan magnetic nanoparticlesNone349–445 nm, Spherical, ζ potential = −9.34 to −33.3 mVBreast cancer (MCF-7)25.61 ± 13 g/mL↑Cytotoxicity; ↑apoptosisNot reported[160]
Lipid nanoparticlesNone70 ± 20 nm, shape and ζ potential = NRTriple negative breast cancer (MDA-MB-231)7 ± 2 μm↑Cytotoxicity↓HER2; ↓survivin; ↓cyclin D1; ↓EGFR[161]
Mesoporous silica nanoparticles-loaded PLGA nanofibersNone150–200 nm, shape and ζ potential = NRBreast cancer (SK-BR-3)55 μM↑Cytotoxicity; ↑apoptosis↑Bax; ↑caspase-3; ↑p53; ↓Bcl-2; ↓hTERT[162]
FarnesolChitosan-encapsulated nickel oxide, tin dioxide nanoparticlesNone34.8 nm, Agglomerated hexagonal structure, ζ potential = NRBreast cancer (MDA-MB-231)17.58 μg/mL↑Cytotoxicity; ↑apoptosis; ↓cell proliferation↓MMP; ↑ROS; ↑G2/M checkpoint arrest[163]
DiterpenoidsAndrographolideSolid lipid nanoparticlesNoneNot reportedHead and neck squamous cell carcinoma (HN6 and
HN30)
6.087 and
11.74 μg/mL
↑Cytotoxicity; ↑apoptosis; ↓cell proliferationNot reported[164]
Albumin nanoparticlesNone100–200 nm, shape and ζ potential = NRCervical cancer (HeLa)39.46 μg/mL↑CytotoxicityNot reported[165]
SclareolHyaluronan-coated PLGA nanoparticlesNone100–150 nm, ζ potential = −30 mV, shape = NRBreast (MCF-7, MDA-MB468) and colon cancer (CaCo-2)50 μM↑CytotoxicityNot reported[166]
Solid lipid nanoparticlesNone88 nm, shape and ζ potential = NRLung cancer (A549)19 μg/mL↓Cell viability; ↑apoptosisNot reported[167]
Carnosic acidAlbumin nanoparticlesNone97.29–144.26 nm,
ζ potential = −21.03 mV, shape = NR
Colon (Caco-2) and breast cancer (MCF-7)2.60 and
6.02 μg/mL
↓Cell viability; ↑apoptosis↑G2/M checkpoint arrest; ↓COX-2; ↓Bcl-2; ↑GCLC; ↑p53[168]
Retinoic acidChitosan nanoparticlesNone313.23 ± 1.75 nm, ζ potential = 2.42 ± 0.04 mV, shape = NRBreast cancer (MCF-7)2.28 ± 0.02 µg/mL↑Cytotoxicity; ↑apoptosis↓Bcl-2; ↑caspase-3; ↑Bax; ↑cleaved PARP; ↑8-oxo-dG [169]
Glutenin NanoparticlesFolic acid~185 nm, Spherical, ζ potential = −3 mV Breast cancer (MCF-7)55.93 µg/mL↑Cytotoxicity; ↑apoptosis; ↓cell proliferation↓MMP; ↑ROS[170]
Solid lipid nanoparticlesNone140–150 nm, Spherical shape, ζ potential = −13 and −19 mVProstate cancer (LNCap)200 μg/mL↑Cytotoxicity; ↑apoptosisNot reported[171]
TriterpenoidsSqualeneCisplatin-nanoprecipitated particlesNone128–160 nm, Spherical, ζ potential = NRColon cancer (HT-29 and
KM-12)
8 μmol/L↑Cytotoxicity; ↑apoptosis; ↓cell proliferation;Not reported[172]
PLGA nanoparticlesNoneNot reportedColon cancer (Caco-2)140 µg/mL↑Cytotoxicity; ↑apoptosis↑ROS generation[173]
LupeolChitosan nanoparticles with cellulose acetate membrane None12 nm, shape and ζ potential = NRSkin cancer (A431)42.2 μg/mL↑CytotoxicityNot reported[174]
Galactosylated liposomesNone100 nm, shape and ζ potential = NRLiver cancer (HepG2)30 µM↑Cytotoxicity; ↑apoptosis↓p-Akt308; ↓p-Akt473 levels[175]
Oleanolic acidAlbumin nanoparticlesCetuximab171 ± 4.8 nm to 180 ± 3.7 nm, ζ potential = − 33.3 ± 3.4 mV, shape = NRLung cancer (A549)4.34 ± 1.90 μg/mL↑Cytotoxicity; ↑apoptosis; ↓cell proliferation↑ROS generation; G0/G1 checkpoint arrest[176]
Ursolic acidGold PLGA nanoparticlesNone80 nm, Spherical, ζ potential = NRCervical cancer (SiHa,
CaSki, and HeLa cells)
100 μM↓Cell viability; ↑apoptosis; ↓cell proliferation; ↓cell migration ↓cell invasion↑Bax; ↓Bcl-2; ↑caspase-3, ↑caspase-8; ↑capase-9; ↓procaspase-3; ↓procaspase-8; ↓procaspase-9; ↑p53; ↑fas; ↓cIAP[177]
Chitosan-coated PLGA nanoparticlesNone250 nm, Spherical, ζ potential = NRBreast cancer (MCF-7 and
MDA-MB-231)
26.74 and 40.67 μM↑CytotoxicityNot reported[178]
PLGA-PEG nanoparticlesNone133.6 ± 0.7 nm, ζ potential = −22.6 ± 2.8 mV, shape = NRPancreatic ductal adenocarcinoma (AsPC-1 and
BxPC-3)
11.7 ± 0.6 and 14.1 ± 2.2 μM↑CytotoxicityNot reported[179]
OridoninSolid lipid nanoparticlesNone108.53 ± 10.92 nm, ζ potential = −37.97 ± 3.78 mV, shape = NRBreast (MCF-7), lung (A549) and liver cancer (HepG2)22.6, 25.3 and 30.1 μM↓Cell viability; ↑apoptosis; ↓cell proliferation↑G2/M checkpoint arrest; ↓G1/G0 checkpoint arrest;[180]
GinsenosideLiposomes combined with paclitaxelNone77.71 ± 3.22 nm, ζ potential = −39.21 ± 1.03 mV, shape = NRGastric cancer (BGC-823)0.04 μg/mL↑Cytotoxicity; ↑apoptosisNot reported[181]
TetraterpenoidsLycopeneEudragit RL 100 polymeric nanoparticlesNone62.10 ± 3.7 nm, Spherical Prostate cancer (LNCaP and
PC-3)
25.43 and 10.03 μg/mL↑CytotoxicityNot reported[182]
Gold nanoemulsionNone25.0 ± 4.2 nm, ζ potential = −32.2 ± 1.8 mVColon cancer (HT-29)0.1 µM↑Cytotoxicity; ↑apoptosis; ↓cell migration↑Bax; ↓Bcl-2; ↓procaspase-8; ↓procaspase-3; ↓procaspase-9; ↑E-cadherin; ↓Akt; ↓NF-κB; ↓MMP-2; ↓MMP-9[183]
CarotenoidNanoemulsionNone15.1 nm, shape and ζ potential = NRColon cancer (HT-29)4.9 μg/mL↑Cytotoxicity; ↑apoptosis; ↓cell proliferation↑G2/M checkpoint arrest; ↑p53; ↑p21; ↓CDK1; ↓CDK2; ↓cyclin A; ↓cyclin B[184]
LuteinChitosan/alginate iron oxide nanoparticlesNone264 ± 6 nm, ζ potential = −13.3 ± 1.6 mV, shape = NRBreast cancer (MCF-7)4.12 ± 0.4 μg/mL↓Cell viabilityNot reported[185]
FucoxanthinPolyvinylpyrrolidone nanoparticlesNone<50 nm, shape and ζ potential = NRColon cancer (Caco-2)20 μM↓Cell viability; ↓cell migration↑Pro-oxidative effect[186]
AstaxanthinChitosan nanoparticlesNone<400 nm, shape and ζ potential = NRMelanoma (B16F10)20 µg/mL↑Antioxidant activity; ↓cell proliferation; ↓cell migrationNot reported[187]
Gold nanoparticlesNone60–120 nm, Spherical, ζ potential = NRBreast cancer (MDA-MB-231)50 μg/mL↑Cytotoxicity; ↑apoptosis; ↓cell proliferationNot reported[188]
β-CaroteneSolid lipid nanoparticlesNone203 ± 7.23 nm, ζ potential = −7.21 ± 0.82 mV, shape = NRBreast cancer (MCF-7)40 μg/mL↑Antioxidant activity; ↑cytotoxicityNot reported[189]
Solid lipid nanoparticlesNone111.78 nm, Spherical, ζ potential = −26.3 ± 1.3 mV, shape = NRBreast cancer (MCF-7)14.89 ± 0.02 μg/mL↑Cytotoxic effectNot reported[190]
Symbols and abbreviations: ↑, increase/upregulation;  ↓, decrease/downregulation; Bax, Bcl-2-associated X protein; Bcl-2, B-cell lymphoma 2; CALNN, cysteine–alanine–leucine–asparagine–asparagine peptide; cIAP, cellular inhibitor of apoptosis protein; CDK, cyclin-dependent kinase; COX-2, cyclooxygenase-2; EGFR, epidermal growth factor receptor; GCLC, glutamate–cysteine ligase catalytic subunit; HER2, human epidermal growth factor receptor 2; hTERT, human telomerase reverse transcriptase; IC50, half maximal inhibitory concentration; MMP, mitochondrial membrane potential; MMP loss, loss of mitochondrial membrane potential; NF-κB, nuclear factor-κB; NR, not reported; p-Akt308/p-Akt473, phosphorylated Akt at threonine 308/serine 473; PARP, poly (ADP-ribose) polymerase; PLGA, poly (lactic-co-glycolic acid); ROS, reactive oxygen species; ζ potential, Zeta potential.
Table 4. In vivo anticancer properties and characteristics of nanomaterials loaded with different terpenoids.
Table 4. In vivo anticancer properties and characteristics of nanomaterials loaded with different terpenoids.
Type of TerpenoidCompoundType of NanocarriersTargeting
Mediator/
Ligand
Size, Shape and ζ PotentialCancer Type (Model)Dosage and Route of AdministrationAntitumor EffectsMechanismsRef.
MonoterpenoidsGeraniolHyaluronic acid-based polymeric nanoconjugateHyaluronic acid110 nm, ζ potential = −10 mV, shape = NRLiver cancer (H22 cells-injected mice)Intravenous injection, 1.0 mg geraniol/kg↓Tumor size; ↓tumor
weight; ↓tumor volume
↓Ki-67[148]
Zinc–tin oxide/dextran/geraniol
nanocomposites
None197.40 nm, Agglomerated, ζ potential = NRColon cancer (DMH-induced rats)20 and 40 mg/kg↓Tumor volume; ↓tumor size; ↓tumor incidence↓COX-2[149]
DiterpenoidsAndrographolidePLGA nanocapsulationNone163 nm, Nanospheres, ζ potential = −57.85 mVCervical cancer (HeLa cells-injected mice)10mg/kg↓Tumor sizeNot reported[191]
Carnosic acidLiposomesTransferrin97.06 ± 3.389 nm,
ζ potential = 2.55 ± 1.26 mV, shape = NR
Liver cancer (HepG2- and SMMC-7721-transplanted BALB/c nude mice)Intraperitoneal injection, 2 mg/kg↓Tumor growth; ↓tumor volume↑Caspase-3; ↑caspase-9; ↑Bax; ↑Bad; ↑PARP; ↓Bcl-2[192]
OridoninPEG-PLGA nanoparticlesNone100 nm, ζ potential = −5 mV, shape = NRBreast cancer (MCF-7 cells-injected BALB/c nude mice)Intraperitoneal injection, 10 mg/kg↓Tumor size; ↓tumor volume; ↓angiogenesisNR[193]
TriterpenoidsUrsolic acidChitosan nanoparticlesFolate160nm, ζ potential = 39.3mV, shape = NRBreast cancer (MCF-7 cells-injected BALB/c mice)Intraperitoneal injection, 12.5mg/kg↓Tumor size; ↓tumor weightNR[194]
Gold PLGA nanoparticlesNone80 nm, Spherical, ζ potential = NRCervical cancer (SiHa and HeLa cells-injected nude mice)Intraperitoneal injection, 20 mg/kg↓Tumor size; ↓tumor weightNR[177]
TetraterpenoidsLycopeneN-isopropylacrylamide with N-vinyl2-pyrrolidone poly(ethyleneglycol)monoacrylate copolymeric nanoparticlesNone<100 nm, Spherical, ζ potential = NRMelanoma (TPA-induced Swiss albino mice)Topical treatment, 1 μg/mLNR↓COX-2, ↓Bcl-2, ↑Bax[195]
AstaxanthinEthylcellulose nanoparticlesNone185 nm, Spherical, ζ potential = NROral cancer (DMBA- induced Syrian hamsters)Oral treatment, 15 mg/kg↓Tumor size; ↓tumor growthCyclin D1; ↓Bcl-2, ↑Bax; ↑caspase-3; ↑caspase-9[196]
β-CaroteneThiolated chitosan -lithocholic acid nanomicellesNone<300 nm, ζ potential = +27.0 mV, shape = NRSkin cancer (DMBA-
induced BALB/c mice)
Topical treatment, 1 mg/mL↓Tumor size; ↓tumor weightNR[197]
Symbols and abbreviations:↑, increase/upregulation;  ↓, decrease/downregulation; Bad, Bcl-2-associated death promoter; Bax, Bcl-2 associated X protein; Bcl-2, B-cell lymphoma 2; COX-2, cyclooxygenase 2; DMBA, 7,12-dimethylbenz[a]anthracene; DMH, 1,2-dimethylhydrazine; NR, not reported; PARP, poly (ADP-ribose) polymerase; PEG-PLGA, polyethylene glycol–poly(lactic-co-glycolic acid); PLGA, poly(lactic-co-glycolic acid); TPA, 12-O-tetradecanoylphorbol-13-acetate; ζ potential, Zeta potential.

7. Challenges and Emerging Trends in Nanoformulated Terpenoids

Although these innovative nanodrug delivery methods of terpenoids have many benefits, there are also several drawbacks in terms of real-world applications. Several of these challenges include the specific description of these nanoproducts, their toxicity and safety characterizations, and the absence of efficient control. Two significant obstacles are the increased cost and manufacturing complexity, as the synthesis and characterization of nanoformulations require several expensive tools. Furthermore, although there are many approved nanomedicines on the list, their clinical uses are limited because of the lack of a specific regulatory standard for the formulation and assessment of these nanoproducts. Since nanotechnology has diverse applications, more comprehensive and updated regulatory standards are required. The governing bodies of multiple countries must work together to develop precise and rigorous rules that can handle major safety issues and guarantee the creation of safe and advantageous nanomedicines for humankind [198].
Nanoformulated terpenoid compounds encapsulated within nanoparticles have shown promise in enhancing cancer diagnosis and therapy. However, several challenges and limitations hinder their widespread clinical applications. One major concern is biocompatibility and toxicity, as variations in nanoparticle size, shape, and surface chemistry can result in unexpected interactions with biological systems, potentially causing side effects on healthy tissues [199]. Additionally, stability and aggregation pose substantial hurdles, particularly for polymeric nanoparticles commonly used to encapsulate terpenoids. Under physiological conditions, these nanoparticles are susceptible to aggregation, potentially undermining colloidal stability, reducing their bioavailability, and hindering their effectiveness in drug delivery and imaging applications [200].
Another limitation is the lack of precise targeting of cancer cells. Many nanoformulated terpenoids exhibit non-specific distribution throughout the body, which not only reduces the therapeutic levels at the tumor site but simultaneously elevates the risk of off-target effects on healthy tissues. This underscores the urgent need for advanced and specific targeting strategies, such as ligand-mediated or stimuli-responsive systems, to increase the selectivity and diagnostic accuracy of these formulations [200]. Furthermore, reliance on the EPR effect for tumor targeting presents another challenge. Although the EPR effect facilitates passive accumulation of nanoparticles in tumor tissues, its effectiveness is highly inconsistent due to tumor heterogeneity, vascular abnormalities, and variations in the tumor microenvironment. These factors can significantly limit the predictability and uniformity of nanoparticle accumulation across different tumor types and patient populations, leading to variable therapeutic and diagnostic outcomes [201].
Nanosized drug delivery methods generate concerns about short- and long-term toxicities and biocompatibility, which should be evaluated by specialized in vivo toxicology investigations in suitable animal models [202]. The important factors are the cost of these medicines and the distribution of nanomedicines globally at the lowest levels of care (government hospitals and primary healthcare). Furthermore, additional investigation is crucial to accurately characterize and interpret these toxicities and their effect on the environment, human body, and human physiology. Researchers are seriously concerned about the ethical application of nanomedicine in society as well as the safety concerns [203]. Addressing these challenges requires ongoing research to develop nanoformulations with improved stability, biocompatibility, and targeted delivery capabilities to increase the efficacy of terpenoid-based cancer diagnostics.
New developments in nanoformulations lead to the formation of multipurpose nanosystems that can perform imaging, diagnostics, and therapy concurrently, paving the way for personalized medicine strategies. Furthermore, the combination of stimulus-responsive nanoparticles and smart materials offers the possibility of regulated drug release, which can be triggered by external forces or physiological factors. Their translational ability is further enhanced by the creation of biocompatible and biodegradable nanocarriers, which will reduce toxicity and increase drug absorption [204]. Moreover, bioinformatics techniques are essential for applications including drug targeting, analysis of protein–protein interactions, and identification of compound–target networks and cancer treatment pathways. A growing trend in bioinformatics, information technology, and omics is the development of resources that provide information on herbal formulations and the bioactive chemicals of medicinal plants. Numerous initiatives do exist as databases, such as the Collective Molecular Activities of Useful Plants (CMAUP), SymMap, Indian Medicinal Plants, Phytochemistry and Therapeutics (IMPPAT), and the Indian Medicinal Plants Database (IMPLAD). Additionally, experts have created new approaches for analyzing the pharmacokinetic characteristics of medications and phytochemicals using in silico methods. These methods can also be applied to in silico screening, elucidation of plausible mechanisms of action, and drug discovery of phytoconstituents [205].
Drugs made using nanocarriers may present a number of difficulties, one of which is obtaining regulatory approval to use clinical trials. As a result, artificial intelligence combined with these nanocarriers may challenge the existing framework for these kinds of innovative interventional treatments. In contrast to wet-lab methods, artificial intelligence (AI) tools offer a logical framework for screening a large range of chemicals using models, which reduces the need for time and resources. Along with other aspects, not limited to drug-target identification, bioactivity, de novo drug design, physicochemical property estimation, and quantitative structure–activity relationship (QSAR)-based drug development, AI has also been used to identify drug toxicity, drug loading, and surface-level characterization. AI-based formation of nanoformulated medications and their targets may enhance the platform for creating innovative anticancer treatments [206]. AI has been used to predict treatment efficacy and to design, characterize, and manufacture drug delivery nanosystems [207]. The integration of in silico modeling or AI and machine learning tools may successfully address the challenges related to terpenoid-loaded nanoformulations in cancer therapy. This enables precise design and optimization of targeted drug delivery systems, thereby improving the therapeutic efficiency and specificity of anticancer compounds (Figure 8).

8. Conclusions and Future Directions

Terpenoids have been recognized as crucial components of current and future potential anticancer drugs. The use of nanotechnology in cancer treatments has demonstrated promise in addressing the issues with conventional treatment strategies, such as those related to instability, high toxicity, poor water solubility, low absorption, multidrug resistance, low bioavailability, and low specificity. Additionally, using multi-compartmental nanocarriers allows the concurrent delivery of several medications, which makes it easier to co-administer herbal and other medications for synergistic effects. In our review, the different classes of terpenoids and their anticancer mechanisms were elaborately discussed. Furthermore, terpene-based nanoformulations are known to be an efficient tool for delivering drugs, as they improve the solubility, bioavailability, biocompatibility, and help to overcome the drawbacks of plant-based terpenoids used for cancer treatment. It is an extremely difficult task for researchers and manufacturers to create terpenoid-based nanoformulations and optimize their dosage either alone or in mixture with other anticancer drugs. Nonetheless, researchers are certain that terpene-based nanoformulations will soon find a place in the arsenal of cancer treatment and tumor therapies. In the future, comprehensive investigations are needed to evaluate the synthesis, drug delivery mechanisms, potential toxic effects, and safety aspects of terpenoid-based nanoformulations through rigorous in vitro and in vivo studies.

Author Contributions

Data curation, A.S.; visualization, A.S.; writing—original draft., A.S.; formal analysis, P.B., C.K., J.K. and C.I.S.; writing—review and editing, P.B., C.K., J.K., C.I.S. and A.B.; conceptualization, C.I.S. and A.B.; supervision, C.I.S. and A.B.; validation, C.I.S. and A.B.; project administration, C.I.S. and A.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

AACT, acetoacetyl-CoA thiolase; ApcMin/+, adenomatous polyposis coli-multiple intestinal neoplasia; AP-1, activator protein-1; BALB/c, Bagg albino c; Bcl-2, B-cell leukemia/lymphoma 2 protein; CCNG1, cyclin G1; DMAPP, dimethylallyl diphosphate; DXP, 1-deoxy-D-xylulose-5-phosphate; DXR, 1-deoxydxylulose 5-phosphate reductoisomerase; DXS, 1-deoxyd-xylulose 5-phosphate synthase; EMT, epithelial–mesenchymal transitions; EPR, enhanced permeability and retention effect; ER, endoplasmic reticulum; ERK, extracellular signal-regulated kinase; FPP, farnesyl pyrophosphate; FPPS, farnesyl pyrophosphate synthase; GGPP, geranylgeranyl pyrophosphate; GGPS, bovine pyrophosphate synthase; GGPPS, geranylgeranyl pyrophosphate synthase; GPP, geranyl pyrophosphate; GPPS, geranyl pyrophosphate synthase; GPP, geranyl pyrophosphate; GPPS, geranyl pyrophosphate synthase; G3P, d-glyceraldehyde 3-phosphate; HDR, (E)-4-hydroxy-3-methylbut-2-enyl diphosphate reductase; HDS, (E)-4-hydroxy-3-methylbut-2-enyl diphosphate synthase; HMLER-TWIST, human mammary luminal epithelial cells/transcription factor; HMG-CoA, 3-hydroxy-3-methyl-glutaryl-CoA; HMGR, 3-hydroxy-3-methylglutaryl-CoA reductase; HMGS, 3-hydroxy-3-methylglutaryl-CoA synthase; HSP60, heat shock protein 60; ICAM-1, intercellular adhesion molecule1; IDI, isopentenyl diphosphate isomerase; IPP, isopentenyl diphosphate; IPPP, isopentenyl pyrophosphate; JAK-STAT, Janus kinases-signal transducer; MCT, 2-C-methyl-d-erythritol 4-phosphate cytidylyltransferase; MDD, mevalonate diphosphate decarboxylase; MDS, 2-C-methyl-d-erythritol 2,4-cyclodiphosphate synthase; MEF2D, myocyte enhancer factor 2D; MEP, 2-C-methyl-D-erythritol-4-phosphate; MK, mevalonate kinase; MNU, N-methyl-N-nitrosourea; MNNG, methyl-N-nitro-N-nitrosoguanidine; MTT, 3-[4,5-dimethylthiazol-2yl]-2,5-diphenyl tetrazolium bromide; MVA, mevalonate; MVAP, mevalonate 5-phosphate; MVAPP, mevalonate diphosphate; NCI-H, National Cancer Institute-Human; NF-κB, nuclear factor-κB; MMPs, metalloproteinases; OSCs, oxidosqualene cyclases; CYP, cytochrome P450; PARP, poly (ADP-ribose) polymerase; PI3K, phosphatidylinositol-3-kinase; PLC/PRF/5, primary liver carcinoma/-patient with recurrence of fibrolamellar carcinoma5; PLGA, poly(lactic-co-glycolic acid); SRB, sulforhodamine B; PMK, phosphomevalonate kinase; PTEN, phosphatase and TENsin homolog deleted on chromosome 10; QSAR, quantitative structure–activity relationship; SCID/bg, severe combined immunodeficiency/beige; SE, squalene epoxidase; SEDDS, self-emulsifying drug delivery systems; SNAIL, snail family transcriptional repressor 1; SLNs, solid lipid nanoparticles; NLCs, nanostructured lipid carriers; TLR4, toll-like receptor 4; TNBC, triple negative breast cancer; TPS, terpene synthase; uPA/uPAR, urokinase plasminogen activator/urokinase-type plasminogen activator receptor; VEGF, vascular endothelial growth factor; VEGF, vascular endothelial growth factor; XIAP, X-linked inhibitor of apoptosis protein.

References

  1. Cao, W.; Qin, K.; Li, F.; Chen, W. Comparative Study of Cancer Profiles Between 2020 and 2022 Using Global Cancer Statistics (GLOBOCAN). J. Natl. Cancer Cent. 2024, 4, 128–134. [Google Scholar] [CrossRef]
  2. Mattiuzzi, C.; Lippi, G. Current Cancer Epidemiology. J. Epidemiol. Glob. Health 2019, 9, 217–222. [Google Scholar] [CrossRef]
  3. Siegel, R.L.; Kratzer, T.B.; Giaquinto, A.N.; Sung, H.; Jemal, A. Cancer Statistics, 2025. Ca 2025, 75, 10. [Google Scholar] [CrossRef]
  4. Jena, D.; Padhi, B.K.; Zahiruddin, Q.S.; Ballal, S.; Kumar, S.; Bhat, M.; Sharma, S.; Kumar, M.R.; Rustagi, S.; Gaidhane, A.M.; et al. Estimation of Burden of Cancer Incidence and Mortality in India: Based on Global Burden of Disease Study 1990–2021. BMC Cancer 2024, 24, 1278. [Google Scholar] [CrossRef]
  5. Wei, G.; Wang, Y.; Yang, G.; Wang, Y.; Ju, R. Recent Progress in Nanomedicine for Enhanced Cancer Chemotherapy. Theranostics 2021, 11, 6370. [Google Scholar] [CrossRef]
  6. Gavas, S.; Quazi, S.; Karpiński, T.M. Nanoparticles for Cancer Therapy: Current Progress and Challenges. Nanoscale Res. Lett. 2021, 16, 173. [Google Scholar] [CrossRef]
  7. Khan, M.S.; Gowda, B.J.; Hasan, N.; Gupta, G.; Singh, T.; Md, S.; Kesharwani, P. Carbon Nanotube-Mediated Platinum-Based Drug Delivery for the Treatment of Cancer: Advancements and Future Perspectives. Eur. Polym. J. 2024, 206, 112800. [Google Scholar] [CrossRef]
  8. Bakrim, S.; El Omari, N.; El Hachlafi, N.; Bakri, Y.; Lee, L.-H.; Bouyahya, A. Dietary Phenolic Compounds as Anticancer Natural Drugs: Recent Update on Molecular Mechanisms and Clinical Trials. Foods 2022, 11, 3323. [Google Scholar] [CrossRef] [PubMed]
  9. Mondal, A.; Gandhi, A.; Fimognari, C.; Atanasov, A.G.; Bishayee, A. Alkaloids for Cancer Prevention and Therapy: Current Progress and Future Perspectives. Eur. J. Pharmacol. 2019, 858, 172472. [Google Scholar] [CrossRef]
  10. Khan, H.; Saeedi, M.; Nabavi, S.M.; Mubarak, M.S.; Bishayee, A. Glycosides from Medicinal Plants as Potential Anticancer Agents: Emerging Trends towards Future Drugs. Curr. Med. Chem. 2019, 26, 2389–2406. [Google Scholar] [CrossRef] [PubMed]
  11. Kamran, S.; Sinniah, A.; Abdulghani, M.A.; Alshawsh, M.A. Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A Scoping Review. Cancers 2022, 14, 1100. [Google Scholar] [CrossRef]
  12. Elekofehinti, O.O.; Iwaloye, O.; Olawale, F.; Ariyo, E.O. Saponins in Cancer Treatment: Current Progress and Future Prospects. J. Pathophysiol. 2021, 28, 250–272. [Google Scholar]
  13. Zheng, H.; Wang, Y.; Ren, Y.; Wang, X.; Sui, L.; Xu, H.; Zheng, C. Design, Synthesis and Biological Evaluation of Sulfur-Containing Tetrahydroxanthones as Potential Anti-Tumor Agents. Bioorg. Med. Chem. Lett. 2025, 121, 130154. [Google Scholar] [CrossRef]
  14. Sati, P.; Sharma, E.; Dhyani, P.; Attri, D.C.; Rana, R.; Kiyekbayeva, L.; Büsselberg, D.; Samuel, S.M.; Sharifi-Rad, J. Paclitaxel and Its Semi-Synthetic Derivatives: Comprehensive Insights into Chemical Structure, Mechanisms of Action, and Anticancer Properties. Eur. J. Med. Res. 2024, 29, 90. [Google Scholar] [CrossRef]
  15. Bakhshi, S.; Shoari, A.; Alibolandi, P.; Ganji, M.; Ghazy, E.; Rahdar, A.; Fathi-karkan, S.; Pandey, S. Emerging Innovations in Vincristine-Encapsulated Nanoparticles: Pioneering a New Era in Oncological Therapeutics. J. Drug Deliv. Sci. Technol. 2024, 91, 105270. [Google Scholar]
  16. Kamle, M.; Pandhi, S.; Mishra, S.; Barua, S.; Kurian, A.; Mahato, D.K.; Rasane, P.; Büsselberg, D.; Kumar, P.; Calina, D. Camptothecin and Its Derivatives: Advancements, Mechanisms and Clinical Potential in Cancer Therapy. Med. Oncol. 2024, 41, 263. [Google Scholar] [PubMed]
  17. Jang, J.Y.; Kim, D.; Im, E.; Kim, N.D. Etoposide as a Key Therapeutic Agent in Lung Cancer: Mechanisms, Efficacy, and Emerging Strategies. Int. J. Mol. Sci. 2025, 26, 796. [Google Scholar] [CrossRef] [PubMed]
  18. Adefegha, S.A.; Oboh, G.; Oluokun, O.O. Food Bioactives: The Food Image behind the Curtain of Health Promotion and Prevention against Several Degenerative Diseases. Stud. Nat. Prod. Chem. 2022, 72, 391–421. [Google Scholar]
  19. Siddiqui, T.; Sharma, V.; Khan, M.U.; Gupta, K. Terpenoids in Essential Oils: Chemistry, Classification, and Potential Impact on Human Health and Industry. Phytomed. Plus 2024, 4, 100549. [Google Scholar]
  20. Li, Y.; Wang, J.; Li, L.; Song, W.; Li, M.; Hua, X.; Wang, Y.; Yuan, J.; Xue, Z. Natural Products of Pentacyclic Triterpenoids: From Discovery to Heterologous Biosynthesis. Nat. Prod. Rep. 2023, 40, 1303–1353. [Google Scholar]
  21. Gozari, M.; Alborz, M.; El-Seedi, H.R.; Jassbi, A.R. Chemistry, Biosynthesis and Biological Activity of Terpenoids and Meroterpenoids in Bacteria and Fungi Isolated from Different Marine Habitats. Eur. J. Med. Chem. 2021, 210, 112957. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, J.-J.; Li, W.-X.; Gao, K.; Jin, X.-J.; Yao, X.-J. Absolute Structures of Monoterpenoids with a δ-Lactone-Containing Skeleton from Ligularia hodgsonii. J. Nat. Prod. 2012, 75, 1184–1188. [Google Scholar] [CrossRef] [PubMed]
  23. Chen, C.; Chen, H.; Huang, S.; Jiang, T.; Wang, C.; Tao, Z.; He, C.; Tang, Q.; Li, P. Volatile DMNT Directly Protects Plants against Plutella Xylostella by Disrupting the Peritrophic Matrix Barrier in Insect Midgut. eLife 2021, 10, e63938. [Google Scholar] [CrossRef]
  24. Silva Santos, A.; Antunes, A.; D’Avila, L.; Bizzo, H.; Souza-Santos, L. The Use of Essential Oils and Terpenes/Terpeneoids in Cosmetic and Perfumery. Perf. Flavorist 2005, 30, 50–55. [Google Scholar]
  25. Fan, W.; Xu, Y.; Qian, M. Current Practice and Future Trends of Aroma and Flavor Research in Chinese Baijiu. In ACS Symposium Series; Guthrie, B., Beauchamp, J.D., Buettner, A., Toth, S., Qian, M.C., Eds.; American Chemical Society: Washington, DC, USA, 2019; Volume 1321, pp. 145–175. ISBN 978-0-8412-3467-3. [Google Scholar]
  26. Pang, B.; Li, J.; Eiben, C.B.; Oksen, E.; Barcelos, C.; Chen, R.; Englund, E.; Sundstrom, E.; Keasling, J.D. Lepidopteran Mevalonate Pathway Optimization in Escherichia coli Efficiently Produces Isoprenol Analogs for Next-Generation Biofuels. Metab. Eng. 2021, 68, 210–219. [Google Scholar] [CrossRef]
  27. Álvarez-Martínez, F.J.; Barrajón-Catalán, E.; Herranz-López, M.; Micol, V. Antibacterial Plant Compounds, Extracts and Essential Oils: An Updated Review on Their Effects and Putative Mechanisms of Action. Phytomedicine 2021, 90, 153626. [Google Scholar] [CrossRef]
  28. Mahizan, N.A.; Yang, S.-K.; Moo, C.-L.; Song, A.A.-L.; Chong, C.-M.; Chong, C.-W.; Abushelaibi, A.; Lim, S.-H.E.; Lai, K.-S. Terpene Derivatives as a Potential Agent against Antimicrobial Resistance (AMR) Pathogens. Molecules 2019, 24, 2631. [Google Scholar] [CrossRef]
  29. Baccouri, B.; Rajhi, I. Potential Antioxidant Activity of Terpenes. In Terpenes and Terpenoids-Recent Advances; IntechOpen: London, UK, 2021; pp. 53–62. [Google Scholar]
  30. Graßmann, J. Terpenoids as Plant Antioxidants. Vitam. Horm. 2005, 72, 505–535. [Google Scholar]
  31. Mohandas, G.G.; Kumaraswamy, M. Antioxidant Activities of Terpenoids from Thuidium tamariscellum (C. Muell.) Bosch. and Sande-Lac. a Moss. Pharmacogn. J. 2018, 10, 645–649. [Google Scholar] [CrossRef]
  32. Del Prado-Audelo, M.L.; Cortés, H.; Caballero-Florán, I.H.; González-Torres, M.; Escutia-Guadarrama, L.; Bernal-Chávez, S.A.; Giraldo-Gomez, D.M.; Magaña, J.J.; Leyva-Gómez, G. Therapeutic Applications of Terpenes on Inflammatory Diseases. Front. Pharmacol. 2021, 12, 704197. [Google Scholar] [CrossRef] [PubMed]
  33. Devi, M.; Bamrah, P.K.; Goyal, R.; Choudhary, M.; Chopra, H. Insights on the Emerging Therapeutic Potential of Terpenoids as Anti-Inflammatory Agents: A Scoping Review. J. Bio-X Res. 2024, 7, 0006. [Google Scholar] [CrossRef]
  34. Ge, J.; Liu, Z.; Zhong, Z.; Wang, L.; Zhuo, X.; Li, J.; Jiang, X.; Ye, X.-Y.; Xie, T.; Bai, R. Natural Terpenoids with Anti-Inflammatory Activities: Potential Leads for Anti-Inflammatory Drug Discovery. Bioorg. Chem. 2022, 124, 105817. [Google Scholar] [CrossRef]
  35. Roy, S.; Ghosh, A.; Majie, A.; Karmakar, V.; Das, S.; Dinda, S.C.; Bose, A.; Gorain, B. Terpenoids as Potential Phytoconstituent in the Treatment of Diabetes: From Preclinical to Clinical Advancement. Phytomedicine 2024, 129, 155638. [Google Scholar] [CrossRef]
  36. Peana, A.T.; Paolo, S.D.; Chessa, M.L.; Moretti, M.D.; Serra, G.; Pippia, P. (−)-Linalool Produces Antinociception in Two Experimental Models of Pain. Eur. J. Pharmacol. 2003, 460, 37–41. [Google Scholar] [CrossRef]
  37. El-Baba, C.; Baassiri, A.; Kiriako, G.; Dia, B.; Fadlallah, S.; Moodad, S.; Darwiche, N. Terpenoids’ Anti-Cancer Effects: Focus on Autophagy. Apoptosis 2021, 26, 491–511. [Google Scholar] [CrossRef]
  38. He, H. Potential and Challenges of Terpenoids in Cancer Therapy: Mechanistic Review and Future Perspectives. Theor. Nat. Sci. 2025, 75, 13–19. [Google Scholar] [CrossRef]
  39. Kaps, A.; Gwiazdoń, P.; Chodurek, E. Nanoformulations for Delivery of Pentacyclic Triterpenoids in Anticancer Therapies. Molecules 2021, 26, 1764. [Google Scholar] [CrossRef] [PubMed]
  40. Masyita, A.; Sari, R.M.; Astuti, A.D.; Yasir, B.; Rumata, N.R.; Emran, T.B.; Nainu, F.; Simal-Gandara, J. Terpenes and Terpenoids as Main Bioactive Compounds of Essential Oils, Their Roles in Human Health and Potential Application as Natural Food Preservatives. Food Chem. X 2022, 13, 100217. [Google Scholar] [CrossRef] [PubMed]
  41. Attri, K.; Sharda, D.; Chudasama, B.; Mahajan, R.L.; Choudhury, D. A Review on Terpenes for Treatment of Gastric Cancer: Current Status and Nanotechnology-Enabled Future. RSC Sustain. 2023, 1, 1109–1124. [Google Scholar] [CrossRef]
  42. Elsori, D.; Pandey, P.; Obaidur Rab, S.; Alharbi, A.; Sahoo, S.; Pandey, S.; Srivastava, M.; Lakhanpal, S.; Saeed, M.; Khan, F. How Effective Are Key Phytocompound Carrying Polysaccharide Nanocarriers as Anti-Breast Cancer Therapy? A Comprehensive Review of the Literature. Int. J. Nanomed. 2025, 20, 8393–8413. [Google Scholar] [CrossRef]
  43. Wang, S.; Su, R.; Nie, S.; Sun, M.; Zhang, J.; Wu, D.; Moustaid-Moussa, N. Application of Nanotechnology in Improving Bioavailability and Bioactivity of Diet-Derived Phytochemicals. J. Nutr. Biochem. 2014, 25, 363–376. [Google Scholar] [CrossRef]
  44. Kashyap, D.; Tuli, H.S.; Yerer, M.B.; Sharma, A.; Sak, K.; Srivastava, S.; Pandey, A.; Garg, V.K.; Sethi, G.; Bishayee, A. Natural Product-Based Nanoformulations for Cancer Therapy: Opportunities and Challenges. Semin. Cancer Biol. 2021, 69, 5–23. [Google Scholar] [CrossRef] [PubMed]
  45. Lagoa, R.; Silva, J.; Rodrigues, J.R.; Bishayee, A. Advances in Phytochemical Delivery Systems for Improved Anticancer Activity. Biotechnol. Adv. 2020, 38, 107382. [Google Scholar] [CrossRef]
  46. Fakhri, S.; Moradi, S.Z.; Faraji, F.; Farhadi, T.; Hesami, O.; Iranpanah, A.; Webber, K.; Bishayee, A. Current Advances in Nanoformulations of Therapeutic Agents Targeting Tumor Microenvironment to Overcome Drug Resistance. Cancer Metastasis Rev. 2023, 42, 959–1020. [Google Scholar] [CrossRef]
  47. Nahata, A.; Seng, L.S. Terpenes and Terpene Based Nanoformulations: A New Prospect for Arthritis Management. Int. J. Med. Pharm. Health Sci. 2024, 1, 196–207. [Google Scholar]
  48. Pandey, P.; Verma, M.; Sanghvi, G.; Roopashree, R.; Joshi, K.K.; Kavitha, V.; Ray, S.; Ramniwas, S.; Singh, A.; Lakhanpal, S.; et al. Plant-Derived Terpenoids Modulating Cancer Cell Metabolism and Cross-Linked Signaling Pathways: An Updated Reviews. Naunyn-Schmiedeb. Arch. Pharmacol. 2025, 398, 8313–8333. [Google Scholar] [CrossRef]
  49. Nayila, I.; Sharif, S.; Lodhi, M.S.; Ullah, R.; Alotaibi, A.; Maqbool, T.; Hameed, S. Formulation, Characterization and Evaluation of Anti-Breast Cancer Activity of 2-Carene Nanoemulsion; in Silico, in Vitro and in Vivo Study. Arab. J. Chem. 2024, 17, 105937. [Google Scholar] [CrossRef]
  50. Negi, K.; Singh, S.; Gahlot, M.S.; Tyagi, S.; Gupta, A. Terpenoids from Medicinal Plants Beneficial for Human Health Care. Int. J. Botany Stud. 2020, 5, 135–138. [Google Scholar]
  51. Evidente, A.; Kornienko, A.; Lefranc, F.; Cimmino, A.; Dasari, R.; Evidente, M.; Mathieu, V.; Kiss, R. Sesterterpenoids with Anticancer Activity. Curr. Med. Chem. 2015, 22, 3502–3522. [Google Scholar] [CrossRef]
  52. Mabou, F.D.; Yossa, I.B.N. TERPENES: Structural Classification and Biological Activities. IOSR J. Pharm. Biol. Sci. 2021, 16, 25–40. [Google Scholar]
  53. Torequl Islam, M.; Quispe, C.; Herrera-Bravo, J.; Rahaman, M.M.; Hossain, R.; Sarkar, C.; Raihan, M.A.; Chowdhury, M.M.; Uddin, S.J.; Shilpi, J.A.; et al. Activities and Molecular Mechanisms of Diterpenes, Diterpenoids, and Their Derivatives in Rheumatoid Arthritis. Evid. Based Complement. Alternat. Med. 2022, 2022, 4787643. [Google Scholar] [CrossRef]
  54. Zhang, C.; Liu, Y. Targeting Cancer with Sesterterpenoids: The New Potential Antitumor Drugs. J. Nat. Med. 2015, 69, 255–266. [Google Scholar] [CrossRef]
  55. Mukherjee, P.K. Chapter 7—Bioactive Phytocomponents and Their Analysis. In Quality Control and Evaluation of Herbal Drugs; Mukherjee, P.K., Ed.; Elsevier: Amsterdam, The Netherlands, 2019; pp. 237–328. ISBN 978-0-12-813374-3. [Google Scholar]
  56. Huang, Y.; Xie, F.-J.; Cao, X.; Li, M.-Y. Research Progress in Biosynthesis and Regulation of Plant Terpenoids. Biotechnol. Biotechnol. Equip. 2021, 35, 1799–1808. [Google Scholar] [CrossRef]
  57. Abdallah, I.I.; Quax, W.J. A Glimpse into the Biosynthesis of Terpenoids. In International Conference on Natural Resources and Life Sciences (NRLS-2016); KnE Life Sciences: Dubai, United Arab Emirates, 2017; pp. 81–98. [Google Scholar]
  58. Tholl, D. Biosynthesis and Biological Functions of Terpenoids in Plants. In Biotechnology of Isoprenoids; Schrader, J., Bohlmann, J., Eds.; Advances in Biochemical Engineering/Biotechnology; Springer International Publishing: Cham, Germany, 2015; Volume 148, pp. 63–106. ISBN 978-3-319-20106-1. [Google Scholar]
  59. Li, C.; Zha, W.; Li, W.; Wang, J.; You, A. Advances in the Biosynthesis of Terpenoids and Their Ecological Functions in Plant Resistance. Int. J. Mol. Sci. 2023, 24, 11561. [Google Scholar] [CrossRef]
  60. Nagegowda, D.A. Plant Volatile Terpenoid Metabolism: Biosynthetic Genes, Transcriptional Regulation and Subcellular Compartmentation. FEBS Lett. 2010, 584, 2965–2973. [Google Scholar] [CrossRef]
  61. Yang, W.; Chen, X.; Li, Y.; Guo, S.; Wang, Z.; Yu, X. Advances in Pharmacological Activities of Terpenoids. Nat. Prod. Commun. 2020, 15, 1934578X20903555. [Google Scholar] [CrossRef]
  62. Powder-George, Y.L.; Ludwiczuk, A.; Skalicka-Woźniak, K.; Georgiev, M.I. Chapter 10—Terpenoids. In Pharmacognosy, 2nd ed.; McCreath, S.B., Clement, Y.N., Eds.; Academic Press: Cambridge, MA, USA, 2024; pp. 253–294. ISBN 978-0-443-18657-8. [Google Scholar]
  63. Liu, Y.; Ma, X.; Liang, H.; Stephanopoulos, G.; Zhou, K. Monoterpenoid Biosynthesis by Engineered Microbes. J. Ind. Microbiol. Biotechnol. 2021, 48, kuab065. [Google Scholar] [CrossRef] [PubMed]
  64. Jiang, S.; Wang, M.; Jiang, Z.; Zafar, S.; Xie, Q.; Yang, Y.; Liu, Y.; Yuan, H.; Jian, Y.; Wang, W. Chemistry and Pharmacological Activity of Sesquiterpenoids from the Chrysanthemum Genus. Molecules 2021, 26, 3038. [Google Scholar] [CrossRef] [PubMed]
  65. Chen, Q.; Li, J.; Ma, Y.; Yuan, W.; Zhang, P.; Wang, G. Occurrence and Biosynthesis of Plant Sesterterpenes (C25), a New Addition to Terpene Diversity. Plant Commun. 2021, 2, 100184. [Google Scholar] [CrossRef]
  66. Chudzik, M.; Korzonek-Szlacheta, I.; Król, W. Triterpenes as Potentially Cytotoxic Compounds. Molecules 2015, 20, 1610–1625. [Google Scholar] [CrossRef]
  67. Maoka, T. Carotenoids as Natural Functional Pigments. J. Nat. Med. 2020, 74, 1–16. [Google Scholar] [CrossRef]
  68. Fink, J.K. Chapter 12—Terpene Resins. In Reactive Polymers Fundamentals and Applications, 2nd ed.; Fink, J.K., Ed.; Plastics Design Library; William Andrew Publishing: Oxford, UK, 2013; pp. 303–315. ISBN 978-1-4557-3149-7. [Google Scholar]
  69. Singh, A.; Debnath, R.; Sharma, A.; Saini, A.; Seni, K.; Chawla, V.; Chawla, P.A. Exploring the Potential of Terpenoids as a Possible Treatment for Cancer: Structure-Activity Relationship and Mechanistic Studies. Curr. Med. Chem. 2024, 31, 1–27. [Google Scholar] [CrossRef]
  70. Tu, S.; Hu, F.; Zhang, J.; Cai, H.; Yang, J. Research Progress on the Signaling Pathway Mechanism of Terpenoids against Breast Cancer. Discov. Oncol. 2025, 16, 433. [Google Scholar] [CrossRef]
  71. Guo, J.; Huang, M.; Hou, S.; Yuan, J.; Chang, X.; Gao, S.; Zhang, Z.; Wu, Z.; Li, J. Therapeutic Potential of Terpenoids in Cancer Treatment: Targeting Mitochondrial Pathways. Cancer Rep. 2024, 7, e70006. [Google Scholar] [CrossRef]
  72. Andreani, T.; Cheng, R.; Elbadri, K.; Ferro, C.; Menezes, T.; Dos Santos, M.R.; Pereira, C.M.; Santos, H.A. Natural Compounds-Based Nanomedicines for Cancer Treatment: Future Directions and Challenges. Drug Deliv. Transl. Res. 2024, 14, 2845–2916. [Google Scholar] [CrossRef]
  73. Mohapatra, P.; Singh, P.; Singh, D.; Sahoo, S.; Sahoo, S.K. Phytochemical Based Nanomedicine: A Panacea for Cancer Treatment, Present Status and Future Prospective. OpenNano 2022, 7, 100055. [Google Scholar] [CrossRef]
  74. Sanna, V.; Chamcheu, J.C.; Pala, N.; Mukhtar, H.; Sechi, M.; Siddiqui, I.A. Nanoencapsulation of Natural Triterpenoid Celastrol for Prostate Cancer Treatment. Int. J. Nanomed. 2015, 10, 6835–6846. [Google Scholar] [CrossRef] [PubMed]
  75. Rahat, I.; Rizwanullah, M.; Gilani, S.J.; Bin-Jummah, M.N.; Imam, S.S.; Kala, C.; Asif, M.; Alshehri, S.; Sharma, S.K. Thymoquinone Loaded Chitosan-Solid Lipid Nanoparticles: Formulation Optimization to Oral Bioavailability Study. J. Drug Deliv. Sci. Technol. 2021, 64, 102565. [Google Scholar]
  76. Lathwal, R. Development and Characterization of Ursolic Acid-Loaded Liposomes for Enhanced Bioavailability and Sustained Anticancer Drug Delivery. Int. J. Sci. Info. 2024, 1, 103–110. [Google Scholar]
  77. Kumar, P.; Singh, A.K.; Raj, V.; Rai, A.; Keshari, A.K.; Kumar, D.; Maity, B.; Prakash, A.; Maiti, S.; Saha, S. Poly(Lactic-Co-Glycolic Acid)-Loaded Nanoparticles of Betulinic Acid for Improved Treatment of Hepatic Cancer: Characterization, in Vitro and in Vivo Evaluations. Int. J. Nanomed. 2018, 13, 975–990. [Google Scholar] [CrossRef]
  78. Sun, Y.; Wang, C.; Li, X.; Lu, J.; Wang, M. Recent Advances in Drug Delivery of Celastrol for Enhancing Efficiency and Reducing the Toxicity. Front. Pharmacol. 2024, 15, 1137289. [Google Scholar] [CrossRef] [PubMed]
  79. Hafez Ghoran, S.; Calcaterra, A.; Abbasi, M.; Taktaz, F.; Nieselt, K.; Babaei, E. Curcumin-Based Nanoformulations: A Promising Adjuvant towards Cancer Treatment. Molecules 2022, 27, 5236. [Google Scholar] [CrossRef]
  80. Kaushik, N.; Borkar, S.B.; Nandanwar, S.K.; Panda, P.K.; Choi, E.H.; Kaushik, N.K. Nanocarrier Cancer Therapeutics with Functional Stimuli-Responsive Mechanisms. J. Nanobiotechnol. 2022, 20, 152. [Google Scholar] [CrossRef] [PubMed]
  81. Atriya, A.; Majee, C.; Mazumder, R.; Choudhary, A.N.; Salahuddin; Mazumder, A.; Dahiya, A.; Priya, N. Insight into the Various Approaches for the Enhancement of Bioavailabilityand Pharmacological Potency of Terpenoids: A Review. Curr. Pharm. Biotechnol. 2023, 24, 1228–1244. [Google Scholar] [CrossRef]
  82. Kumar, R.; Sharma, M. Herbal Nanomedicine Interactions to Enhance Pharmacokinetics, Pharmacodynamics, and Therapeutic Index for Better Bioavailability and Biocompatibility of Herbal Formulations. Mater. NanoSci. 2018, 5, 35–60. [Google Scholar]
  83. Lôbo, G.C.; Paiva, K.L.; Silva, A.L.G.; Simões, M.M.; Radicchi, M.A.; Báo, S.N. Nanocarriers Used in Drug Delivery to Enhance Immune System in Cancer Therapy. Pharmaceutics 2021, 13, 1167. [Google Scholar] [CrossRef]
  84. Hadkar, V.M.; Mohanty, C.; Selvaraj, C.I. Biopolymeric Nanocarriers in Cancer Therapy: Unleashing the Potency of Bioactive Anticancer Compounds for Enhancing Drug Delivery. RSC Adv. 2024, 14, 25149–25173. [Google Scholar] [CrossRef] [PubMed]
  85. Liao, Z.; Wong, S.W.; Yeo, H.L.; Zhao, Y. Smart Nanocarriers for Cancer Treatment: Clinical Impact and Safety. NanoImpact 2020, 20, 100253. [Google Scholar] [CrossRef]
  86. Lasoń, E. Topical Administration of Terpenes Encapsulated in Nanostructured Lipid-Based Systems. Molecules 2020, 25, 5758. [Google Scholar] [CrossRef]
  87. Milan, A.; Mioc, A.; Prodea, A.; Mioc, M.; Buzatu, R.; Ghiulai, R.; Racoviceanu, R.; Caruntu, F.; Şoica, C. The Optimized Delivery of Triterpenes by Liposomal Nanoformulations: Overcoming the Challenges. Int. Mol. Sci. 2022, 23, 1140. [Google Scholar] [CrossRef]
  88. Rehman, M.; Tahir, N.; Sohail, M.F.; Qadri, M.U.; Duarte, S.O.; Brandão, P.; Esteves, T.; Javed, I.; Fonte, P. Lipid-Based Nanoformulations for Drug Delivery: An Ongoing Perspective. Pharmaceutics 2024, 16, 1376. [Google Scholar] [CrossRef]
  89. Nigade, P.M.; Patil, S.L.; Tiwari, S.S. Self Emulsifying Drug Delivery System (SEDDS): A Review. Int. J. Pharm. Biol. Sci. 2012, 2, 42–52. [Google Scholar]
  90. Kumari, A.; Yadav, S.K.; Yadav, S.C. Biodegradable Polymeric Nanoparticles Based Drug Delivery Systems. Colloids Surf. B Biointerfaces 2010, 75, 1–18. [Google Scholar] [CrossRef] [PubMed]
  91. Chevalier, Y.; Bolzinger, M.-A. Emulsions Stabilized with Solid Nanoparticles: Pickering Emulsions. Colloids Surf. A Physicochem. Eng. Asp. 2013, 439, 23–34. [Google Scholar] [CrossRef]
  92. Sundar, S.; Kumar Prajapati, V. Drug Targeting to Infectious Diseases by Nanoparticles Surface Functionalized with Special Biomolecules. Curr. Med. Chem. 2012, 19, 3196–3202. [Google Scholar] [CrossRef] [PubMed]
  93. Adeola, H.A.; Sabiu, S.; Adekiya, T.A.; Aruleba, R.T.; Aruwa, C.E.; Oyinloye, B.E. Prospects of Nanodentistry for the Diagnosis and Treatment of Maxillofacial Pathologies and Cancers. Heliyon 2020, 6, e04890. [Google Scholar] [CrossRef] [PubMed]
  94. Elsori, D.; Pandey, P.; Ramniwas, S.; Kumar, R.; Lakhanpal, S.; Rab, S.O.; Siddiqui, S.; Singh, A.; Saeed, M.; Khan, F. Naringenin as Potent Anticancer Phytocompound in Breast Carcinoma: From Mechanistic Approach to Nanoformulations Based Therapeutics. Front. Pharmacol. 2024, 15, 1406619. [Google Scholar] [CrossRef]
  95. Wahi, A.; Bishnoi, M.; Raina, N.; Singh, M.A.; Verma, P.; Gupta, P.K.; Kaur, G.; Tuli, H.S.; Gupta, M. Recent Updates on Nano-Phyto-Formulations Based Therapeutic Intervention for Cancer Treatment. Oncol. Res. 2023, 32, 19. [Google Scholar] [CrossRef]
  96. Das, C.A.; Kumar, V.G.; Dhas, T.S.; Karthick, V.; Kumar, C.V. Nanomaterials in Anticancer Applications and Their Mechanism of Action-A Review. Nanomed. Nanotechnol. Biol. Med. 2023, 47, 102613. [Google Scholar] [CrossRef]
  97. Silva, B.I.; Nascimento, E.A.; Silva, C.J.; Silva, T.G.; Aguiar, J.S. Anticancer Activity of Monoterpenes: A Systematic Review. Mol. Biol. Rep. 2021, 48, 5775–5785. [Google Scholar] [CrossRef]
  98. Batista, F.A.; Fontele, S.B.C.; Santos, L.K.B.; Filgueiras, L.A.; Nascimento, S.Q.; de Castro e Sousa, J.M.; Gonçalves, J.C.R.; Mendes, A.N. Synthesis, Characterization of α-Terpineol-Loaded PMMA Nanoparticles as Proposed of Therapy for Melanoma. Mater. Today Commun. 2020, 22, 100762. [Google Scholar] [CrossRef]
  99. Khan, I.; Bahuguna, A.; Kumar, P.; Bajpai, V.K.; Kang, S.C. In Vitro and in Vivo Antitumor Potential of Carvacrol Nanoemulsion against Human Lung Adenocarcinoma A549 Cells via Mitochondrial Mediated Apoptosis. Sci. Rep. 2018, 8, 144. [Google Scholar] [CrossRef]
  100. Khan, I.; Bhardwaj, M.; Shukla, S.; Lee, H.; Oh, M.-H.; Bajpai, V.K.; Huh, Y.S.; Kang, S.C. Carvacrol Encapsulated Nanocarrier/Nanoemulsion Abrogates Angiogenesis by Downregulating COX-2, VEGF and CD31 in Vitro and in Vivo in a Lung Adenocarcinoma Model. Colloids Surf. B Biointerfaces 2019, 181, 612–622. [Google Scholar] [CrossRef]
  101. Nordin, N.; Yeap, S.K.; Rahman, H.S.; Zamberi, N.R.; Mohamad, N.E.; Abu, N.; Masarudin, M.J.; Abdullah, R.; Alitheen, N.B. Antitumor and Anti-Metastatic Effects of Citral-Loaded Nanostructured Lipid Carrier in 4T1-Induced Breast Cancer Mouse Model. Molecules 2020, 25, 2670. [Google Scholar] [CrossRef]
  102. Ahmed, T.A.; Almehmady, A.M.; Alharbi, W.S.; Alshehri, A.A.; Almughem, F.A.; Altamimi, R.M.; Alshabibi, M.A.; Omar, A.M.; El-Say, K.M. Incorporation of Perillyl Alcohol into Lipid-Based Nanocarriers Enhances the Antiproliferative Activity in Malignant Glioma Cells. Biomedicines 2023, 11, 2771. [Google Scholar] [CrossRef]
  103. Yu, H.; Ning, N.; He, F.; Xu, J.; Zhao, H.; Duan, S.; Zhao, Y. Targeted Delivery of Geraniol via Hyaluronic Acid-Conjugation Enhances Its Anti-Tumor Activity Against Prostate Cancer. Int. J. Nanomed. 2024, 19, 155–169. [Google Scholar] [CrossRef]
  104. Han, H.D.; Cho, Y.-J.; Cho, S.K.; Byeon, Y.; Jeon, H.N.; Kim, H.-S.; Kim, B.-G.; Bae, D.-S.; Lopez-Berestein, G.; Sood, A.K. Linalool-Incorporated Nanoparticles as a Novel Anticancer Agent for Epithelial Ovarian Carcinoma. Mol. Cancer Ther. 2016, 15, 618–627. [Google Scholar] [CrossRef]
  105. Li, X.; Wu, Y.; Wang, S.; Liu, J.; Zhang, T.; Wei, Y.; Zhu, L.; Bai, W.; Ye, T.; Wang, S. Menthol Nanoliposomes Enhanced Anti-Tumor Immunotherapy by Increasing Lymph Node Homing of Dendritic Cell Vaccines. Clin. Immunol. 2022, 244, 109119. [Google Scholar] [CrossRef]
  106. Abu-Izneid, T.; Rauf, A.; Shariati, M.A.; Khalil, A.A.; Imran, M.; Rebezov, M.; Uddin, M.S.; Mahomoodally, M.F.; Rengasamy, K.R. Sesquiterpenes and Their Derivatives-Natural Anticancer Compounds: An Update. Pharmacol. Res. 2020, 161, 105165. [Google Scholar] [CrossRef] [PubMed]
  107. Parvez, S.; Karole, A.; Mudavath, S.L. Fabrication, Physicochemical Characterization and In Vitro Anticancer Activity of Nerolidol Encapsulated Solid Lipid Nanoparticles in Human Colorectal Cell Line. Colloids Surf. B Biointerfaces 2022, 215, 112520. [Google Scholar] [CrossRef] [PubMed]
  108. Rasedee, A.; Abdul, A.B.; Zeenathul, N.A.; Rahman, H.; Yeap, S.; Wan Ghani, W.N.H.; Othman, H.H.; How, C.W. Zerumbone-Loaded Nanostructured Lipid Carrier Induces G2/M Cell Cycle Arrest and Apoptosis via Mitochondrial Pathway in a Human Lymphoblastic Leukemia Cell Line. Int. J. Nanomed. 2014, 9, 527–538. [Google Scholar] [CrossRef] [PubMed]
  109. Alamoudi, A.J.; Badr-Eldin, S.M.; Ahmed, O.A.; Fahmy, U.A.; Elbehairi, S.E.I.; Alfaifi, M.Y.; Asfour, H.Z.; Mohamed, G.A.; Ibrahim, S.R.; Abdel-Naim, A.B. Optimized Bilosome-Based Nanoparticles Enhance Cytotoxic and pro-Apoptotic Activity of Costunolide in LS174T Colon Cancer Cells. Biomed. Pharmacother. 2023, 168, 115757. [Google Scholar] [CrossRef]
  110. Kordi, S.; Zarghami, N.; Akbarzadeh, A.; Rahmati, Y.M.; Ghasemali, S.; Barkhordari, A.; Tozihi, M. A Comparison of the Inhibitory Effect of Nano-Encapsulated Helenalin and Free Helenalin on Telomerase Gene Expression in the Breast Cancer Cell Line, by Real-Time PCR. Artif. Cells Nanomed. Biotechnol. 2016, 44, 695–703. [Google Scholar] [CrossRef] [PubMed]
  111. Firouzi Amandi, A.; Jokar, E.; Eslami, M.; Dadashpour, M.; Rezaie, M.; Yazdani, Y.; Nejati, B. Enhanced Anti-Cancer Effect of Artemisinin- and Curcumin-Loaded Niosomal Nanoparticles against Human Colon Cancer Cells. Med. Oncol. 2023, 40, 170. [Google Scholar] [CrossRef]
  112. Lalami, Z.A.; Tafvizi, F.; Naseh, V.; Salehipour, M. Characterization and Optimization of Co-Delivery Farnesol-Gingerol Niosomal Formulation to Enhance Anticancer Activities against Breast Cancer Cells. J. Drug Deliv. Sci. Technol. 2022, 72, 103371. [Google Scholar] [CrossRef]
  113. Indumathi, T.; Suriyaprakash, J.; Alarfaj, A.A.; Hirad, A.H.; Jaganathan, R.; Mathanmohun, M. Synergistic Effects of CuO/TiO2 -chitosan-farnesol Nanocomposites: Synthesis, Characterization, Antimicrobial, and Anticancer Activities on Melanoma Cells SK-MEL-3. J. Basic Microbiol. 2024, 64, 2300505. [Google Scholar] [CrossRef] [PubMed]
  114. Mondal, J.; Khuda-Bukhsh, A.R. Cisplatin and Farnesol Co-Encapsulated PLGA Nano-Particles Demonstrate Enhanced Anti-Cancer Potential against Hepatocellular Carcinoma Cells in Vitro. Mol. Biol. Rep. 2020, 47, 3615–3628. [Google Scholar] [CrossRef]
  115. Pirali-Hamedani, Z.; Abbasi, A.; Hassan, Z.M. Synthesis of Artemether-Loaded Albumin Nanoparticles and Measurement of Their Anti-Cancer Effects. Biomedicines 2022, 10, 2713. [Google Scholar] [CrossRef]
  116. Ma, C.; Gao, L.; Song, K.; Gu, B.; Wang, B.; Pu, W.; Chen, H. Exploring the Therapeutic Potential of Diterpenes in Gastric Cancer: Mechanisms, Efficacy, and Clinical Prospects. Biomol. Biomed. 2025, 25, 1–15. [Google Scholar] [CrossRef]
  117. Zheng, Y.; Kong, F.; Liu, S.; Liu, X.; Pei, D.; Miao, X. Membrane Protein-Chimeric Liposome-Mediated Delivery of Triptolide for Targeted Hepatocellular Carcinoma Therapy. Drug Deliv. 2021, 28, 2033–2043. [Google Scholar] [CrossRef]
  118. Li, Z.; Wu, X.; Li, J.; Yao, L.; Sun, L.; Shi, Y.; Zhang, W.; Lin, J.; Liang, D.; Li, Y. Antitumor Activity of Celastrol Nanoparticles in a Xenograft Retinoblastoma Tumor Model. Int. J. Nanomed. 2012, 7, 2389. [Google Scholar] [CrossRef]
  119. Velhal, K.; Sah, P.M.; Naik, H.S.; Raut, R.; Patil, S.; Yamgar, R.; Lakkakula, J.; Uddin, I. Synergistic Nanoformulation: Streamlined One-Pot Synthesis Enhances Paclitaxel Functionalization Gold Nanoparticles for Potent Anticancer Activity. Cell Biochem. Biophy. 2025, 83, 3187–3203. [Google Scholar] [CrossRef]
  120. Borges, G.S.M.; Prazeres, P.H.D.M.; de Souza, Â.M.; Yoshida, M.I.; Vilela, J.M.C.; e Silva, A.T.M.; Oliveira, M.S.; Gomes, D.A.; Andrade, M.S.; de Souza-Fagundes, E.M. Nanostructured Lipid Carriers as a Novel Tool to Deliver Sclareol: Physicochemical Characterisation and Evaluation in Human Cancer Cell Lines. Braz. J. Pharm. Sci. 2021, 57, e18497. [Google Scholar] [CrossRef]
  121. Chatterjee, S.; Chakraborty, P.; Dutta, S.; Karak, S.; Mahalanobis, S.; Ghosh, N.; Dewanjee, S.; Sil, P.C. Formulation of Carnosic-Acid-Loaded Polymeric Nanoparticles: An Attempt to Endorse the Bioavailability and Anticancer Efficacy of Carnosic Acid against Triple-Negative Breast Cancer. ACS Appl. Bio Mater. 2024, 7, 1656–1670. [Google Scholar] [CrossRef] [PubMed]
  122. Kiruthiga, C.; Balan, D.J.; Prasath, N.H.; Manikandakrishnan, M.; Jafni, S.; Prabhu, N.M.; Pandian, S.K.; Devi, K.P. Synergistic Induction of Apoptosis in Lung Cancer Cells through Co-Delivery of PLGA Phytol/α-Bisabolol Nanoparticles. Naunyn-Schmiedeb. Arch. Pharmacol. 2024, 397, 5131–5144. [Google Scholar] [CrossRef]
  123. Zhang, Y.; Zhao, J.; Sun, J.; Huang, L.; Li, Q. Targeting Lung Cancer Initiating Cells by All-Trans Retinoic Acid-Loaded Lipid-PLGA Nanoparticles with CD133 Aptamers. Exp. Ther. Med. 2018, 16, 4639–4649. [Google Scholar] [CrossRef]
  124. Roy, P.; Das, S.; Mondal, A.; Chatterji, U.; Mukherjee, A. Nanoparticle Engineering Enhances Anticancer Efficacy of Andrographolide in MCF-7 Cells and Mice Bearing EAC. Curr. Pharmaceut. Biotechnol. 2012, 13, 2669–2681. [Google Scholar] [CrossRef]
  125. Balakarthikeyan, J.; Mayakrishnan, V.; Kannappan, P.; Al-Ghamdi, S.; Alrudian, N.A.; Alqahtani, M.S.; El-Bidawy, M.H.; Albasheer, K.; Gamil, S.; Alsanousi, N. Cetuximab-Conjugated Andrographolide Loaded Chitosan-Pectin Composite Nanoparticles for Colorectal Cancer. J. King Saud Univ. Sci. 2024, 36, 103261. [Google Scholar] [CrossRef]
  126. Shen, Y.; Ma, H. Oridonin-Loaded Lipid-Coated Calcium Phosphate Nanoparticles: Preparation, Characterization, and Application in A549 Lung Cancer. Pharm. Dev. Technol. 2022, 27, 598–605. [Google Scholar] [CrossRef]
  127. Parikh, N.R.; Mandal, A.; Bhatia, D.; Siveen, K.S.; Sethi, G.; Bishayee, A. Oleanane Triterpenoids in the Prevention and Therapy of Breast Cancer: Current Evidence and Future Perspectives. Phytochem. Rev. 2014, 13, 793–810. [Google Scholar] [CrossRef] [PubMed]
  128. Kumbham, S.; Paul, M.; Itoo, A.; Ghosh, B.; Biswas, S. Oleanolic Acid-Conjugated Human Serum Albumin Nanoparticles Encapsulating Doxorubicin as Synergistic Combination Chemotherapy in Oropharyngeal Carcinoma and Melanoma. Int. J. Pharm. 2022, 614, 121479. [Google Scholar] [CrossRef]
  129. Man, D.K.W.; Casettari, L.; Cespi, M.; Bonacucina, G.; Palmieri, G.F.; Sze, S.C.W.; Leung, G.P.H.; Lam, J.K.W.; Kwok, P.C.L. Oleanolic Acid Loaded PEGylated PLA and PLGA Nanoparticles with Enhanced Cytotoxic Activity against Cancer Cells. Mol. Pharm. 2015, 12, 2112–2125. [Google Scholar] [CrossRef]
  130. Silva, A.M.; Alvarado, H.L.; Abrego, G.; Martins-Gomes, C.; Garduño-Ramirez, M.L.; García, M.L.; Calpena, A.C.; Souto, E.B. In Vitro Cytotoxicity of Oleanolic/Ursolic Acids-Loaded in PLGA Nanoparticles in Different Cell Lines. Pharmaceutics 2019, 11, 362. [Google Scholar] [CrossRef]
  131. Lee, K.-J.; Lee, J.M.; Shin, J.-S.; Kim, D.-H.; An, J.H. Cytotoxic Effect of Oleanolic Acid-Coated Magnetic Nanoparticles on Reversal of Multidrug Resistance in A549 Lung Cancer Cells. J. Nanosci. Nanotechnol. 2016, 16, 11031–11034. [Google Scholar] [CrossRef]
  132. Zhang, H.; Li, X.; Ding, J.; Xu, H.; Dai, X.; Hou, Z.; Zhang, K.; Sun, K.; Sun, W. Delivery of Ursolic Acid (UA) in Polymeric Nanoparticles Effectively Promotes the Apoptosis of Gastric Cancer Cells through Enhanced Inhibition of Cyclooxygenase 2 (COX-2). Int. J. Pharm. 2013, 441, 261–268. [Google Scholar] [CrossRef] [PubMed]
  133. Zhou, M.; Yi, Y.; Liu, L.; Lin, Y.; Li, J.; Ruan, J.; Zhong, Z. Polymeric Micelles Loading with Ursolic Acid Enhancing Anti-Tumor Effect on Hepatocellular Carcinoma. J. Cancer 2019, 10, 5820. [Google Scholar] [CrossRef]
  134. Zare-Zardini, H.; Alemi, A.; Taheri-Kafrani, A.; Hosseini, S.A.; Soltaninejad, H.; Hamidieh, A.A.; Haghi Karamallah, M.; Farrokhifar, M.; Farrokhifar, M. Assessment of a New Ginsenoside Rh2 Nanoniosomal Formulation for Enhanced Antitumor Efficacy on Prostate Cancer: An In Vitro Study. Drug Des. Dev. Ther. 2020, 14, 3315–3324. [Google Scholar] [CrossRef] [PubMed]
  135. Rahman, M.; Beg, S.; Alharbi, K.S.; Alruwaili, N.K.; Alotaibi, N.H.; Alzarea, A.I.; Almalki, W.H.; Alenezi, S.K.; Altowayan, W.M.; Alshammari, M.S. Implications of Solid Lipid Nanoparticles of Ganoderic Acid for the Treatment and Management of Hepatocellular Carcinoma. J. Pharm. Innov. 2021, 16, 359–370. [Google Scholar] [CrossRef]
  136. Rahman, M.; Al-Ghamdi, S.A.; Alharbi, K.S.; Beg, S.; Sharma, K.; Anwar, F.; Al-Abbasi, F.A.; Kumar, V. Ganoderic Acid Loaded Nano-Lipidic Carriers Improvise Treatment of Hepatocellular Carcinoma. Drug Deliv. 2019, 26, 782–793. [Google Scholar] [CrossRef]
  137. Lu, J.; Zhang, A.; Zhang, F.; Linhardt, R.J.; Zhu, Z.; Yang, Y.; Zhang, T.; Lin, Z.; Zhang, S.; Zhao, H. Ganoderenic Acid D-Loaded Functionalized Graphene Oxide-Based Carrier for Active Targeting Therapy of Cervical Carcinoma. Biomed. Pharmacother. 2023, 164, 114947. [Google Scholar] [CrossRef]
  138. Kumari, S.; Goyal, A.; Garg, M. Phytochemistry and Pharmacological Update on Tetraterpenoids. Nat. Prod. J. 2021, 11, 617–628. [Google Scholar] [CrossRef]
  139. Zare, M.; Norouzi Roshan, Z.; Assadpour, E.; Jafari, S.M. Improving the Cancer Prevention/Treatment Role of Carotenoids through Various Nano-Delivery Systems. Crit. Rev. Food Sci. Nutr. 2021, 61, 522–534. [Google Scholar] [CrossRef]
  140. Shejawal, K.P.; Randive, D.S.; Bhinge, S.D.; Bhutkar, M.A.; Todkar, S.S.; Mulla, A.S.; Jadhav, N.R. Green Synthesis of Silver, Iron and Gold Nanoparticles of Lycopene Extracted from Tomato: Their Characterization and Cytotoxicity against COLO320DM, HT29 and Hella Cell. J. Mater. Sci. Mater. Med. 2021, 32, 19. [Google Scholar] [CrossRef]
  141. Tian, B.; Li, J.; Pang, R.; Dai, S.; Li, T.; Weng, Y.; Jin, Y.; Hua, Y. Gold Nanoparticles Biosynthesized and Functionalized Using a Hydroxylated Tetraterpenoid Trigger Gene Expression Changes and Apoptosis in Cancer Cells. ACS Appl. Mater. Interfaces 2018, 10, 37353–37363. [Google Scholar] [CrossRef] [PubMed]
  142. Bolla, P.K.; Gote, V.; Singh, M.; Yellepeddi, V.K.; Patel, M.; Pal, D.; Gong, X.; Sambalingam, D.; Renukuntla, J. Preparation and Characterization of Lutein Loaded Folate Conjugated Polymeric Nanoparticles. J. Microencapsul. 2020, 37, 502–516. [Google Scholar] [CrossRef] [PubMed]
  143. Shanmugham, V.; Subban, R. Capsanthin-Loaded Micelles: Preparation, Characterization and in Vitro Evaluation of Cytotoxicity Using MDA-MB-231 Breast Cancer Cell Line. Food Technol. Biotechnol. 2022, 60, 350–360. [Google Scholar] [CrossRef]
  144. Ravi, H.; Kurrey, N.; Manabe, Y.; Sugawara, T.; Baskaran, V. Polymeric Chitosan-Glycolipid Nanocarriers for an Effective Delivery of Marine Carotenoid Fucoxanthin for Induction of Apoptosis in Human Colon Cancer Cells (Caco-2 Cells). Mater. Sci. Eng. C 2018, 91, 785–795. [Google Scholar] [CrossRef] [PubMed]
  145. Noviendri, D.; Irwandi Jaswir, M.T.; Mohammad, F. Analysis of Apoptosis-Inducing Effect of Free Fucoxanthin and Fucoxanthin-Loaded PLGA Microsphere on Human Lung Cancer H1299 Cell Lines. Int. J. Agric. Biol. 2024, 31, 174–182. [Google Scholar]
  146. Bharathiraja, S.; Manivasagan, P.; Oh, Y.-O.; Moorthy, M.S.; Seo, H.; Bui, N.Q.; Oh, J. Astaxanthin Conjugated Polypyrrole Nanoparticles as a Multimodal Agent for Photo-Based Therapy and Imaging. Int. J. Pharm. 2017, 517, 216–225. [Google Scholar] [CrossRef]
  147. Rodenak-Kladniew, B.; Gambaro, R.; Cisneros, J.S.; Huck-Iriart, C.; Padula, G.; Castro, G.R.; Chain, C.Y.; Islan, G.A. Enhanced Anticancer Activity of Encapsulated Geraniol into Biocompatible Lipid Nanoparticles against A549 Human Lung Cancer Cells. J. Drug Deliv. Sci. Technol. 2023, 80, 104159. [Google Scholar] [CrossRef]
  148. Duan, S.; Xia, Y.; Tian, X.; Cui, J.; Zhang, X.; Yang, Q.; Zhao, T.; Lin, Y.; Zhang, F.; Zhang, X. A Multi-Bioresponsive Self-Assembled Nano Drug Delivery System Based on Hyaluronic Acid and Geraniol against Liver Cancer. Carbohydr. Polym. 2023, 310, 120695. [Google Scholar] [CrossRef]
  149. Han, W.; Shi, W.; Yao, Q.; Wu, H.; Lei, X.; Guo, G. Anticancer Activity of Zinc-Tin Oxide/Dextran/Geraniol Nanocomposites Against 1, 2-Dimethylhydrazine-Induced Colon Cancer in Rats: In Vitro and In Vivo Study. J. Drug Deliv. Sci. Technol. 2024, 98, 105819. [Google Scholar] [CrossRef]
  150. Ghanbariasad, A.; Osanloo, M.; Hatami, S.; Khaksar, S.; Zarenezhad, E.; Ranjbar, R.; Alipanah, H. Synthesis, Characterization, and Development of Alpha-Pinene Nanoemulsion as an Apoptotic Inducer with Cytotoxicity Activity on Human Melanoma and Breast Cancer. Chem. Pap. 2024, 78, 1181–1191. [Google Scholar] [CrossRef]
  151. Rahmani, H.; Ghanbariasad, A.; Meshkibaf, M.H.; Molazade, A.; Heiran, R.; Safari, M.; Osanloo, M. Chitosan Nanoparticles Containing α-Pinene and Rosmarinus officinalis L. Essential Oil: Effects on Human Melanoma Cells’ Viability and Expression of Apoptosis-Involved Genes. Polym. Bull. 2024, 81, 2505–2523. [Google Scholar] [CrossRef]
  152. Jabir, M.S.; Taha, A.A.; Sahib, U.I.; Taqi, Z.J.; Al-Shammari, A.M.; Salman, A.S. Novel of Nano Delivery System for Linalool Loaded on Gold Nanoparticles Conjugated with CALNN Peptide for Application in Drug Uptake and Induction of Cell Death on Breast Cancer Cell Line. Mater. Sci. Eng. C. 2019, 94, 949–964. [Google Scholar] [CrossRef]
  153. Rodenak-Kladniew, B.; Islan, G.A.; de Bravo, M.G.; Durán, N.; Castro, G.R. Design, Characterization and in Vitro Evaluation of Linalool-Loaded Solid Lipid Nanoparticles as Potent Tool in Cancer Therapy. Colloids Surf. B Biointerfaces 2017, 154, 123–132. [Google Scholar] [CrossRef]
  154. Jabir, M.; Sahib, U.I.; Taqi, Z.; Taha, A.; Sulaiman, G.; Albukhaty, S.; Al-Shammari, A.; Alwahibi, M.; Soliman, D.; Dewir, Y.H.; et al. Linalool-Loaded Glutathione-Modified Gold Nanoparticles Conjugated with CALNN Peptide as Apoptosis Inducer and NF-κB Translocation Inhibitor in SKOV-3 Cell Line. Int. J. Nanomed. 2020, 15, 9025–9047. [Google Scholar] [CrossRef]
  155. Jalali, A.; Bari, I.J.; Salehzadeh, A. Menthol Conjugated Magnetic Iron Oxide Nanoparticles Induce Apoptosis and Increase Caspase-8 Gene Expression in Gastric Cancer Cell Line. BioNanoScience 2024, 14, 5276–5285. [Google Scholar] [CrossRef]
  156. Osanloo, M.; Sedaghati, F.; Zarenezhad, E.; Amani, A. Novel Nanocarrier for Melanoma Treatment: Chitosan-Gum Arabic Nanoparticles Containing Menthol. BioNanoScience 2024, 14, 2638–2648. [Google Scholar] [CrossRef]
  157. Alipanah, H.; Farjam, M.; Zarenezhad, E.; Roozitalab, G.; Osanloo, M. Chitosan Nanoparticles Containing Limonene and Limonene-Rich Essential Oils: Potential Phytotherapy Agents for the Treatment of Melanoma and Breast Cancers. BMC Complement. Med. Ther. 2021, 21, 186. [Google Scholar] [CrossRef]
  158. Swathi, B.; Thangaraj, R.; Gayathri, D.; Subha, V.; Thiyagarajan, P.; Thiruvengadam, M.; Venkidasamy, B. Green Synthesis of β-Caryophyllene-Coated Silver Nanoparticles: Biophysical Characterization and Evaluation of Antibacterial, Cytotoxic, and Larvicidal Activities. BioNanoScience 2025, 15, 102. [Google Scholar] [CrossRef]
  159. Kamaraj, C.; Balasubramani, G.; Siva, C.; Raja, M.; Balasubramanian, V.; Raja, R.K.; Tamilselvan, S.; Benelli, G.; Perumal, P. Ag Nanoparticles Synthesized Using β-Caryophyllene Isolated from Murraya Koenigii: Antimalarial (Plasmodium Falciparum 3D7) and Anticancer Activity (A549 and HeLa Cell Lines). J. Clust. Sci. 2017, 28, 1667–1684. [Google Scholar] [CrossRef]
  160. Natesan, S.; Ponnusamy, C.; Sugumaran, A.; Chelladurai, S.; Palaniappan, S.S.; Palanichamy, R. Artemisinin Loaded Chitosan Magnetic Nanoparticles for the Efficient Targeting to the Breast Cancer. Int. J. Biol. Macromol. 2017, 104, 1853–1859. [Google Scholar] [CrossRef] [PubMed]
  161. Zhang, Y.J.; Gallis, B.; Taya, M.; Wang, S.; Ho, R.J.; Sasaki, T. pH-Responsive Artemisinin Derivatives and Lipid Nanoparticle Formulations Inhibit Growth of Breast Cancer Cells in Vitro and Induce down-Regulation of HER Family Members. PLoS ONE 2013, 8, e59086. [Google Scholar] [CrossRef]
  162. Eslami Vaghar, M.; Dadashpour, M.; Derakhshan, E.; Vahedian, V.; Shahrtash, S.A.; Firouzi Amandi, A.; Eslami, M.; Hasannia, M.; Mehrabi, Z.; Roshangar, L. Artemisinin-Loaded Mesoporous Silica Nanoparticles/Electrospun Poly(Lactic-Co-Glycolic Acid) Composite Nanofibers for Enhanced Anticancer Efficiency in Breast Cancer Cells. Cancer Nanotechnol. 2024, 15, 58. [Google Scholar] [CrossRef]
  163. Mickymaray, S.; Al Aboody, M.S.; Eraqi, M.M.; Alhoqail, W.A.; Alothaim, A.S.; Suresh, K.; Arulselvan, P. Chitosan-Encapsulated Nickel Oxide, Tin Dioxide, and Farnesol Nanoparticles: Antimicrobial and Anticancer Properties in Breast Cancer Cells. Int. J. Biol. Macromol. 2023, 248, 125799. [Google Scholar] [CrossRef]
  164. Li, H.; Qu, X.; Qian, W.; Song, Y.; Wang, C.; Liu, W. Andrographolide-loaded Solid Lipid Nanoparticles Enhance Anti-cancer Activity against Head and Neck Cancer and Precancerous Cells. Oral Dis. 2022, 28, 142–149. [Google Scholar] [CrossRef] [PubMed]
  165. Lavanya, S.K.; Patil, S.; Sivapatham, S. Synthesis and Characterization of Andrographolide: Albumin Nanoparticles and Their Anticancer Activity in HeLa Cancer Cell Line. Int. J. Nutr. Pharmacol. Neurol. Dis. 2024, 14, 137–141. [Google Scholar] [CrossRef]
  166. Cosco, D.; Mare, R.; Paolino, D.; Salvatici, M.C.; Cilurzo, F.; Fresta, M. Sclareol-Loaded Hyaluronan-Coated PLGA Nanoparticles: Physico-Chemical Properties and in Vitro Anticancer Features. Int. J. Biol. Macromol. 2019, 132, 550–557. [Google Scholar] [CrossRef]
  167. Hamishehkar, H.; Bahadori, M.B.; Vandghanooni, S.; Eskandani, M.; Nakhlband, A.; Eskandani, M. Preparation, Characterization and Anti-Proliferative Effects of Sclareol-Loaded Solid Lipid Nanoparticles on A549 Human Lung Epithelial Cancer Cells. J. Drug Deliv. Sci. Technol. 2018, 45, 272–280. [Google Scholar] [CrossRef]
  168. Khella, K.F.; Abd El Maksoud, A.I.; Hassan, A.; Abdel-Ghany, S.E.; Elsanhoty, R.M.; Aladhadh, M.A.; Abdel-Hakeem, M.A. Carnosic Acid Encapsulated in Albumin Nanoparticles Induces Apoptosis in Breast and Colorectal Cancer Cells. Molecules 2022, 27, 4102. [Google Scholar] [CrossRef]
  169. Dogan, M. Analysis of the Mechanisms Underlying the Anticancer and Biological Activity of Retinoic Acid and Chitosan Nanoparticles Containing Retinoic Acid. Med. Oncol. 2024, 41, 251. [Google Scholar] [CrossRef]
  170. Rajeshkumar, R.R.; Pavadai, P.; Panneerselvam, T.; Pandian, S.R.K.; Kumar, A.S.K.; Maszczyk, P.; Babkiewicz, E.; Kabilan, S.J.; Kunjiappan, S. Enhanced Delivery of Retinoic Acid to Breast Cancer Cells by Folate Receptor-Targeted Folic Acid-Conjugated Glutenin Nanoparticles for Promising Treatment of Breast Cancer. J. Polym. Environ. 2024, 32, 2120–2139. [Google Scholar] [CrossRef]
  171. Akanda, M.H.; Rai, R.; Slipper, I.J.; Chowdhry, B.Z.; Lamprou, D.; Getti, G.; Douroumis, D. Delivery of Retinoic Acid to LNCap Human Prostate Cancer Cells Using Solid Lipid Nanoparticles. Int. J. Pharm. 2015, 493, 161–171. [Google Scholar] [CrossRef]
  172. Kotelevets, L.; Chastre, E.; Caron, J.; Mougin, J.; Bastian, G.; Pineau, A.; Walker, F.; Lehy, T.; Desmaële, D.; Couvreur, P. A Squalene-Based Nanomedicine for Oral Treatment of Colon Cancer. Cancer Res. 2017, 77, 2964–2975. [Google Scholar] [CrossRef] [PubMed]
  173. Bidooki, S.H.; Quero, J.; Sánchez-Marco, J.; Herrero-Continente, T.; Marmol, I.; Lasheras, R.; Sebastian, V.; Arruebo, M.; Osada, J.; Rodriguez-Yoldi, M.J. Squalene in Nanoparticles Improves Antiproliferative Effect on Human Colon Carcinoma Cells Through Apoptosis by Disturbances in Redox Balance. Int. J. Mol. Sci. 2024, 25, 13048. [Google Scholar] [CrossRef]
  174. Ravichandran, S.; Kandaswamy, K.; Muthu, K. Evaluation of Lupeol-Chitosan Nanoparticles Infused Cellulose Acetate Membranes for Enhanced in-Vitro Anticancer and Antidiabetic Activities. Chemosphere 2024, 351, 141149. [Google Scholar] [CrossRef]
  175. Zhang, J.; Hu, X.; Zheng, G.; Yao, H.; Liang, H. In Vitro and in Vivo Antitumor Effects of Lupeol-Loaded Galactosylated Liposomes. Drug Deliv. 2021, 28, 709–718. [Google Scholar] [CrossRef] [PubMed]
  176. Shukla, V.N.; Mehata, A.K.; Setia, A.; Kumari, P.; Mahto, S.K.; Muthu, M.S.; Mishra, S.K. EGFR Targeted Albumin Nanoparticles of Oleanolic Acid: In Silico Screening of Nanocarrier, Cytotoxicity and Pharmacokinetics for Lung Cancer Therapy. Int. J. Biol. Macromol. 2023, 246, 125719. [Google Scholar] [CrossRef]
  177. Wang, S.; Meng, X.; Dong, Y. Ursolic Acid Nanoparticles Inhibit Cervical Cancer Growth in Vitro and in Vivo via Apoptosis Induction. Int. J. Oncol. 2017, 50, 1330–1340. [Google Scholar] [CrossRef]
  178. Payomhom, P.; Panyain, N.; Sakonsinsiri, C.; Wongtrakoongate, P.; Lertsuwan, K.; Pissuwan, D.; Katewongsa, K.P. Chitosan-Coated Poly(Lactic-Co-Glycolic Acid) Nanoparticles Loaded with Ursolic Acid for Breast Cancer Therapy. ACS Appl. Nano Mater. 2024, 7, 5383–5395. [Google Scholar] [CrossRef]
  179. Markowski, A.; Migdał, P.; Zygmunt, A.; Zaremba-Czogalla, M.; Gubernator, J. Evaluation of the in Vitro Cytotoxic Activity of Ursolic Acid PLGA Nanoparticles against Pancreatic Ductal Adenocarcinoma Cell Lines. Materials 2021, 14, 4917. [Google Scholar] [CrossRef] [PubMed]
  180. Wang, L.; Wang, S.; Chen, R.; Wang, Y.; Li, H.; Wang, Y.; Chen, M. Oridonin Loaded Solid Lipid Nanoparticles Enhanced Antitumor Activity in MCF-7 Cells. J. Nanomater. 2014, 2014, 903646. [Google Scholar] [CrossRef]
  181. Hong, C.; Wang, D.; Liang, J.; Guo, Y.; Zhu, Y.; Xia, J.; Qin, J.; Zhan, H.; Wang, J. Novel Ginsenoside-Based Multifunctional Liposomal Delivery System for Combination Therapy of Gastric Cancer. Theranostics 2019, 9, 4437. [Google Scholar] [CrossRef]
  182. Goswami, A.; Patel, N.; Bhatt, V.; Raval, M.; Kundariya, M.; Sheth, N. Lycopene Loaded Polymeric Nanoparticles for Prostate Cancer Treatment: Formulation, Optimization Using Box-Behnken Design and Cytotoxicity Studies. J. Drug Deliv. Sci. Technol. 2022, 67, 102930. [Google Scholar] [CrossRef]
  183. Chen, B.-H.; Huang, R.-F.S.; Wei, Y.-J.; Stephen Inbaraj, B. Inhibition of Colon Cancer Cell Growth by Nanoemulsion Carrying Gold Nanoparticles And&nbsp;Lycopene. Int. J. Nanomed. 2015, 10, 2823–2846. [Google Scholar] [CrossRef]
  184. Hsu, H.J.; Huang, R.F.; Kao, T.H.; Inbaraj, B.S.; Chen, B.H. Preparation of Carotenoid Extracts and Nanoemulsions from Lycium barbarum L. and Their Effects on Growth of HT-29 Colon Cancer Cells. Nanotechnology 2017, 28, 135103. [Google Scholar] [CrossRef] [PubMed]
  185. Bulatao, B.P.; Nalinratana, N.; Jantaratana, P.; Vajragupta, O.; Rojsitthisak, P.; Rojsitthisak, P. Lutein-Loaded Chitosan/Alginate-Coated Fe3O4 Nanoparticles as Effective Targeted Carriers for Breast Cancer Treatment. Int. J. Biol. Macromol. 2023, 242, 124673. [Google Scholar] [CrossRef]
  186. Sui, Y.; Gu, Y.; Lu, Y.; Yu, C.; Zheng, J.; Qi, H. Fucoxanthin@ Polyvinylpyrrolidone Nanoparticles Promoted Oxidative Stress-Induced Cell Death in Caco-2 Human Colon Cancer Cells. Mar. Drugs 2021, 19, 92. [Google Scholar] [CrossRef]
  187. Hwang, E.J.; Jeong, Y.-I.; Lee, K.-J.; Yu, Y.-B.; Ohk, S.-H.; Lee, S.-Y. Anticancer Activity of Astaxanthin-Incorporated Chitosan Nanoparticles. Molecules 2024, 29, 529. [Google Scholar] [CrossRef]
  188. Bharathiraja, S.; Manivasagan, P.; Quang Bui, N.; Oh, Y.-O.; Lim, I.G.; Park, S.; Oh, J. Cytotoxic Induction and Photoacoustic Imaging of Breast Cancer Cells Using Astaxanthin-Reduced Gold Nanoparticles. Nanomaterials 2016, 6, 78. [Google Scholar] [CrossRef]
  189. Jain, A.; Sharma, G.; Thakur, K.; Raza, K.; Shivhare, U.S.; Ghoshal, G.; Katare, O.P. Beta-Carotene-Encapsulated Solid Lipid Nanoparticles (BC-SLNs) as Promising Vehicle for Cancer: An Investigative Assessment. AAPS PharmSciTech 2019, 20, 100. [Google Scholar] [CrossRef]
  190. Dutta, R.S.; Elhassan, G.O.; Devi, T.B.; Bhattacharjee, B.; Singh, M.; Jana, B.K.; Sahu, S.; Mazumder, B.; Sahu, R.K.; Khan, J. Enhanced Efficacy of β-Carotene Loaded Solid Lipid Nanoparticles Optimized and Developed via Central Composite Design on Breast Cancer Cell Lines. Heliyon 2024, 10, e28457. [Google Scholar] [CrossRef]
  191. Sanati, P.; Chua, L.S.; Nasiri, R.; Hashemi, S.-S. Nanoencapsulation of Andrographolide Rich Extract for the Inhibition of Cervical and Neuroblastoma Cancer Cells. J. Biomed. Nanotechnol. 2020, 16, 1370–1380. [Google Scholar] [CrossRef]
  192. Liu, X.; Dong, S.; Dong, M.; Li, Y.; Sun, Z.; Zhang, X.; Wang, Y.; Teng, L.; Wang, D. Transferrin-Conjugated Liposomes Loaded with Carnosic Acid Inhibit Liver Cancer Growth by Inducing Mitochondria-Mediated Apoptosis. Int. J. Pharm. 2021, 607, 121034. [Google Scholar] [CrossRef] [PubMed]
  193. Zhao, Y.; Xiao, W.; Peng, W.; Huang, Q.; Wu, K.; Evans, C.E.; Liu, X.; Jin, H. Oridonin-Loaded Nanoparticles Inhibit Breast Cancer Progression through Regulation of ROS-Related Nrf2 Signaling Pathway. Front. Bioeng. Biotechnol. 2021, 9, 600579. [Google Scholar] [CrossRef] [PubMed]
  194. Jin, H.; Pi, J.; Yang, F.; Jiang, J.; Wang, X.; Bai, H.; Shao, M.; Huang, L.; Zhu, H.; Yang, P. Folate-Chitosan Nanoparticles Loaded with Ursolic Acid Confer Anti-Breast Cancer Activities In Vitro and In Vivo. Sci. Rep. 2016, 6, 30782. [Google Scholar] [CrossRef]
  195. Bano, S.; Ahmed, F.; Khan, F.; Chaudhary, S.C.; Samim, M. Targeted Delivery of Thermoresponsive Polymeric Nanoparticle-Encapsulated Lycopene: In Vitro Anticancer Activity and Chemopreventive Effect on Murine Skin Inflammation and Tumorigenesis. RSC Adv. 2020, 10, 16637–16649. [Google Scholar] [CrossRef]
  196. Kowshik, J.; Nivetha, R.; Ranjani, S.; Venkatesan, P.; Selvamuthukumar, S.; Veeravarmal, V.; Nagini, S. Astaxanthin Inhibits Hallmarks of Cancer by Targeting the PI3K/NF-κΒ/STAT3 Signalling Axis in Oral Squamous Cell Carcinoma Models. IUBMB Life 2019, 71, 1595–1610. [Google Scholar] [CrossRef]
  197. Azhar, F.; Naureen, H.; Shahnaz, G.; Hamdani, S.D.A.; Kiani, M.H.; Khattak, S.; Manna, M.K.; Babar, M.M.; Rajadas, J.; Rahdar, A. Development of Chitosan Based β-Carotene Mucoadhesive Formulation for Skin Cancer Treatment. Int. J. Biol. Macromol. 2023, 253, 126659. [Google Scholar] [CrossRef]
  198. Joshi, M.; Pathak, K.; Dhaneshwar, S. Nanotechnology-Based Strategies for Effective Delivery of Phytoconstituents for the Management of Rheumatoid Arthritis. Pharmacol. Res. Mod. Chin. Med. 2022, 2, 100061. [Google Scholar] [CrossRef]
  199. Ahmad, A.; Imran, M.; Sharma, N. Precision Nanotoxicology in Drug Development: Current Trends and Challenges in Safety and Toxicity Implications of Customized Multifunctional Nanocarriers for Drug-Delivery Applications. Pharmaceutics 2022, 14, 2463. [Google Scholar] [CrossRef]
  200. Jeevanandam, J.; San Chan, Y.; Danquah, M.K. Nano-Formulations of Drugs: Recent Developments, Impact and Challenges. Biochimie 2016, 128, 99–112. [Google Scholar] [CrossRef]
  201. Chaurasiya, B.; Mahanty, A.; Roy, D.; Shen, Y.; Tu, J.; Sun, C. Influence of Tumor Microenvironment on the Distribution and Elimination of Nano-Formulations. Curr. Drug Metabol. 2016, 17, 783–798. [Google Scholar] [CrossRef]
  202. Teja, P.K.; Mithiya, J.; Kate, A.S.; Bairwa, K.; Chauthe, S.K. Herbal Nanomedicines: Recent Advancements, Challenges, Opportunities and Regulatory Overview. Phytomedicine 2022, 96, 153890. [Google Scholar] [CrossRef]
  203. Kumari, S.; Goyal, A.; Sönmez Gürer, E.; Algın Yapar, E.; Garg, M.; Sood, M.; Sindhu, R.K. Bioactive Loaded Novel Nano-Formulations for Targeted Drug Delivery and Their Therapeutic Potential. Pharmaceutics 2022, 14, 1091. [Google Scholar] [CrossRef] [PubMed]
  204. Vandana, K.; Kumar, V.; Rathour, A. Advancements In Herbal-Nano Formulations: A Systematic Review. Educ. Adm. Theory Pract. 2024, 30, 11099–11109. [Google Scholar]
  205. Shrihastini, V.; Muthuramalingam, P.; Adarshan, S.; Sujitha, M.; Chen, J.-T.; Shin, H.; Ramesh, M. Plant Derived Bioactive Compounds, Their Anti-Cancer Effects and in Silico Approaches as an Alternative Target Treatment Strategy for Breast Cancer: An Updated Overview. Cancers 2021, 13, 6222. [Google Scholar] [CrossRef] [PubMed]
  206. Kour, S.; Biswas, I.; Sheoran, S.; Arora, S.; Sheela, P.; Duppala, S.K.; Murthy, D.K.; Pawar, S.C.; Singh, H.; Kumar, D. Artificial Intelligence and Nanotechnology for Cervical Cancer Treatment: Current Status and Future Perspectives. J. Drug Deliv. Sci. Technol. 2023, 83, 104392. [Google Scholar] [CrossRef]
  207. Heydari, S.; Masoumi, N.; Esmaeeli, E.; Ayyoubzadeh, S.M.; Ghorbani-Bidkorpeh, F.; Ahmadi, M. Artificial Intelligence in Nanotechnology for Treatment of Diseases. J. Drug Target. 2024, 32, 1247–1266. [Google Scholar] [CrossRef]
Figure 1. Schematic illustration of various biological applications of terpenoids.
Figure 1. Schematic illustration of various biological applications of terpenoids.
Cancers 17 03013 g001
Figure 2. The chemical structures of different classes of terpenoids. Chemical structures were drawn using ChemAxon’s MarvinSketch software (version 17.21.0).
Figure 2. The chemical structures of different classes of terpenoids. Chemical structures were drawn using ChemAxon’s MarvinSketch software (version 17.21.0).
Cancers 17 03013 g002
Figure 3. Terpenoid biosynthesis via the cytosolic MVA and plastidic MEP pathways, both producing isoprenoid precursors as building blocks for diverse terpenoids. Abbreviations: DMAPP, dimethylallyl diphosphate; DXP, 1-deoxy-D-xylulose 5-phosphate; DXR, 1-deoxy-D-xylulose 5-phosphate reductoisomerase; FPP, farnesyl diphosphate; FPPS, farnesyl diphosphate synthase; GGPP, geranylgeranyl diphosphate; GGPPS, geranylgeranyl diphosphate synthase; GPP, geranyl diphosphate; GPPS, geranyl diphosphate synthase; HMGR, 3-hydroxy-3-methylglutaryl-CoA reductase; HMGS, 3-hydroxy-3-methylglutaryl-CoA synthase; IDI, isopentenyl diphosphate isomerase; IPP, isopentenyl diphosphate; MEP, methylerythritol phosphate; MVA, mevalonate; MK, mevalonate kinase; MDD, mevalonate diphosphate decarboxylase; OSCs, oxidosqualene cyclases; PMK, phosphomevalonate kinase; SE, squalene epoxidase; TPS, terpene synthase.
Figure 3. Terpenoid biosynthesis via the cytosolic MVA and plastidic MEP pathways, both producing isoprenoid precursors as building blocks for diverse terpenoids. Abbreviations: DMAPP, dimethylallyl diphosphate; DXP, 1-deoxy-D-xylulose 5-phosphate; DXR, 1-deoxy-D-xylulose 5-phosphate reductoisomerase; FPP, farnesyl diphosphate; FPPS, farnesyl diphosphate synthase; GGPP, geranylgeranyl diphosphate; GGPPS, geranylgeranyl diphosphate synthase; GPP, geranyl diphosphate; GPPS, geranyl diphosphate synthase; HMGR, 3-hydroxy-3-methylglutaryl-CoA reductase; HMGS, 3-hydroxy-3-methylglutaryl-CoA synthase; IDI, isopentenyl diphosphate isomerase; IPP, isopentenyl diphosphate; MEP, methylerythritol phosphate; MVA, mevalonate; MK, mevalonate kinase; MDD, mevalonate diphosphate decarboxylase; OSCs, oxidosqualene cyclases; PMK, phosphomevalonate kinase; SE, squalene epoxidase; TPS, terpene synthase.
Cancers 17 03013 g003
Figure 4. Anticancer mechanism of terpenoids. (A) Terpenoids trigger cell cycle arrest and apoptosis via p53, Bax/Bak, and caspases. (B) Terpenoids promote autophagy via AMPK/ROS and inhibit PI3K/Akt/mTOR. Abbreviations: AMPK, AMP-activated protein kinase; Apaf1, apoptotic protease activating factor 1; ATG, autophagy-related protein; Bax, Bcl-2-associated X protein; Bak, Bcl-2 homologous antagonist/killer; Bcl-2, B-cell lymphoma 2; Bcl-xL, B-cell lymphoma-extra-large; c-Jun, cellular Jun proto-oncogene; c-Met, mesenchymal–epithelial transition factor; Cdc2, cell division cycle protein 2; Cdk1, cyclin-dependent kinase 1; Cyt. c, cytochrome c; ERK, extracellular signal-regulated kinase; Glut-1, glucose transporter 1; HSF-1, heat shock factor 1; HSP70, heat shock protein 70; JAK2/STAT3, Janus kinase 2/signal transducer and activator of transcription 3; JNK, c-Jun N-terminal kinase; LC3II, microtubule-associated protein 1A/1B-light chain 3; MAPK, mitogen-activated protein kinase; MMP-9, matrix metalloproteinase-9; mTOR, mammalian target of rapamycin; NF-κβ, nuclear factor-κβ; PI3K, phosphatidylinositol-3-kinase; ROS, reactive oxygen species.
Figure 4. Anticancer mechanism of terpenoids. (A) Terpenoids trigger cell cycle arrest and apoptosis via p53, Bax/Bak, and caspases. (B) Terpenoids promote autophagy via AMPK/ROS and inhibit PI3K/Akt/mTOR. Abbreviations: AMPK, AMP-activated protein kinase; Apaf1, apoptotic protease activating factor 1; ATG, autophagy-related protein; Bax, Bcl-2-associated X protein; Bak, Bcl-2 homologous antagonist/killer; Bcl-2, B-cell lymphoma 2; Bcl-xL, B-cell lymphoma-extra-large; c-Jun, cellular Jun proto-oncogene; c-Met, mesenchymal–epithelial transition factor; Cdc2, cell division cycle protein 2; Cdk1, cyclin-dependent kinase 1; Cyt. c, cytochrome c; ERK, extracellular signal-regulated kinase; Glut-1, glucose transporter 1; HSF-1, heat shock factor 1; HSP70, heat shock protein 70; JAK2/STAT3, Janus kinase 2/signal transducer and activator of transcription 3; JNK, c-Jun N-terminal kinase; LC3II, microtubule-associated protein 1A/1B-light chain 3; MAPK, mitogen-activated protein kinase; MMP-9, matrix metalloproteinase-9; mTOR, mammalian target of rapamycin; NF-κβ, nuclear factor-κβ; PI3K, phosphatidylinositol-3-kinase; ROS, reactive oxygen species.
Cancers 17 03013 g004
Figure 5. Overview of the various advantages of nanoformulated terpenoid-based anticancer drugs.
Figure 5. Overview of the various advantages of nanoformulated terpenoid-based anticancer drugs.
Cancers 17 03013 g005
Figure 6. Schematic representation of different nanocarriers used in targeted anticancer drug delivery.
Figure 6. Schematic representation of different nanocarriers used in targeted anticancer drug delivery.
Cancers 17 03013 g006
Figure 7. Schematic diagram showing the mechanisms of action of nanoformulated terpenoids.
Figure 7. Schematic diagram showing the mechanisms of action of nanoformulated terpenoids.
Cancers 17 03013 g007
Figure 8. Schematic diagram of a synergistic approach combining nanoformulated terpenoids with advanced bioinformatics tools and artificial intelligence to optimize cancer therapy.
Figure 8. Schematic diagram of a synergistic approach combining nanoformulated terpenoids with advanced bioinformatics tools and artificial intelligence to optimize cancer therapy.
Cancers 17 03013 g008
Table 2. Comparative overview of nanocarriers for terpenoid delivery in cancer therapy.
Table 2. Comparative overview of nanocarriers for terpenoid delivery in cancer therapy.
Type of NanocarrierRoute of
Administration
AdvantagesDisadvantagesRef.
Self-emulsifying drug delivery systemsOralEnhance solubility and absorption of hydrophobic terpenoidsLimited drug loading and requires high surfactant content[89]
Polymeric nanoparticlesOral and IVSustained/controlled release; biodegradable; and targeted deliveryComplex synthesis; morphology and size dependency; and burst release possible[90]
Pickering emulsionsOralSurfactant-free; good physical stabilityScale-up challenges[91]
LiposomesOral, IV, and topicalEncapsulate both hydrophilic and lipophilic terpenoids; biocompatible; enhance solubility and systemic availabilityStability issues; drug leakage; and high cost[87]
Solid lipid nanoparticles/nanostructured lipid carriersOral and IVEncapsulate hydrophobic terpenoids; high stability; controlled release; and reduced toxicityLimited drug loading and risk of drug expulsion[86]
Ethosomes, phytosomes, niosomes, and invasomesOral, transdermal, and topicalEnhance skin permeability, absorption, and stabilityLimited systemic application and variable reproducibility[86,87]
Inorganic nanoparticlesIVEasy surface modification and theranostic potentialLong-term toxicity concerns and poor biodegradability[83]
Abbreviations: IV, intravenous.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sharmila, A.; Bhadra, P.; Kishore, C.; Selvaraj, C.I.; Kavalakatt, J.; Bishayee, A. Nanoformulated Terpenoids in Cancer: A Review of Therapeutic Applications, Mechanisms, and Challenges. Cancers 2025, 17, 3013. https://doi.org/10.3390/cancers17183013

AMA Style

Sharmila A, Bhadra P, Kishore C, Selvaraj CI, Kavalakatt J, Bishayee A. Nanoformulated Terpenoids in Cancer: A Review of Therapeutic Applications, Mechanisms, and Challenges. Cancers. 2025; 17(18):3013. https://doi.org/10.3390/cancers17183013

Chicago/Turabian Style

Sharmila, Arunagiri, Priyanka Bhadra, Chandra Kishore, Chinnadurai Immanuel Selvaraj, Joachim Kavalakatt, and Anupam Bishayee. 2025. "Nanoformulated Terpenoids in Cancer: A Review of Therapeutic Applications, Mechanisms, and Challenges" Cancers 17, no. 18: 3013. https://doi.org/10.3390/cancers17183013

APA Style

Sharmila, A., Bhadra, P., Kishore, C., Selvaraj, C. I., Kavalakatt, J., & Bishayee, A. (2025). Nanoformulated Terpenoids in Cancer: A Review of Therapeutic Applications, Mechanisms, and Challenges. Cancers, 17(18), 3013. https://doi.org/10.3390/cancers17183013

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop