ABSTRACT
The present study focuses on the nanoscale formulation technique for pharmacological agent delivery, avoiding the shortcomings of conventional protocols, such as rapid pharmaceutical clearance, limited site-specific accuracy and reduced biological accessibility. The goal of this is that because nanoscale particles can improve stability, control pharmaceutical release and increase penetration, they have emerged as a promising drug delivery technology. Targeted therapies for conditions like septicemia and bronchial asthma can be developed by attaching particular molecular binders to nanoscale particles. This reduces toxicity while maximizing therapeutic efficacy. In summary, this analysis highlights the medicinal delivery potential of nanoscale formulations, including the mechanisms of action of botanical constituents such as flavonoids and triterpenoidal compounds. It also implies that the administration of inflammatory conditions may benefit greatly from the use of nanoscale particles loaded with these compounds.
INTRODUCTION
Defending against harmful stimuli like allergens or damaged tissues, inflammatory responses can also result from uncontrollably responding to a range of illnesses, including infections from bacteria, metabolic diseases, cardiac abnormalities and malignant tumours; it is illustrated in (Figure 1). These conditions have a significant financial burden on social systems (Tables 1 and 2). While there are many different therapeutic options to reduce inflammation, such as corticosteroids, non-corticosteroids and immunomodulatory drugs, they are frequently harmful when taken at the higher dosages needed to achieve therapeutic plasma concentrations (Ghasemianet al., 2016; Bagadet al., 2013; Lópezet al., 2021). Therefore, investigating phytogenic treatments is essential to overcome the drawbacks of synthetic pharmaceuticals. Highly valued for their taste, scent, or medicinal properties, botanical substances have been utilized to treat a range of illnesses and metabolic issues in addition to lowering inflammation and neutralizing reactive oxygen species. The purpose of this review is to assess botanical ingredients’ ability to reduce inflammation in a variety of disorders, emphasizing their benefits over traditional treatment approaches and investigating their potential as effective substitutes for treating inflammatory diseases.

Figure 1:
A graphic representation of the various forms of both acute and chronic inflammation.
Sl. No. | Phytoconstituents | Plant source | Active compound | Mechanism of action | References |
---|---|---|---|---|---|
1 | Flavanoid | Sambucus canadensis L., Ginkgo biloba L. and Hypericum perforatum L. | Quercetin | Suppression of cyclooxygenase 2 blocking AP-1’s attachment the leptin receptor, to extracellular signal-regulated kinase Ob-Ra and Mitogen-Activated Protein Kinase (MAPK) K/p38 kinase. | (Mirzaet al., 2023; Kwaket al., 2016; Shabiret al., 2022; Liet al., 2016; Serafiniet al., 2010; Ferrazet al.,2020; Cunhaet al., 2010; Sachset al., 2004; Ntalouka and Tsirivako, 2023; Choyet al., 2019; Krishnaet al., 2024). |
2 | Flavanoids | Salvadora persica | Luteolin-8 CfucopyraNOSide (LU8C-FP). | Decrease in IL-6. | (Lvet al., 2011; Azizet al., 2018). |
3 | Flavnoids | Ruta graveolens | Rutin | Both Nuclear factor E2-related factor (Nrf) stimulation and Nuclear factor (NF-κB) repression are involved. | (Álvarezet al., 2016; Ganeshpurkaret al., 2017; Raufet al., 2017;Farzaeiet al., 2019). |
4 | Flavanoids | Reseda luteola L | Luteolin | NF-κB, TNF-α and IL-6 reduction inhibition. | (Arampatziset al., 2023; Al-Khayriet al.,2022; Choyet al., 2019; Muruganathanet al., 2022). |
5 | Flavanoids | Sumach from Venice (Rhus cotinus L.) | Fisetin | Reduced levels of Tumor Necrosis Factor (TNF-α), Interleukin (IL-1β and IL-8). | (Grynkiewicz and Demchuk, 2019; Grynkiewicz and Demchuk, 2019; Choyet al., 2019; Garget al., 2019). |
6 | Triterpenoids | Souroubeasympetala | Betulinic acid | By modulating the NF-kB pathway, betulinic acid reduces the generation of IL-6 and inhibits the formation of nitric oxide. | (Punianiet al., 2014; Jeonget al., 2009; Linet al., 2015; Yunet al., 2003). |
7 | Triterpenoids | Lisgustrum lucidum | Oleanolic acid | Blocking Liposaccharide (LPS) release, which is a mediator of the synthesis of Cell Adhesion Molecules (CAMs) and High Mobility Group Box 1 (HMGB1). | (VYASet al., 2014; Xiaet al., 2011; Yanget al., 2012; Anderssonet al., 2011; Leeet al., 2013). |
8 | Triterpenoids | Herbs (such as organum, marjoram, rosemary, lavender and thyme), fruits (such as apple peel) and flowers. Australian sambucus. | Ursolic acid | Lower the quantities of IL-1β, IL-6, IL-8 and TNF-α, which are indicators of inflammation. | (Woźniaket al., 2015; Jägeret al., 2009; Szakielet al., 2012; Zhaoet al., 2023). |
9 | Triterpenoids | Americans’ ginseng Sebastianiaadenophora, Crataevanurvala, Bombax ceiba, Leptadeniahastata, Hematanthussucuuba, Shea butter plant species include andriedelianum. | Lupeol acid | Modifying the mRNA unique to IL-2 and TNF-alpha. | (Saleem, 2009; Beveridgeet al., 2002; Fernándezet al., 2001; Vasconceloset al., 2008; Yamashitaet al., 2002; Lucettiet al., 2010). |
10 | Triterpenoids | Centella asiatica | Asiatic acid | Control of NOS, IL-1β, COX-2 and IL-6. | (Nagooret al., 2018; Kambleet al., 2017; Patilet al., 2015; Yanget al., 2018). |
11 | Triterpenoids | Olea europaea | Maslinic acid | Preventing the synthesis of Tumour Necrosis Factor alpha (TNF-α) and nitric oxide. | (Lozanoet al., 2014; Ooiet al., 2023; Luet al., 2009; Bannoet al., 2005; Kimet al., 2005; Huanget al., 2011). |
12 | Triterpenoids | Planta odorum, Nerium oleander | Oleandrin | Reduced Phosphatidylinositol 3-Kinase (PI3K), phosphorylated Ak strain transforming (Akt) and inhibition of NF-κB. | (Zhaiet al., 2022; Khanet al., 2010; Atayet al., 2018). |
Sl. No. | Plant-based composites | Nanoformulation | Active ingredient | Inflammation-related illness | References |
---|---|---|---|---|---|
1 | Flavanoids | PLGA Nanoparticles | Quercetin | Antimicrobialactivity | (Kapset al., 2021). |
2 | Triterpenoids | Gold Nanoparticles | Betulinic acid | Anticancer disease | (Miocet al., 2018). |
3 | Triterpenoids | Pentacyclic triterpenoid – Lupeol Nanoemulsion. | Lupeol | Antimalarial illness | (Dwivediet al., 2023). |
4 | Triterpenoids | Liposomes | Ursolic acid | Anti malignant tumor action | (Sysaket al., 2023). |
5 | Triterpenoids | Chitoson Nanodrug Carrier. | Ursolic acid | Antitumor activity | (Shaoet al., 2020). |
6 | Flavanoids | Silver Nanoparticles with Quercetin. | Quercetin | The anti-inflammatory property. | (Wanget al., 2021). |
7 | Triterpenoids | Acid ursolic nanosuspension. | Ursolic acid | Diabetes type 2 | (Nieet al., 2020). |
8 | Flavanoids | Particles of nanoscale Acetate phthalate of cellulose, silica nanoparticles and nanoparticles The polycaprol act one particles of nanoscale Silica nanoparticles, polycaprolactone nanoparticles and cellulose acetate phthalate. | Quercetin | Anti-rheumaticeffect | (Guanet al., 2021). |
9 | Flavanoids | Both solid lipid and polymer nanoparticles, solid lipid nanoemulsions, elastic liposomes and nanostructured lipid transporters. | Quercetin, Rutin and Lutein | Topical infectious disorders | (Dwivediet al., 2023; Dermanet al., 2020; Viscusiet al., 2023). |
10 | Flavanoids | Metallic oxide, silver and gold nanoparticles. | Quercetin | Viral disease | (Jannatet al., 2021). |
Drug Delivery Systems
The efficiency of flavonoids as anti-inflammatory agents and their bioavailability have been greatly increased by the application of nanotechnology. Nanoscale particles, such as polylactic acid, Poly (Lactic-Co-Glycolic Acid) (PLGA) and polyoxymethylene glycol, are commonly utilized for phytochemical component transfer and pharmacokinetic parameter modification. For example, it has been shown that polyoxymethylene glycol nanoscale particles increase quercetin’s stability, solubilization and circulation persistence in the vascular system (Wanget al., 2013). Many morbidities, including high blood sugar levels, elevated blood pressure, infections of the upper respiratory tract, pancreatic and mammary carcinomas and heart abnormalities, have been linked to quercetin’s possible therapeutic advantages (Bennetet al., 2012; Yuet al., 2007; Kumariet al., 2012; Cunicoet al., 2020; Bose and Michniak, 2013; Liet al., 2009). However, its poor clinical relevance and efficacy are caused by low intrinsic bioactivity (<10%), considerable metabolic degradation (>40%), fast physiological elimination (<1 hr), limited hydro solubility (1 μg/mL) and inactive substances.
To get around these challenges, a variety of delivery modalities have been developed, including inclusion complexes and micelles, phospholipid vesicles, inorganic nanoscale particulates, polymeric nanoscale particulates/microspheres and other nanomaterials. Pentacyclic triterpenoidal substances have not been utilized in medicine despite having advantageous properties, Thus, the efficacy of in vivo therapy is not always correlated with its biological consequences (Kapset al., 2021; Ghante and Jamkhande, 2019; Szakielet al., 2012; Xiaet al., 2017; Yuet al., 2020; Shaoet al., 2020; Patraet al., 2018). Figure 2 presents an overview of the general benefits of Drug Delivery Mechanisms (DDMs).

Figure 2:
Efficacy of Drug Delivery systems.
Nano Drug Delivery Systems
Numerous research investigations have been carried out to determine which botanical compounds and entities from dietary and medicinal flora are effective for treating a variety of clinical disorders. On the other hand, rapid metabolism, limited permeability and instability can all hinder the bio utilization of such herbal bioactive substances, which is essential to their effectiveness (Yadavet al., 2011). In order to overcome the drawbacks of conventional therapy, nanoscale pharmaceutical delivery has become a viable method for delivering active ingredients. Pharmacological researchers have taken notice of the special qualities of nano vehicles, such as their increased surface area, quick evacuation from the stomach, controlled delivery to specific sites, increased internalization of cells, better bio utilization and biodistribution, affordability, patient compliance and increased healing efficacy (Yetisginet al., 2020).
Solid Lipid Nanovesicles (SLNs), nanoscale lipid-based entities, have attracted considerable interest worldwide despite the absence of polymeric materials with regulatory approval and their exorbitant price. The stiff core lipid matrix of SLNs provides increased stability of active principles, one of the many benefits they have over liposomal entities (Mishraet al., 2018). SLNs also demonstrate improved entrapment efficiency, regulated release for lipophilic drugs and appropriate modification for targeted therapeutics (Satapathyet al., 2021). Additionally, SLNs are superior to polymeric nanovesicles in that they can be produced on a large scale, are more stable and do not require organic solvents (Rizviet al., 2019). Many studies have revealed advantageous phytochemical entities from medicinal and dietary flora that can treat a range of pathological disorders. One possible way to improve the distribution of active principles while addressing these issues is through nanoscale pharmaceutical delivery.
Pharmacologists have been drawn to nano vehicles because of their intriguing properties, which include enhanced pharmacokinetics, precise targeting and reduced cytotoxicity. Because of their superior toughness, improved encapsulation efficiency and mass-production feasibility, Lipid-Based Nano Capsules (LNCs) have become the preferred option over polymeric nano capsules. To be more precise, LNCs are very histocompatible since they are made of endogenous and biodegradable lipids; they are more robust because of their stiff lipid core; and they are made with less effort because they don’t require complicated polymeric structures during the manufacturing process. In addition, LNCs are more histocompatible than polymeric nano capsules and may be customized for precision therapies, which makes them a desirable choice for effective phytochemical drug administration (Rawatet al., 2023; Sharmaet al., 2019).
Challenges in Formulating Nano Formulations
Nanoscale entities with customized architecture, size and dispersal can often be produced by adjusting fabrication processes, reductive catalysts and stabilizing agents (Royet al., 2019). Due to variations in the phytochemical extracts used, the morphology and dimensions of the nanoscale entities are significantly influenced by them. Consequently, standardizing and producing nanoscale objects with the necessary size, topology and surface characteristics requires optimization of the manufacturing paradigm (Patil and Chandrasekaran, 2020). Although state-of-the-art nanoscale formulations for drug delivery systems are constantly being developed and provide several advantages, there are still some barriers that keep them from being widely deployed. Researchers have challenges while developing and manufacturing nanoscale formulations due to a variety of factors, such as (i) biomaterial characteristics; (ii) fabrication restrictions; (iii) payload capability; (iv) stability; and (v) biological hurdles (Yetisginet al., 2020; Shaoet al., 2020).
The characteristics of biomaterials, such as their biodegradability or surface moiety charge, can affect their membrane permeability, cytotoxicity and biocompatibility (Santoset al., 2019). Producing consistent product batches free of unwanted fabrication byproducts requires an effective and scalable production strategy for nanoscale formulations (Kahramanet al., 2017).
Pharmacokinetics of Nano Formulations
Distinct delivery systems allow Nano Formulations to be introduced into the body and precisely target particular body locations. Nanoparticle characteristics including size, concentration, surface electronegativity and hydrogen ion concentration have a significant influence on pharmacokinetic processes, such as absorption and dissolution. A rise in the bloodstream’s level of nanoparticles during a single day that is dose-dependent has been demonstrated by empirical evidence, with the liver, kidney and lungs exhibiting the most significant accumulation over a 72-hr period. The results emphasize the significant impact of nanoparticle properties on pharmacokinetic mechanisms and possible toxicity, indicating that urine excretion is the main method of eliminating tiny nanoparticles, while feces and the biliary system are the main routes of elimination for bigger ones. The quantity of nanoparticles in circulation rises dose-dependently and is linked to increasing toxicity; the organs most impacted are the liver and splenic ones.
To minimize potential negative effects, it is imperative to customize the size and concentration of nanoparticles according to the the area of the target and the degree of inflammation. Moreover, surface modification using Ethylene Diamine Tetra Methylene Phosphonic Acid (EDTMP) has been shown to lessen the cytotoxicity of nanoparticles by lessening the oxidative stress that is caused by high nanoparticle concentrations (Imperialeet al., 2019; Chen et al., 2017).
Inflammation
In response to external stimuli, the immune system produces inflammation as a defense mechanism in response to things like pathogen invasions, cellular destruction, toxic substances, or ionizing radiation (Medzhitov, 2010; Ferreroet al., 2007; Nathan and Ding, 2010; Zhouet al., 2016). Usually, this defensive reaction restores tissue homeostasis and reduces inflammation by reducing the likelihood of infection or trauma through cellular and molecular processes. Leukocyte mobilization, pro-inflammatory mediator release and altered vascular permeability are all significant outcomes of this process (Chertovet al., 2000). Irritation manifests as irritation, edema, heat, discomfort and tissue dysfunction because of localized inflamed, vascular and immune cell reactions to injury or infection (Takeuchiet al., 2010). An array of factors, mostly pertaining to the upper respiratory tract and nasal cavities, can cause inflammation, such as pathogenic invasions, severe temperature swings, toxic chemicals, physical trauma and abnormalities in health.
A defective multi systemic response to airway stimulation, including effector cells and pro-inflammatory mediators, is the hallmark of asthma, a chronic inflammatory airway illness. Sepsis, on the other hand, is a potentially fatal reaction to pathogenic invasion that releases pro-inflammatory mediators and damages numerous organs. Individuals may have symptoms like hypotension, pyrexia, decreased urine production, impaired hemostasis, increased vascular permeability and loss of smooth muscle tone. These illnesses have been treated with synthetic medications such as corticosteroids, leukotriene receptor antagonists and antimicrobials (Yatooet al., 2018; Jamtshoet al., 2024; Bjorkman, 1998; Ricciotti and FitzGerald, 2011; Rainsford,et al., 2007; Bermas,et al., 2014; Shaikhet al., 2012; Juthaniet al., 2017; Moskovtchenko and Cognet, 1976; Ayroldiet al., 2012; Celotti and Laufer, 2001).
These pharmacological categories, although therapeutically beneficial, are also associated with a wide range of adverse reactions, such as fever, gastrointestinal distress, nasal blockage, aural infections, cutaneous manifestations, pruritic eruptions, bronchial irritation, tussive episodes, sinusoidal infections, upper respiratory tract disorders, atopic eczema, cranial discomfort, nausea, laryngeal inflammation, pharyngeal inflammation, viral afflictions, dyspneic episodes, odontalgic pain, vertiginous sensations, gastric dyspepsia and increased levels of hepatocellular enzymes. In contrast, a number of plant components (including flavonoids and triterpenoids) found in phytotherapeutic treatments have been demonstrated to improve immunological response and offer numerous other advantages, which helps to mitigate the drawbacks of traditional treatment approaches (Altavillaet al., 2009; Linet al., 2008; Aggarwalet al., 2011). Figure 3 illustrates the mechanistic effects of different coactivational substances and receptors.

Figure 3:
The method by which NFκB operates.
In the cytosol, where the Rel and p50 proteins are found, NF-κβ forms a complex with the inhibitory protein Iκβα when it is in its active state. Membrane receptors pick up extracellular signals, which then activate Iκb Kinase (IKK). The IKK then phosphorylates the Iκβα protein, which causes it to get ubiquitinated and eventually be degraded by the proteasome (by the canonical pathway). RelB prefers the non-canonical NF-κβ activation route through RelB. Response Elements (RE) are DNA bindings that induce NF-κβ to translocate into the nucleus and bind. Coactivators and RNA polymerase are drawn to the DNA/NF-κβ complex, which causes the latter to produce mRNA from the DNA and change the cell.
Function of Triterpenoids and Flavonoids in Inflammation
Due to differences in their chemical structures, flavonoids may be divided into six groups: flavones, phenols, flavanonols, isoflavonoids, flavan-3-ols and anthocyanins, are found in a wide range of plant materials, such as fruits, flowers and leaves (Choyet al., 2019; Pollastri and Tattini, 2011; Barrecaet al., 2017). Studies have indicated that in the presence of chronic inflammation, triterpenoids and flavonoids impact several redox pathways, including Nuclear Factor kappa B (NFκB). The transcriptional regulation NFκB is activated by inflammatory stimuli, leading to the suppression of the Inhibiting kappa B (IκB) kinase. This finally results in cellular death and a rise in pro-inflammatory cytokines, including Tumor Necrosis Factor-alpha (TNF-α). It has been observed that flavonoids and triterpenoids reduce oxidative stress in a variety of medical conditions, as well as the concentrations Elevated blood serum levels of TNF-α or Interleukin-1 beta (IL-1β), as well as blood mononuclear cells’ NFκB transcriptional activity (Chekalinaet al., 2018; Indraet al., 2013; Tanget al., 2014; Oyagbemiet al., 2018; Lvet al., 2011; Garget al., 2019; Renet al., 2018; Xionget al., 2018; Leeet al., 2012; Raniet al., 2016; Hanet al., 2015; Suchalet al., 2016; Kashyapet al., 2016).
Referring to Figure 5, Figure 4 depicts the inflammatory mechanism. Diverse treatment methods, such as corticosteroidal and non-corticosteroidal ones, are being continually investigated to manage acute inflammatory responses. Unfortunately, these techniques frequently fall short of the intended therapeutic result because of drawbacks including non-specific pharmacokinetic distribution, limited bioavailability, or short half-lives, which force doctors to prescribe higher dosages and cause unintended side effects. Nanoscale particles are becoming more and more popular as a solution to these problems because of their ability to target specific molecular entities, minimize toxicity and negative effects and facilitate controlled release in affected cellular and tissue structures. They also have controlled dimensions, morphology and surface charge.

Figure 4:
A number of infections and allergens in the environment cause immune cells to release specific cytokines and chemokines, which chemotactically trigger eosinophil infiltration through endothelial cells at the site of inflammation and intensify the inflammatory process.

Figure 5:
Recent nanomedicines for the treatment of inflammation.
Nanoscale formulations demonstrate a significant advantage in regulating maintaining the proper ratio of pro-to-anti-inflammatory effectors and using endocytic absorption to target cell phagocytic or inflammatory sentinels in particular. In addition, precision targeting of particular cell populations with nanovehicular systems might greatly improve immune response or intracellular internalization to suppress inflammation. Anti-inflammatory nanopharmaceuticals are the product of significant efforts to develop anti-inflammatory tactics that overcome the constraints of conventional pharmaceutical forms (Maioet al., 2021; Caiet al., 2021; Jubileeet al., 2024; Costanzoet al., 2016).
CONCLUSION
The treatment of inflammation could be completely transformed by the combination of nanotechnology and herbal products. Improved patient outcomes can be achieved by using Nano Formulations to optimize drug administration, improve biocompatibility and minimize side effects. This strategy offers a viable treatment for chronic inflammatory illnesses while addressing the drawbacks of traditional therapies. Researchers can develop effective and tailored medicines by using Nano Formulations, which will ultimately change the way disease management is handled. Approaches to personalized treatment are made possible by the adaptability of Nano Formulations, which enable targeted moieties and customized designs. To fully achieve the potential of Nano Formulations and make them a reality for patients, more research is required. Nano Formulations have the potential to significantly improve the lives of millions of people globally by serving as a cornerstone of inflammatory control with further innovation. It appears that the therapy of inflammation has a bright future and Nano Formulations will likely be crucial in determining that future. To realize their full potential, ongoing research is essential.
Cite this article:
Ravikumar L, Velmurugan R, Jahnavi P, Vidiyala P, Kumar JP, Rahul A, et al. Nanoscale Formulations for Targeted Drug Delivery: Enhancing Stability, Release Control and Therapeutic Efficacy. Int. J. Pharm. Investigation. 2025;15(3):313-24.
REFERENCE
- Aggarwal, B. B., Prasad, S., Reuter, S., Kannappan, R., Yadev, V. R., Park, B., Kim, J. H., Gupta, S. C., Phromnoi, K., Sundaram, C., Prasad, S., Chaturvedi, M. M., & Sung, B. (2011, October). Identification of novel anti-inflammatory agents from Ayurvedic medicine for prevention of chronic diseases: “reverse pharmacology” and “bedside to bench” approach. Current Drug Targets, 12(11), 1595–1653. https://doi.org/10.2174/138945011798109464, PubMed: 21561421, PubMed Central: PMC3170500
- Al-Khayri, J. M., Sahana, G. R., Nagella, P., Joseph, B. V., Alessa, F. M., & Al-Mssallem, M. Q. (2022, May 2). Flavonoids as potential anti-inflammatory molecules: A review. Molecules, 27(9), 2901. https://doi.org/10.3390/molecules27092901, PubMed: 35566252, PubMed Central: PMC9100260
- Altavilla, D., Squadrito, F., Bitto, A., Polito, F., Burnett, B. P., Di Stefano, V., & Minutoli, L. (2009, August). Flavocoxid, a dual inhibitor of cyclooxygenase and 5-lipoxygenase, blunts pro-inflammatory phenotype activation in endotoxin-stimulated macrophages. British Journal of Pharmacology, 157(8), 1410–1418. https://doi.org/10.1111/j.1476-5381.2009.00322.x, PubMed: 19681869, PubMed Central: PMC2765324
- Álvarez, R., Araya, H., Navarro-Lisboa, R., & Lopez de Dicastillo, C. (2016, December). Evaluation of polyphenol content and antioxidant capacity of fruits and vegetables using a modified enzymatic extraction. Food Technology and Biotechnology, 54(4), 462–467. https://doi.org/10.17113/ftb.54.04.16.4497, PubMed: 28115904, PubMed Central: PMC5253986
- Andersson, U., & Tracey, K. J. (2011). HMGB1 is a therapeutic target for sterile inflammation and infection. Annual Review of Immunology, 29, 139–162. https://doi.org/10.1146/annurev-immunol-030409-101323, PubMed: 21219181, PubMed Central: PMC4536551
- Arampatzis, A. S., Pampori, A., Droutsa, E., Laskari, M., Karakostas, P., Tsalikis, L., Barmpalexis, P., Dordas, C., & Assimopoulou, A. N. (2023, November 22). Occurrence of luteolin in the Greek flora, isolation of luteolin and its action for the treatment of periodontal diseases. Molecules, 28(23), 7720. https://doi.org/10.3390/molecules28237720, PubMed: 38067450, PubMed Central: PMC10707704
- Atay Balkan, İ., Gören, A. C., Kırmızıbekmez, H., & Yeşilada, E. Y. (2018). Evaluation of the in vitro anti-inflammatory activity of nerium oleander L. flower extracts and activity-guided isolation of the active constituents. Records of Natural Products, 12(2), 128–141. https://doi.org/10.25135/rnp.15.17.05.100
- Ayroldi, E., Cannarile, L., Migliorati, G., Nocentini, G., Delfino, D. V., & Riccardi, C. (2012, December). Mechanisms of the anti-inflammatory effects of glucocorticoids: Genomic and nongenomic interference with MAPK signaling pathways. FASEB Journal, 26(12), 4805–4820. https://doi.org/10.1096/fj.12-216382 [EPub]. PubMed: 22954589
- Aziz, N., Kim, M.-Y., & Cho, J. Y. (2018, October 28). Anti-inflammatory effects of luteolin: A review of in vitro, in vivo and in silico studies. Journal of Ethnopharmacology, 225, 342–358. https://doi.org/10.1016/j.jep.2018.05.019 [EPub]. PubMed: 29801717
- Bagad, A. S., Joseph, J. A., Bhaskaran, N., & Agarwal, A. (2013). Comparative evaluation of anti-inflammatory activity of curcuminoids, turmerones and aqueous extract of Curcuma longa. Advances in Pharmacological Sciences, 2013, Article 805756. https://doi.org/10.1155/2013/805756 [EPub]. PubMed: 24454348, PubMed Central: PMC3885190
- Banno, N., Akihisa, T., Tokuda, H., Yasukawa, K., Taguchi, Y., Akazawa, H., Ukiya, M., Kimura, Y., Suzuki, T., & Nishino, H. (2005, October). Anti-inflammatory and antitumor-promoting effects of the triterpene acids from the leaves of Eriobotrya japonica. Biological and Pharmaceutical Bulletin, 28(10), 1995–1999. https://doi.org/10.1248/bpb.28.1995, PubMed: 16204964
- Barreca, D., Gattuso, G., Bellocco, E., Calderaro, A., Trombetta, D., Smeriglio, A., Laganà, G., Daglia, M., Meneghini, S., & Nabavi, S. M. (2017, July 8). Flavanones: Citrus phytochemical with health-promoting properties. Bio Factors, 43(4), 495–506. https://doi.org/10.1002/biof.1363 [EPub]. PubMed: 28497905
- Bennet, D., Marimuthu, M., Kim, S., & An, J. (2012). Dual drug-loaded nanoparticles on self-integrated scaffold for controlled delivery. International Journal of Nanomedicine, 7, 3399–3419. https://doi.org/10.2147/IJN.S32800 [EPub]. PubMed: 22888222, PubMed Central: PMC3414208
- Bermas, B. L. (2014, May). Non-steroidal anti inflammatory drugs, glucocorticoids and disease modifying anti-rheumatic drugs for the management of rheumatoid arthritis before and during pregnancy. Current Opinion in Rheumatology, 26(3), 334–340. https://doi.org/10.1097/BOR.0000000000000054, PubMed: 24663106
- Beveridge, T. H. J., Li, T. S. C., & Drover, J. C. G. (2002, February 13). Phytosterol content in American ginseng seed oil. Journal of Agricultural and Food Chemistry, 50(4), 744–750. https://doi.org/10.1021/jf010701v, PubMed: 11829639
- Bjorkman, D. J. (1998, July 27). The effect of aspirin and nonsteroidal anti-inflammatory drugs on prostaglandins. The American Journal of Medicine, 105(1B), 8S–12S. https://doi.org/10.1016/s0002-9343(98)00069-2, PubMed: 9715829
- Bose, S., & Michniak-Kohn, B. (2013, February 14). Preparation and characterization of lipid based nano systems for topical delivery of quercetin. European Journal of Pharmaceutical Sciences, 48(3), 442–452. https://doi.org/10.1016/j.ejps.2012.12.005 [EPub] 2012 Dec 13. PubMed: 23246734
- Cai, Z., Wang, S., & Li, J. (2021, December 20). Treatment of inflammatory bowel disease: A comprehensive review. Frontiers in Medicine, 8, Article 765474. https://doi.org/10.3389/fmed.2021.765474, PubMed: 34988090, PubMed Central: PMC8720971
- Celotti, F., & Laufer, S. (2001, May). Anti-inflammatory drugs: New multitarget compounds to face an old problem. The dual inhibition concept. Pharmacological Research, 43(5), 429–436. https://doi.org/10.1006/phrs.2000.0784, PubMed: 11394934
- Chekalina, N., Burmak, Y., Petrov, Y., Borisova, Z., Manusha, Y., Kazakov, Y., & Kaidashev, I. (2018, September–October). Quercetin reduces the transcriptional activity of NF-kB in stable coronary artery disease. Indian Heart Journal, 70(5), 593–597. https://doi.org/10.1016/j.ihj.2018.04.006 [EPub]. PubMed: 30392493, PubMed Central: PMC6204471
- Chen, L., Deng, H., Cui, H., Fang, J., Zuo, Z., Deng, J., Li, Y., Wang, X., & Zhao, L. (2018). Inflammatory responses and inflammation-associated diseases in organs. Oncotarget, 9(6), 7204–7218. https://doi.org/10.18632/oncotarget.23208, PubMed: 29467962, PubMed Central: PMC5805548
- Chertov, O., Yang, D., Howard, O. M., & Oppenheim, J. J. (2000, October). Leukocyte granule proteins mobilize innate host defenses and adaptive immune responses. Immunological Reviews, 177, 68–78. https://doi.org/10.1034/j.1600-065x.2000.17702.x, PubMed: 11138786
- Choy, K. W., Murugan, D., Leong, X.-F., Abas, R., Alias, A., & Mustafa, M. R. (2019, October 31). Flavonoids as natural anti-inflammatory agents targeting nuclear factor-kappa B (NFκB) signaling in cardiovascular diseases: A mini review. Frontiers in Pharmacology, 10, 1295. https://doi.org/10.3389/fphar.2019.01295, PubMed: 31749703, PubMed Central: PMC6842955
- Costanzo, P., Cariati, L., Desiderio, D., Sgammato, R., Lamberti, A., Arcone, R., Salerno, R., Nardi, M., Masullo, M., & Oliverio, M. (2016, March 28). Design, synthesis and evaluation of donepezil-like compounds as AChE and BACE-1 inhibitors. ACS Medicinal Chemistry Letters, 7(5), 470–475. https://doi.org/10.1021/acsmedchemlett.5b00483, PubMed: 27190595, PubMed Central: PMC4867475
- Cunha, T. M., Roman-Campos, D., Lotufo, C. M., Duarte, H. L., Souza, G. R., Verri, W. A., Jr., Funez, M. I., Dias, Q. M., Schivo, I. R., Domingues, A. C., Sachs, D., Chiavegatto, S., Teixeira, M. M., Hothersall, J. S., Cruz, J. S., Cunha, F. Q., & Ferreira, S. H. (2010, March 2). Morphine peripheral analgesia depends on activation of the PI3Kgamma/AKT/nNOS/NO/KATP signaling pathway. Proceedings of the National Academy of Sciences of the United States of America, 107(9), 4442–4447. https://doi.org/10.1073/pnas.0914733107 [EPub]. PubMed: 20147620, PubMed Central: PMC2840166
- Cunico, L. P., Cobo, A. M., Al-Hamimi, S., & Turner, C. (2020, November 27). Solubility and thermal degradation of quercetin in CO2-expanded liquids. Molecules, 25(23), 5582. https://doi.org/10.3390/molecules25235582, PubMed: 33261120, PubMed Central: PMC7730818
- Derman, S., Uzunoglu, D., Acar, T., Arasoglu, T., Ucak, S., Ozalp, V. C., & Mansuroglu, B. (2020, winter). Antioxidant activity and hemocompatibility study of quercetin loaded Plga nanoparticles. Iranian Journal of Pharmaceutical Research, 19(1), 424–435. https://doi.org/10.22037/ijpr.2020.1101000, PubMed: 32922498, PubMed Central: PMC7462503
- Dwivedi, K., Mandal, A. K., Afzal, O., Altamimi, A. S. A., Sahoo, A., Alossaimi, M. A., Almalki, W. H., Alzahrani, A., Barkat, M. A., Almeleebia, T. M., Mir Najib Ullah, S. N., & Rahman, M. (2023, August 18). Emergence of nano-based formulations for effective delivery of flavonoids against topical infectious disorders. Gels, 9(8), 671. https://doi.org/10.3390/gels9080671, PubMed: 37623126, PubMed Central: PMC10453850
- Farzaei, M. H., Singh, A. K., Kumar, R., Croley, C. R., Pandey, A. K., Coy-Barrera, E., Kumar Patra, J., Das, G., Kerry, R. G., Annunziata, G., Tenore, G. C., Khan, H., Micucci, M., Budriesi, R., Momtaz, S., Nabavi, S. M., & Bishayee, A. (2019, October 8). Targeting inflammation by flavonoids: Novel therapeutic strategy for metabolic disorders. International Journal of Molecular Sciences, 20(19), 4957. https://doi.org/10.3390/ijms20194957, PubMed: 31597283, PubMed Central: PMC6801776
- Fernández, M. A., de las Heras, B., García, M. D., Sáenz, M. T., & Villar, A. (2001, November). New insights into the mechanism of action of the anti-inflammatory triterpene lupeol. The Journal of Pharmacy and Pharmacology, 53(11), 1533–1539. https://doi.org/10.1211/0022357011777909, PubMed: 11732756
- Ferraz, C. R., Carvalho, T. T., Manchope, M. F., Artero, N. A., Rasquel-Oliveira, F. S., Fattori, V., Casagrande, R., & Verri, W. A., Jr. (2020, February 10). Therapeutic potential of flavonoids in pain and inflammation: Mechanisms of action, pre-clinical and clinical data and pharmaceutical development. Molecules, 25(3), 762. https://doi.org/10.3390/molecules25030762, PubMed: 32050623, PubMed Central: PMC7037709
- Ferrero-Miliani, L., Nielsen, O. H., Andersen, P. S., & Girardin, S. E. (2007, February). Chronic inflammation: Importance of NOD2 and NALP3 in interleukin-1beta generation. Clinical and Experimental Immunology, 147(2), 227–235. https://doi.org/10.1111/j.1365-2249.2006.03261.x, PubMed: 17223962, PubMed Central: PMC1810472
- Ganeshpurkar, A., & Saluja, A. K. (2017, February). The pharmacological potential of Rutin. Saudi Pharmaceutical Journal, 25(2), 149–164. https://doi.org/10.1016/j.jsps.2016.04.025 [EPub] 2016 Apr 30. PubMed: 28344465, PubMed Central: PMC5355559
- Garg, S., Malhotra, R. K., Khan, S. I., Sarkar, S., Susrutha, P. N., Singh, V., Goyal, S., Nag, T. C., Ray, R., Bhatia, J., & Arya, D. S. (2019, March 15). Fisetin attenuates isoproterenol-induced cardiac ischemic injury in vivo by suppressing RAGE/NF-κB mediated oxidative stress, apoptosis and inflammation. Phytomedicine, 56, 147–155. https://doi.org/10.1016/j.phymed.2018.09.187 [EPub] 2018 Sep 19. PubMed: 30668335
- Ghante, M. H., & Jamkhande, P. G. (2019, June). Role of pentacyclic triterpenoids in chemoprevention and anticancer treatment: An overview on targets and underling mechanisms. Journal of Pharmacopuncture, 22(2), 55–67. https://doi.org/10.3831/KPI.201.22.007 [EPub]. PubMed: 31338244, PubMed Central: PMC6645347
- Ghasemian, M., Owlia, S., & Owlia, M. B. (2016). Review of anti-inflammatory herbal medicines. Advances in Pharmacological Sciences, 2016, Article 9130979. https://doi.org/10.1155/2016/9130979 [EPub]. PubMed: 27247570, PubMed Central: PMC4877453
- Grynkiewicz, G., & Demchuk, O. M. (2019, October 30). New perspectives for fisetin. Frontiers in Chemistry, 7, 697. https://doi.org/10.3389/fchem.2019.00697, PubMed: 31750288, PubMed Central: PMC6842927
- Guan, F., Wang, Q., Bao, Y., & Chao, Y. (2021, February 11). Anti-rheumatic effect of quercetin and recent developments in nano formulation. RSC Advances, 11(13), 7280–7293. https://doi.org/10.1039/d0ra08817j, PubMed: 35423269, PubMed Central: PMC8695102
- Han, S., Wu, H., Li, W., & Gao, P. (2015, May). Protective effects of genistein in homocysteine-induced endothelial cell inflammatory injury. Molecular and Cellular Biochemistry, 403(1–2), 43–49. https://doi.org/10.1007/s11010-015-2335-0 [EPub]. PubMed: 25626894
- Huang, L., Guan, T., Qian, Y., Huang, M., Tang, X., Li, Y., & Sun, H. (2011, December 15). Anti-inflammatory effects of maslinic acid, a natural triterpene, in cultured cortical astrocytes via suppression of nuclear factor-kappa B. European Journal of Pharmacology, 672(1–3), 169–174. https://doi.org/10.1016/j.ejphar.2011.09.175 [EPub]. PubMed: 21970807
- Imperiale, J. C., Schlachet, I., Lewicki, M., Sosnik, A., & Biglione, M. M. (2019, November 11). Oral pharmacokinetics of a chitosan-based nano- drug delivery system of interferon alpha. Polymers, 11(11), 1862. https://doi.org/10.3390/polym11111862, PubMed: 31718060, PubMed Central: PMC6918283
- Indra, M. R., Karyono, S., Ratnawati, R., & Malik, S. G. (2013, July 16). Quercetin suppresses inflammation by reducing ERK1/2 phosphorylation and NF kappa B activation in leptin-induced human umbilical vein endothelial cells (HUVECs). BMC Research Notes, 6, 275. https://doi.org/10.1186/1756-0500-6-275, PubMed: 23856194, PubMed Central: PMC3725156
- Jäger, S., Trojan, H., Kopp, T., Laszczyk, M. N., & Scheffler, A. (2009, June 4). Pentacyclic triterpene distribution in various plants – Rich sources for a new group of multi-potent plant extracts. Molecules, 14(6), 2016–2031. https://doi.org/10.3390/molecules14062016, PubMed: 19513002, PubMed Central: PMC6254168
- Jamtsho, T., Loukas, A., & Wangchuk, P. (2024, June 21). Pharmaceutical potential of remedial plants and helminths for treating inflammatory bowel disease. Pharmaceuticals, 17(7), 819. https://doi.org/10.3390/ph17070819, PubMed: 39065669, PubMed Central: PMC11279646
- Jannat, K., Paul, A. K., Bondhon, T. A., Hasan, A., Nawaz, M., Jahan, R., Mahboob, T., Nissapatorn, V., Wilairatana, P., Pereira, M. L., & Rahmatullah, M. (2021, November 8). Nanotechnology applications of flavonoids for viral diseases. Pharmaceutics, 13(11), 1895. https://doi.org/10.3390/pharmaceutics13111895, PubMed: 34834309, PubMed Central: PMC8625292
- Jeong, W., Hong, S. S., Kim, N., Yang, Y. T., Shin, Y. S., Lee, C., Hwang, B. Y., & Lee, D. (2009, June). Bioactive triterpenoids from Callistemon lanceolatus. Archives of Pharmacal Research, 32(6), 845–849. https://doi.org/10.1007/s12272-009-1605-3 [EPub]. PubMed: 19557361
- Jubilee, R., Komala, M., & Patel, S. (2024, June 24). Therapeutic potential of resveratrol and lignans in the management of tuberculosis. Cell Biochemistry and Biophysics, 82(3), 1809–1823. https://doi.org/10.1007/s12013-024-01378-7 [Epub ahead of print]. PubMed: 38914838
- Juthani, V. V., Clearfield, E., & Chuck, R. S. (2017, July 3). Non-steroidal anti-inflammatory drugs versus corticosteroids for controlling inflammation after uncomplicated cataract surgery. The Cochrane Database of Systematic Reviews, 7(7), Article CD010516. https://doi.org/10.1002/14651858.CD010516.pub2, PubMed: 28670710, PubMed Central: PMC5580934
- Kahraman, E., Güngör, S., & Özsoy, Y. (2017, November). Potential enhancement and targeting strategies of polymeric and lipid-based nanocarriers in dermal drug delivery. Therapeutic Delivery, 8(11), 967–985. https://doi.org/10.4155/tde-2017-0075, PubMed: 29061106
- Kamble, S. M., Patel, H. M., Goyal, S. N., Noolvi, M. N., Mahajan, U. B., Ojha, S., & Patil, C. R. (2017). In silico Evidence for Binding of pentacyclic triterpenoids to Keap1-Nrf2 protein–protein Binding Site. Combinatorial Chemistry and High Throughput Screening, 20(3), 215–234. https://doi.org/10.2174/1386207319666161214111822, PubMed: 28024463
- Kaps, A., Gwiazdoń, P., & Chodurek, E. (2021, March 21). Nanoformulations for delivery of pentacyclic triterpenoids in anticancer therapies. Molecules, 26(6), 1764. https://doi.org/10.3390/molecules26061764, PubMed: 33801096, PubMed Central: PMC8004206
- Kashyap, D., Sharma, A., Tuli, H. S., Punia, S., & Sharma, A. K. (2016). Ur solic acid and oleanolic acid: Pentacyclic terpenoids with promising anti-inflammatory activities. Recent Patents on Inflammation and Allergy Drug Discovery, 10(1), 21–33. https://doi.org/10.2174/1872213×10666160711143904, PubMed: 27531153
- Khan, I., Kant, C., Sanwaria, A., & Meena, L. (2010, October). Acute cardiac toxicity of nerium oleander/indicum poisoning (kaner) poisoning. Heart Views, 11(3), 115–116. https://doi.org/10.4103/1995-705X.76803, PubMed: 21577379, PubMed Central: PMC3089829
- Kim, D.-H., Han, K.-M., Chung, I.-S., Kim, D.-K., Kim, S.-H., Kwon, B.-M., Jeong, T.-S., Park, M.-H., Ahn, E.-M., & Baek, N.-I. (2005, May). Triterpenoids from the flower of Campsis grandiflora K. Schum. as human acyl-CoA: Cholesterol acyltransferase inhibitors. Archives of Pharmacal Research, 28(5), 550–556. https://doi.org/10.1007/BF02977757, PubMed: 15974441
- Kumari, A., Kumar, V., & Yadav, S. K. (2012). Plant extract synthesized PLA nanoparticles for controlled and sustained release of quercetin: A green approach. PLOS One, 7(7), Article e41230. https://doi.org/10.1371/journal.pone.0041230 [EPub]. PubMed: 22844443, PubMed Central: PMC3402536
- Kwak, J.-H., Seo, J. M., Kim, N.-H., Arasu, M. V., Kim, S., Yoon, M. K., & Kim, S.-J. (2017, September). Variation of quercetin glycoside derivatives in three onion (Allium cepa L.) varieties. Saudi Journal of Biological Sciences, 24(6), 1387–1391. https://doi.org/10.1016/j.sjbs.2016.05.014 [EPub] 2016 May 27. PubMed: 28855836, PubMed Central: PMC5562462
- L, A., Krishna Kumar, J., & Shanmugam, R. (2024, January 5). LA, Krishna Kumar J, Shanmugam R. Formulation of quercetin mouthwash and anti-microbial potential against critical pathogens: An in vitro evaluation. Cureus, 16(1), Article e51688. https://doi.org/10.7759/cureus.51688. PMID: 383(Muruganathan, et al., 2022)06. PubMed Central: PMC10838391
- Lee, W., Ku, S.-K., & Bae, J.-S. (2012, September). Barrier protective effects of rutin in LPS-induced inflammation in vitro and in vivo. Food and Chemical Toxicology, 50(9), 3048-3055. https://doi.org/10.1016/j.fct.2012.06.013 [EPub]. PubMed: 22721984
- Lee, W., Yang, E.-J., Ku, S.-K., Song, K.-S., & Bae, J.-S. (2013, February). Anti-inflammatory effects of oleanolic acid on LPS-induced inflammation in vitro and in vivo. Inflammation, 36(1), 94-102. https://doi.org/10.1007/s10753-012-9523-9, PubMed: 22875543
- Li, H., Zhao, X., Ma, Y., Zhai, G., Li, L., & Lou, H. (2009, February 10). Enhancement of gastrointestinal absorption of quercetin by solid lipid nanoparticles. Journal of Controlled Release, 133(3), 238–244. https://doi.org/10.1016/j.jconrel.2008.10.002 [EPub] 2008 Oct 12. PubMed: 18951932
- Li, Y., Yao, J., Han, C., Yang, J., Chaudhry, M. T., Wang, S., Liu, H., & Yin, Y. (2016, March 15). Quercetin, inflammation and immunity. Nutrients, 8(3), 167. https://doi.org/10.3390/nu8030167, PubMed: 26999194, PubMed Central: PMC4808895
- Lin, A.-S., Lin, C.-R., Du, Y.-C., Lübken, T., Chiang, M. Y., Chen, I.-H., Wu, C.-C., Hwang, T.-L., Chen, S.-L., Yen, M.-H., Chang, F.-R., & Wu, Y.-C. (2009, February). Acasiane A and B and farnesirane A and B, diterpene derivatives from the roots of Acacia farnesiana. Planta Medica, 75(3), 256–261. https://doi.org/10.1055/s-0028-1112201 [EPub] 2008 Dec 19. PubMed: 19101886
- Lin, C.-K., Tseng, C.-K., Chen, K.-H., Wu, S.-H., Liaw, C.-C., & Lee, J.-C. (2015, September). Betulinic acid exerts anti-hepatitis C virus activity via the suppression of NF-κB- and MAPK-ERK1/2-mediated COX-2 expression. British Journal of Pharmacology, 172(18), 4481–4492. https://doi.org/10.1111/bph.13233 [EPub]. PubMed: 26102077, PubMed Central: PMC4562509
- López Zúñiga, M. Á., Moreno-Moral, A., Ocaña-Granados, A., Padilla-Moreno, F. A., Castillo-Fernández, A. M., Guillamón-Fernández, D., Ramírez-Sánchez, C., Sanchez-Palop, M., Martínez-Colmenero, J., Pimentel-Villar, M. A., Blázquez-Roselló, S., Moreno-Sánchez, J. J., López-Vílchez, M., Prior-Sánchez, I., Jódar-Moreno, R., & López Ruz, M. Á. (2021, January 28). High-dose corticosteroid pulse therapy increases the survival rate in COVID-19 patients at risk of hyper-inflammatory response. PLOS One, 16(1), Article e0243964. https://doi.org/10.1371/journal.pone.0243964, PubMed: 33507958, PubMed Central: PMC7842890
- Lozano-Mena, G., Sánchez-González, M., Juan, M. E., & Planas, J. M. (2014, August 4). Maslinic acid, a natural phytoalexin-type triterpene from olives-A promising nutraceutical? Molecules, 19(8), 11538–11559. https://doi.org/10.3390/molecules190811538, PubMed: 25093990, PubMed Central: PMC6271970
- Lu, H., Xi, C., Chen, J., & Li, W. (2009, September). [Determination of triterpenoid acids in leaves of Eriobotrya japonica collected at in different seasons]. Zhongguo Zhong Yao Za Zhi, 34(18), 2353–2355. PubMed: 20030087
- Lucetti, D. L., Lucetti, E. C., Bandeira, M. A. M., Veras, H. N., Silva, A. H., Leal, L. K. A., Lopes, A. A., Alves, V. C., Silva, G. S., Brito, G. A., & Viana, G. B. (2010, December 17). Anti-inflammatory effects and possible mechanism of action of lupeol acetate isolated from Himatanthus drasticus (Mart.) Plumel. Journal of Inflammation, 7, 60. https://doi.org/10.1186/1476-9255-7-60, PubMed: 21167055, PubMed Central: PMC3019217
- Lv, L., Lv, L., Zhang, Y., & Kong, Q. (2011, December). Luteolin prevents LPS-induced TNF-α expression in cardiac myocytes through inhibiting NF-κB signaling pathway. Inflammation, 34(6), 620–629. https://doi.org/10.1007/s10753-010-9271-7, PubMed: 21076936
- Maio, A., Pibiri, I., Morreale, M., Mantia, F. P. L., & Scaffaro, R. (2021, June 29). An overview of functionalized graphene nanomaterials for advanced applications. Nanomaterials, 11(7), 1717. https://doi.org/10.3390/nano11071717, PubMed: 34209928, PubMed Central: PMC8308136
- Medzhitov, R. (2010). Inflammation 2010: New adventures of an old flame. Cell, 140(6), 771–776. https://doi.org/10.1016/j.cell.2010.03.006, PubMed: 20303867
- Mioc, M., Pavel, I. Z., Ghiulai, R., Coricovac, D. E., Farcaş, C., Mihali, C.-V., Oprean, C., Serafim, V., Popovici, R. A., Dehelean, C. A., Shtilman, M. I., Tsatsakis, A. M., & Şoica, C. (2018, May 3). The cytotoxic effects of betulin-conjugated gold nanoparticles as stable formulations in normal and melanoma cells. Frontiers in Pharmacology, 9, 429. https://doi.org/10.3389/fphar.2018.00429, PubMed: 29773989, PubMed Central: PMC5943567
- Mirza, M. A., Mahmood, S., Hilles, A. R., Ali, A., Khan, M. Z., Zaidi, S. A. A., Iqbal, Z., & Ge, Y. (2023, November 20). Quercetin as a therapeutic product: Evaluation of its pharmacological action and clinical applications-A review. Pharmaceuticals, 16(11), 1631. https://doi.org/10.3390/ph16111631, PubMed: 38004496, PubMed Central: PMC10674654
- Mishra, V., Bansal, K. K., Verma, A., Yadav, N., Thakur, S., Sudhakar, K., & Rosenholm, J. M. (2018, October 18). Solid lipid nanoparticles: Emerging colloidal Nano drug delivery systems. Pharmaceutics, 10(4), 191. https://doi.org/10.3390/pharmaceutics10040191, PubMed: 30340327, PubMed Central: PMC6321253
- Moskovtchenko, J. F., & Cognet, J. B. (1976). Classification des corticoïdes [Classification of corticoids]. Annales de l’Anesthesiologie Française, 17(4), 399–405. French. PMID: 9878.
- Muruganathan, N., Dhanapal, A. R., Baskar, V., Muthuramalingam, P., Selvaraj, D., Aara, H., Shiek Abdullah, M. Z., & Sivanesan, I. (2022, November 20). Recent updates on source, biosynthesis and therapeutic potential of natural flavonoid luteolin: A review. Metabolites, 12(11), 1145. https://doi.org/10.3390/metabo12111145, PubMed: 36422285, PubMed Central: PMC9696498
- Nagoor Meeran, M. F., Goyal, S. N., Suchal, K., Sharma, C., Patil, C. R., & Ojha, S. K. (2018, September 4). Pharmacological properties, molecular mechanisms and pharmaceutical development of Asiatic acid: A pentacyclic triterpenoid of therapeutic promise. Frontiers in Pharmacology, 9, 892. https://doi.org/10.3389/fphar.2018.00892, PubMed: 30233358, PubMed Central: PMC6131672
- Nathan, C., & Ding, A. (2010, March 19). Nonresolving inflammation. Cell, 140(6), 871–882. https://doi.org/10.1016/j.cell.2010.02.029, PubMed: 20303877
- Nie, X., Chen, Z., Pang, L., Wang, L., Jiang, H., Chen, Y., Zhang, Z., Fu, C., Ren, B., & Zhang, J. (2020, December 16). Oral Nano drug delivery systems for the treatment of type 2 diabetes mellitus: An available administration strategy for antidiabetic phytocompounds. International Journal of Nanomedicine, 15, 10215–10240. https://doi.org/10.2147/IJN.S285134, PubMed: 33364755, PubMed Central: PMC7751584
- Ntalouka, F., & Tsirivakou, A. (2023, March 1). Luteolin: A promising natural agent in management of pain in chronic conditions. Frontiers in Pain Research, 4, Article 1114428. https://doi.org/10.3389/fpain.2023.1114428, PubMed: 36937566, PubMed Central: PMC10016360
- Ooi, K. X., Poo, C. L., Subramaniam, M., Cordell, G. A., & Lim, Y. M. (2023, February). Maslinic acid exerts anticancer effects by targeting cancer hallmarks. Phytomedicine, 110, Article 154631. https://doi.org/10.1016/j.phymed.2022.154631 [EPub] 2022 Dec 25. PubMed: 36621168
- Oyagbemi, A. A., Omobowale, T. O., Ola-Davies, O. E., Asenuga, E. R., Ajibade, T. O., Adejumobi, O. A., Afolabi, J. M., Ogunpolu, B. S., Falayi, O. O., Saba, A. B., Adedapo, A. A., & Yakubu, M. A. (2018, November). Luteolin-mediated KIM-1/NF-kB/Nrf2 signaling pathways protects sodium fluoride-induced hypertension and cardiovascular complications. BioFactors, 44(6), 518–531. https://doi.org/10.1002/biof.1449 [EPub]. PubMed: 30474894
- Patil, K. R., Mohapatra, P., Patel, H. M., Goyal, S. N., Ojha, S., Kundu, C. N., & Patil, C. R. (2015, May 4). Pentacyclic triterpenoids inhibit IKKβ mediated activation of NF-κB pathway: In silico and in vitro Evidences. PLOS One, 10(5), Article e0125709. https://doi.org/10.1371/journal.pone.0125709, PubMed: 25938234, PubMed Central: PMC4418667
- Patil, S., & Chandrasekaran, R. (2020, October 26). Biogenic nanoparticles: A comprehensive perspective in synthesis, characterization, application and its challenges. Journal, Genetic Engineering & Biotechnology, 18(1), 67. https://doi.org/10.1186/s43141-020-00081-3, PubMed: 33104931, PubMed Central: PMC7588575
- Patra, J. K., Das, G., Fraceto, L. F., Campos, E. V. R., Rodriguez-Torres, M. D. P., Acosta-Torres, L. S., Diaz-Torres, L. A., Grillo, R., Swamy, M. K., Sharma, S., Habtemariam, S., & Shin, H.-S. (2018, September 19). Nano based drug delivery systems: Recent developments and future prospects. Journal of Nanobiotechnology, 16(1), 71. https://doi.org/10.1186/s12951-018-0392-8, PubMed: 30231877, PubMed Central: PMC6145203
- Pollastri, S., & Tattini, M. (2011, November). Flavonols: Old compounds for old roles. Annals of Botany, 108(7), 1225–1233. https://doi.org/10.1093/aob/mcr234 [EPub]. PubMed: 21880658, PubMed Central: PMC3197460
- Puniani, E., Cayer, C., Kent, P., Mullally, M., Sánchez-Vindas, P., Poveda Álvarez, L., Cal, V., Merali, Z., Arnason, J. T., & Durst, T. (2015, May). Ethnopharmacology of Souroubea sympetala and Souroubea gilgii (Marcgraviaceae) and identification of betulinic acid as an anxiolytic principle. Phytochemistry, 113, 73–78. https://doi.org/10.1016/j.phytochem.2014.02.017 [EPub] 2014 Mar 15. PubMed: 24641939
- Rainsford, K. D. (2007). Anti-inflammatory drugs in the 21st century. Sub-Cellular Biochemistry, 42, 3–27. https://doi.org/10.1007/1-4020-5688-5_1, PubMed: 17612044
- Rani, N., Bharti, S., Bhatia, J., Nag, T. C., Ray, R., & Arya, D. S. (2016, April 25). Chrysin, a PPAR-γ agonist improves myocardial injury in diabetic rats through inhibiting AGE-RAGE mediated oxidative stress and inflammation. Chemico-Biological Interactions, 250, 59–67. https://doi.org/10.1016/j.cbi.2016.03.015 [EPub]. PubMed: 26972669
- Rauf, A., Imran, M., Patel, S., Muzaffar, R., & Bawazeer, S. S. (2017, December). Rutin: Exploitation of the flavonol for health and homeostasis. Biomedicine and Pharmacotherapy, 96, 1559-1561. https://doi.org/10.1016/j.biopha.2017.08.136 [EPub]. PubMed: 28888794
- Rawat, R., Chouhan, R. S., Sadhu, V., & Sharma, M. (2023, May 8). Clarithromycin-loaded submicron-sized carriers: Pharmacokinetics and pharmacodynamic evaluation. Materials, 16(9), 3593. https://doi.org/10.3390/ma16093593, PubMed: 37176475, PubMed Central: PMC10179782
- Ren, K., Jiang, T., Zhou, H.-F., Liang, Y., & Zhao, G.-J. (2018). Apigenin retards atherogenesis by promoting ABCA1-mediated cholesterol efflux and suppressing inflammation. Cellular Physiology and Biochemistry, 47(5), 2170-2184. https://doi.org/10.1159/000491528 [EPub]. PubMed: 29975943
- Ricciotti, E., & FitzGerald, G. A. (2011, May). Prostaglandins and inflammation. Arteriosclerosis, Thrombosis, and Vascular Biology, 31(5), 986–1000. https://doi.org/10.1161/ATVBAHA.110.207449, PubMed: 21508345, PubMed Central: PMC3081099
- Rizvi, S. Z. H., Shah, F. A., Khan, N., Muhammad, I., Ali, K. H., Ansari, M. M., Din, F. U., Qureshi, O. S., Kim, K.-W., Choe, Y.-H., Kim, J.-K., & Zeb, A. (2019, April 5). Simvastatin-loaded solid lipid nanoparticles for enhanced anti-hyperlipidemic activity in hyperlipidemia animal model. International Journal of Pharmaceutics, 560, 136-143. https://doi.org/10.1016/j.ijpharm.2019.02.002 [EPub]. PubMed: 30753932
- Roy, A., Bulut, O., Some, S., Mandal, A. K., & Yilmaz, M. D. (2019, January 21). Green synthesis of silver nanoparticles: Biomolecule-nanoparticle organizations targeting antimicrobial activity. RSC Advances, 9(5), 2673-2702. https://doi.org/10.1039/c8ra08982e, PubMed: 35520490, PubMed Central: PMC9059941
- Sachs, D., Cunha, F. Q., & Ferreira, S. H. (2004, March 9). Peripheral analgesic blockade of hypernociception: Activation of arginine/NO/cGMP/protein kinase G/ATP-sensitive K+ channel pathway. Proceedings of the National Academy of Sciences of the United States of America, 101(10), 3680–3685. https://doi.org/10.1073/pnas.0308382101 [EPub]. PubMed: 14990791, PubMed Central: PMC373522
- Saleem, M. (2009, November 28). Lupeol, a novel anti-inflammatory and anti-cancer dietary triterpene. Cancer Letters, 285(2), 109–115. https://doi.org/10.1016/j.canlet.2009.04.033 [EPub]. PubMed: 19464787, PubMed Central: PMC2764818
- Santos, A., Veiga, F., & Figueiras, A. (2019, December 21). Dendrimers as pharmaceutical excipients: Synthesis, properties, toxicity and biomedical applications. Materials, 13(1), 65. https://doi.org/10.3390/ma13010065, PubMed: 31877717, PubMed Central: PMC6981751
- Satapathy, M. K., Yen, T.-L., Jan, J.-S., Tang, R.-D., Wang, J.-Y., Taliyan, R., & Yang, C.-H. (2021, July 31). Solid lipid nanoparticles (SLNs): An advanced drug delivery system targeting brain through BBB. Pharmaceutics, 13(8), 1183. https://doi.org/10.3390/pharmaceutics13081183, PubMed: 34452143, PubMed Central: PMC8402065
- Serafini, M., Peluso, I., & Raguzzini, A. (2010, August). Flavonoids as anti-inflammatory agents. The Proceedings of the Nutrition Society, 69(3), 273–278. https://doi.org/10.1017/S002966511000162X [EPub]. PubMed: 20569521
- Shabir, I., Pandey, V. K., Dar, A. H., Pandiselvam, R., Manzoor, S., Mir, S. A., Shams, R., Dash, K. K., Fayaz, U., Khan, S. A., Jeevarathinam, G., Zhang, Y., Rusu, A. V., & Trif, M. (2022, September 22). Nutritional profile, phytochemical compounds, biological activities and utilisation of onion peel for food applications: A review. Sustainability, 14(19), Article 11958. https://doi.org/10.3390/su141911958
- Shaikh, S., Verma, H., Yadav, N., Jauhari, M., & Bullangowda, J. (2012). Applications of steroid in clinical practice: A review. International Scholarly Research Notices, 2012, Article 985495.
- Shao, J., Fang, Y., Zhao, R., Chen, F., Yang, M., Jiang, J., Chen, Z., Yuan, X., & Jia, L. (2020, November). Evolution from small molecule to nano-drug delivery systems: An emerging approach for cancer therapy of ursolic acid. Asian Journal of Pharmaceutical Sciences, 15(6), 685–700. https://doi.org/10.1016/j.ajps.2020.03.001 [EPub]. PubMed: 33363625, PubMed Central: PMC7750806
- Sharma, M., Sharma, S., & Wadhwa, J. (2019, December). Improved uptake and therapeutic intervention of curcumin via designing binary lipid nanoparticulate formulation for oral delivery in inflammatory bowel disorder. Artificial Cells, Nanomedicine, and Biotechnology, 47(1), 45–55. https://doi.org/10.1080/21691401.2018.1543191, PubMed: 30663410
- Suchal, K., Malik, S., Gamad, N., Malhotra, R. K., Goyal, S. N., Chaudhary, U., Bhatia, J., Ojha, S., & Arya, D. S. (2016). Kaempferol attenuates myocardial ischemic injury via inhibition of MAPK signaling pathway in experimental model of myocardial ischemia-reperfusion injury. Oxidative Medicine and Cellular Longevity, 2016, Article 7580731. https://doi.org/10.1155/2016/7580731 [EPub]. Erratum in: Oxidative Medicine and Cellular Longevity. (2020, June 15), 2020, Article 4530278. https://doi.org/10.1155/2020/4530278, PubMed: 32655768, PubMed Central: PMC4819110.
- Sysak, S., Czarczynska-Goslinska, B., Szyk, P., Koczorowski, T., Mlynarczyk, D. T., Szczolko, W., Lesyk, R., & Goslinski, T. (2023, May 2). Metal nanoparticle-flavonoid connections: Synthesis, physicochemical and biological properties, as well as potential applications in medicine. Nanomaterials, 13(9), 1531. https://doi.org/10.3390/nano13091531, PubMed: 37177076, PubMed Central: PMC10180592
- Szakiel, A., Pączkowski, C., Pensec, F., & Bertsch, C. (2012, June). Fruit cuticular waxes as a source of biologically active triterpenoids. Phytochemistry Reviews: Proceedings of the Phytochemical Society of Europe, 11(2–3), 263–284. https://doi.org/10.1007/s11101-012-9241-9 [EPub]. PubMed: 23519009, PubMed Central: PMC3601259
- Takeuchi, O., & Akira, S. (2010, March 19). Pattern recognition receptors and inflammation. Cell, 140(6), 805–820. https://doi.org/10.1016/j.cell.2010.01.022, PubMed: 20303872
- Tang, X.-L., Liu, J.-X., Dong, W., Li, P., Li, L., Zheng, Y.-Q., & Hou, J.-C. (2014, June). [Intervention effect of quercetin on inflammatory secretion of cardiac fibroblasts]. Zhongguo Zhong Yao Za Zhi, 39(12), 2314–2317. PubMed: 25244766
- Vasconcelos, J. F., Teixeira, M. M., Barbosa-Filho, J. M., Lúcio, A. S., Almeida, J. R., de Queiroz, L. P., Ribeiro-Dos-Santos, R., & Soares, M. B. (2008, September). The triterpenoid lupeol attenuates allergic airway inflammation in a murine model. International Immunopharmacology, 8(9), 1216–1221. https://doi.org/10.1016/j.intimp.2008.04.011 [EPub]. Erratum in: International Immunopharmacology. (2008, December 10), 8(12), 1714. https://doi.org/10.1016/j.intimp.2008.07.008, PubMed: 18602067.
- Viscusi, G., Paolella, G., Lamberti, E., Caputo, I., & Gorrasi, G. (2023, February 17). Quercetin-loaded polycaprolactone-polyvinylpyrrolidone electrospun membranes for health application: Design, characterization, modeling and cytotoxicity studies. Membranes, 13(2), 242. https://doi.org/10.3390/membranes13020242, PubMed: 36837745, PubMed Central: PMC9965405
- Vyas, N., & Argal, A. (2014). Isolation and characterization of oleanolic acid from roots of Lantana camara. Asian Journal of Pharmaceutical and Clinical Research, 7(7), 189–191. https://journals.innovareacademics.in/index.php/ajpcr/article/view/1116
- Wang, L., Yin, Q., Liu, C., Tang, Y., Sun, C., & Zhuang, J. (2021, July 5). Nanoformulations of ursolic acid: A modern natural anticancer molecule. Frontiers in Pharmacology, 12, Article 706121. https://doi.org/10.3389/fphar.2021.706121, PubMed: 34295253, PubMed Central: PMC8289884
- Wang, S., Su, R., Nie, S., Sun, M., Zhang, J., Wu, D., & Moustaid-Moussa, N. (2014, April). Application of nanotechnology in improving bioavailability and bioactivity of diet-derived phytochemicals. The Journal of Nutritional Biochemistry, 25(4), 363–376. https://doi.org/10.1016/j.jnutbio.2013.10.002 [EPub] 2013 Nov 5. PubMed: 24406273, PubMed Central: PMC3959237
- Woźniak, Ł., Skąpska, S., & Marszałek, K. (2015, November 19). Ursolic acid-A pentacyclic triterpenoid with a wide spectrum of pharmacological activities. Molecules, 20(11), 20614–20641. https://doi.org/10.3390/molecules201119721, PubMed: 26610440, PubMed Central: PMC6332387
- Xia, E.-Q., Wang, B.-W., Xu, X.-R., Zhu, L., Song, Y., & Li, H.-B. (2011). Microwave-assisted extraction of oleanolic acid and ursolic acid from Ligustrum lucidum Ait. International Journal of Molecular Sciences, 12(8), 5319–5329. https://doi.org/10.3390/ijms12085319 [EPub]. PubMed: 21954361, PubMed Central: PMC3179168
- Xia, X., Liu, H., Lv, H., Zhang, J., Zhou, J., & Zhao, Z. (2017, May 9). Preparation, characterization and in vitro/vivo studies of oleanolic acid-loaded lactoferrin nanoparticles. Drug Design, Development and Therapy, 11, 1417–1427. https://doi.org/10.2147/DDDT.S133997, PubMed: 28533680, PubMed Central: PMC5431734
- Xiong, D., Hu, W., Ye, S.-T., & Tan, Y.-S. (2018, November 17). Isoliquiritigenin alleviated the Ang II-induced hypertensive renal injury through suppressing inflammation cytokines and oxidative stress-induced apoptosis via Nrf2 and NF-κB pathways. Biochemical and Biophysical Research Communications, 506(1), 161–168. https://doi.org/10.1016/j.bbrc.2018.09.013 [EPub]. PubMed: 30340829
- Yadav, D. Y., Suri, S., Choudhary, A., Sikender, M., Hemant, H., Beg, M. N., Garg, V., Ahmad, A., & Asif, M. (2011). Novel approach: Herbal remedies and natural products in pharmaceutical science as nano drug delivery systems.
- Yamashita, K., Lu, H., Lu, J., Chen, G., Yokoyama, T., Sagara, Y., Manabe, M., & Kodama, H. (2002, November). Effect of three triterpenoids, lupeol, betulin and betulinic acid on the stimulus-induced superoxide generation and tyrosyl phosphorylation of proteins in human neutrophils. Clinica Chimica Acta; International Journal of Clinical Chemistry, 325(1–2), 91–96. https://doi.org/10.1016/s0009-8981(02)00252-8, PubMed: 12367771
- Yang, C., Guo, Y., Huang, T.-S., Zhao, J., Huang, X.-J., Tang, H.-X., An, N., Pan, Q., Xu, Y.-Z., & Liu, H.-F. (2018, November). Asiatic acid protects against cisplatin-induced acute kidney injury via anti-apoptosis and anti-inflammation. Biomedicine and Pharmacotherapy, 107, 1354–1362. https://doi.org/10.1016/j.biopha.2018.08.126 [EPub]. PubMed: 30257350
- Yang, E.-J., Lee, W., Ku, S.-K., Song, K.-S., & Bae, J.-S. (2012, May). Anti-inflammatory activities of oleanolic acid on HMGB1 activated HUVECs. Food and Chemical Toxicology, 50(5), 1288–1294. https://doi.org/10.1016/j.fct.2012.02.026 [EPub]. PubMed: 22386814
- Yatoo, M. I., Gopalakrishnan, A., Saxena, A., Parray, O. R., Tufani, N. A., Chakraborty, S., Tiwari, R., Dhama, K., & Iqbal, H. M. N. (2018). Anti-inflammatory drugs and herbs with special emphasis on herbal medicines for countering inflammatory diseases and disorders – A review. Recent Patents on Inflammation and Allergy Drug Discovery, 12(1), 39–58. https://doi.org/10.2174/1872213X12666180115153635, PubMed: 29336271
- Yetisgin, A. A., Cetinel, S., Zuvin, M., Kosar, A., & Kutlu, O. (2020, May 8). Therapeutic nanoparticles and their targeted delivery applications. Molecules, 25(9), 2193. https://doi.org/10.3390/molecules25092193, PubMed: 32397080, PubMed Central: PMC7248934
- Yu, D., Kan, Z., Shan, F., Zang, J., & Zhou, J. (2020, December 7). Triple strategies to improve oral bioavailability by fabricating Coamorphous forms of ursolic acid with piperine: Enhancing water-solubility, permeability and inhibiting cytochrome P450 isozymes. Molecular Pharmaceutics, 17(12), 4443–4462. https://doi.org/10.1021/acs.molpharmaceut.0c00443 [EPub]. PubMed: 32926628
- Yu, Y.-B., Miyashiro, H., Nakamura, N., Hattori, M., & Park, J. C. (2007, July). Effects of triterpenoids and flavonoids isolated from Alnus firma on HIV-1 viral enzymes. Archives of Pharmacal Research, 30(7), 820–826. https://doi.org/10.1007/BF02978831, PubMed: 17703732
- Yun, Y., Han, S., Park, E., Yim, D., Lee, S., Lee, C.-K., Cho, K., & Kim, K. (2003, December). Immunomodulatory activity of betulinic acid by producing pro-inflammatory cytokines and activation of macrophages. Archives of Pharmacal Research, 26(12), 1087–1095. https://doi.org/10.1007/BF02994763, PubMed: 14723345
- Zhai, J., Dong, X., Yan, F., Guo, H., & Yang, J. (2022, February 21). Oleandrin: A systematic review of its natural sources, structural properties, detection methods, pharmacokinetics and toxicology. Frontiers in Pharmacology, 13, Article 822726. https://doi.org/10.3389/fphar.2022.822726, PubMed: 35273501, PubMed Central: PMC8902680
- Zhao, M., Wu, F., Tang, Z., Yang, X., Liu, Y., Wang, F., & Chen, B. (2023, September 28). Anti-inflammatory and antioxidant activity of ursolic acid: A systematic review and meta-analysis. Frontiers in Pharmacology, 14, Article 1256946. https://doi.org/10.3389/fphar.2023.1256946, PubMed: 37841938, PubMed Central: PMC10568483
- Zhou, Y., Hong, Y., & Huang, H. (2016). Triptolide attenuates inflammatory response in membranous glomerulo-nephritis rat via downregulation of NF-κB signaling pathway. Kidney and Blood Pressure Research, 41(6), 901–910. https://doi.org/10.1159/000452591 [EPub]. PubMed: 27871079