Next Article in Journal
Special Issue “Role of Molecular Dynamics Simulations and Related Methods in Drug Discovery”
Previous Article in Journal
Redefining Chemotherapy-Related Headaches: From Pathobiology to Differential Diagnosis and Management
Previous Article in Special Issue
Liver-Targeted Scutellarin Nanoemulsion Alleviates Fibrosis with Ancillary Modulation of the Gut–Liver Microbiota
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Harnessing Moringa oleifera for Immune Modulation in Cancer: Molecular Mechanisms and Therapeutic Potential

by
Mounir Tilaoui
1,*,
Jamal El Karroumi
2,
Hassan Ait Mouse
3 and
Abdelmajid Zyad
3
1
Laboratory of Biotechnology, Environment, Agrifood, and Health, Department of Biology, Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
2
Laboratory of Biomolecular Chemistry, Natural Substances and Reactivity, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech 40000, Morocco
3
Laboratory of Agro-Industrial and Medical Biotechnology, Team of Experimental Oncology and Natural Substances, Cellular and Molecular Immuno-Pharmacology, Faculty of Sciences and Technology, Sultan Moulay Slimane University, BP 523, Beni-Mellal 23000, Morocco
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2026, 27(1), 263; https://doi.org/10.3390/ijms27010263 (registering DOI)
Submission received: 25 July 2025 / Revised: 26 September 2025 / Accepted: 19 December 2025 / Published: 26 December 2025
(This article belongs to the Special Issue Natural Products in Drug Discovery and Development)

Abstract

Moringa oleifera, widely recognized as the horseradish tree or drumstick tree, is classified within the Moringaceae family, which comprises 13 species predominantly distributed across tropical and subtropical regions. The plant possesses a variety of therapeutic, nutritional, and beneficial health properties, including its potential to enhance the immune system. The present work provides extensive bibliographic research addressing the chemical composition of Moringa oleifera and its immunomodulatory properties with a focus on the cellular and molecular mechanisms involved in the regulation of immune function, which is crucial in unchecked cell proliferation and metastasis. The chemical composition of Moringa oleifera, including kaempferol, chlorogenic acid, quercetin, and niazimicin, varies between different biological parts of the plant (seeds, leaves, roots, and stems). The presence of these various chemical compounds contributes to the plant’s effect on the immune response via different pathways. Several studies indicate that Moringa oleifera mitigates inflammation by suppressing key pro-inflammatory mediators, such as TNF-α, IL-1β, inducible nitric oxide synthase (iNOS), prostaglandin E2 (PGE-2), and cyclooxygenase-2 (COX-2), while simultaneously enhancing anti-inflammatory mediators through activation of PPAR-γ. Furthermore, the immunomodulatory properties and possible application in health promotion and disease prevention, especially in cancer therapy, are discussed. Studies indicate that Moringa oleifera can modulate the tumor microenvironment (TME) by reducing Treg polarization, enhancing NK cell cytotoxicity, and prompting the proliferation and clonal expansion of CD8+ and CD4+ T lymphocytes. Together, Moringa oleifera could be considered for the treatment of conditions related to immune dysregulation, such as cancer.

1. Introduction

The treatment of immune-mediated inflammatory diseases, such as asthma, allergies and autoimmune diseases due to disorders of the immune system requires the modulation of the innate and adaptive immune responses [1]. Immunomodulation refers to the regulation or alteration of the immune system’s activity in response to a given internal or external stimulus, often achieved through the administration of specific compounds or drugs [2]. There are numerous chemical immunomodulators, including immunosuppressive and immunostimulatory agents such as dexamethasone, fluticasone, prednisolone, prednisone, and hydrocortisone, which have been used to treat and modulate various inflammatory diseases.
Recombinant therapeutic proteins have gained increasing importance as promising treatments for immunodeficiency, infectious diseases, and cancer [3,4,5]. Cyclosporin A is an example of a fungal cyclic peptide widely used as an immunosuppressant drug to prevent rejection in organ-transplant recipients and to treat patients with autoimmune diseases. Nevertheless, most of these conventional drugs have been associated with several adverse effects [6,7]. Duodenal mucosa and gastric mucosal epithelium damage are the commonest adverse effects caused by non-steroidal anti-inflammatory drugs [8,9,10,11]. Other side effects, notably increased skin fragility and reduced bone marrow function, have been linked to the use of immunosuppressive drugs such as corticosteroids [12,13,14,15].
Cyclosporin A is known to cause a variety of adverse metabolic side effects and toxicities, including hypomagnesemia, hypertension, hypercalciuria, hyperkalemia, hepatotoxicity, nephrotoxicity, and gingival hypertrophy [16]. Therefore, new, safer, and more effective drugs are required as alternatives to current treatments.
Natural products are still a significant source of leads for new immunomodulatory drugs. Many plants were reported to exhibit immunomodulatory activities and to modulate immune functions. The present work provides extensive bibliographic research addressing the chemical composition of Moringa oleifera and its effect on the immune system, exploring its cellular activities and its molecular impacts on immunoglobulins, regulatory pathways, and cytokines.

2. Methods/Literature Search Strategy

This comprehensive narrative review was conducted to explore the immunomodulatory and therapeutic potential of Moringa oleifera, especially on cancer therapy, with an emphasis on molecular mechanisms, and the phytochemical composition of its various parts (leaves, seeds, pods, flowers, and roots). Relevant studies published primarily between 2020 and 2025 were conducted across databases of PubMed/Medline, Scopus, Web of Science, Google Scholar, and Cochrane Library to identify pertinent articles. The search terms used included “Moringa oleifera” “chemical composition” “immune system” “immune modulation” “ant-inflammatory” “cytokines” “immunotherapy” “cancer” and “molecular mechanism”. Boolean operators (AND, OR) were used to effectively optimize the combination of these keywords for the literature research.
Studies were selected based on their relevance to preclinical, mechanistic, and clinical evidence, prioritizing peer-reviewed reports. Articles lacking methodological rigor, sufficient experimental detail, or unrelated to the topic were excluded. Although a formal risk-of-bias assessment was not conducted, priority was given to high-quality experimental and clinical studies to ensure the robustness, reliability, and validity of the synthesized findings.

3. Chemical Composition of Moringa oleifera

Each component of the plant, the leaf, root, bark, seed, flower, and pod, contains important nutritional resources [17]. Over 100 chemical constituents have been identified in Moringa oleifera, including proteins, fats, vitamins, minerals, carbohydrates, and dietary fibers [18]. Moreover, a multitude of phytochemicals have been identified in various parts of this plant, each offering diverse health-promoting properties [19,20].

3.1. Seeds

The seeds are spherical and enclosed within a semi-permeable brown seed coat, characterized by three papery white wings. A single tree can produce approximately 15,000 to 25,000 seeds per year [21,22], each with an average mass of 0.3 g, of which about 25% is attributed to the seed coat [23]. The oil extracted from Moringa oleifera seeds, globally known as Ben oil, has a yield of approximately 40% [24]. According to Corbett et al., the seed oil of Moringa oleifera is of the oleic type with a high content of oleic acid [25]. This characteristic is particularly advantageous in the context of growing preference for replacing polyunsaturated vegetable oils with those rich in monounsaturated fatty acids, which exhibit high oxidative and thermal stability, even under high-temperature conditions such as frying. Moringa oleifera seed oil contains all the major fatty acids present in olive oil and is also distinguished by its notable concentration of behenic acid (C22:0), ranging from 5 to 6%. In addition, it contains small amounts of gadoleic acid (C20:1) and lignoceric acid (C24:0), along with trace levels of erucic acid (C22:1) and cerotic acid (C26:0). Figure 1 illustrates the main fatty acids found in the oil of Moringa oleifera seeds [26]. A study has shown that Moringa oil is rich in tocopherols, which are fat-soluble vitamins and powerful natural antioxidants [27].

3.2. Leaves

Nutritional analyses have shown that Moringa oleifera leaves are rich in vitamins, minerals, and proteins [28]. Through the process of hydrodistillation, the leaves of Moringa oleifera yielded a pale-yellow oil, accounting for 0.05% of the dry mass [29]. The first studies on Moringa oleifera date back to the 1970s. Recent comprehensive phytochemical analyses have identified more than 90 to over 100 compounds in Moringa oleifera leaves, including phenols, flavonoids, amino acids, and sugars, highlighting its diverse bioactive profile [20,30]. In 1982, several sugars such as L-arabinose, D-galactose, glucuronic acid, L-rhamnose, D-mannose, and D-xylose were identified in the following proportions: 14.5:11.3:3:2:1:1 [31]. Subsequently, in 1994, two nitrile glycosides, niazirin and niazirinin, were isolated by Shaheen Faizi et al. [32]. The same authors successfully identified glucosinolates, which, upon enzymatic hydrolysis by myrosianse, can produce isothiocyanates such as 4-[(α-L-rhamnosyloxy) benzyl]isothiocyanate, known for their various biological activities [33,34,35,36]. Moringa oleifera is known for its antioxidant activities, which are attributed to the presence of several flavonoids such as rutin, kaempferol, and quercetin [37] (Figure 2).
Recent investigations suggest that isothiocyanate and astragalin serve as key chemical markers for standardizing Moringa oleifera extracts [38], given their well-documented anti-inflammatory and antioxidant properties [39]. To quantify isothiocyanate, a specific HPLC-based cyclocondensation method was developed and applied to analyze extracts from various plant parts, such as leaves, immature seeds, mature seeds, and pods. The leaves of Moringa oleifera are particularly abundant in bioactive compounds, among which phenolic acids represent a prominent class [40]. Several studies have been conducted to identify and quantify these compounds with Moringa oleifera leaf extract. Among the most frequently reported phenolic acids are gallic acid, ferulic acid, and o-coumaric acid [41]. Figure 3 illustrates the major phenolic acids identified in Moringa oleifera leaves.

3.3. Roots

While the leaves of Moringa oleifera are the most commonly utilized, the roots also exhibit notable nutritional and therapeutic properties [19,42]. The root composition includes carbohydrates, proteins, minerals, and dietary fiber, along with important levels of antioxidant compounds such as phenolics and flavonoids [43]. Although these bioactive constituents are present in smaller concentrations compared to the leaves, the roots still provide essential vitamins, notably vitamin C, vitamin E, and multiple B-complex vitamins.

3.4. Stems

In addition to the roots, other parts of Moringa oleifera, including the stem, bark, and flowers, also exhibit a rich profile of bioactive phytochemicals [44]. While their chemical composition largely overlaps with that of leaves, these plant components represent significant sources of alkaloids, flavonoids, and glycosides. Moreover, compounds including steroids, terpenoids, and phenolic acids have also been identified [45]. It is worth noting that the chemical composition may be influenced by several factors, such as the plant’s geographical origin, age, genetic variability, and the extraction techniques employed [46,47,48,49].

4. The Impact of Moringa oleifera on the Immune System

4.1. Moringa oleifera Effect on Innate and Adaptive Immunity

Moringa oleifera is a widely known medicinal plant indigenous to the Indian subcontinent. It has been used for centuries in Ayurvedic medicine for its healing, nutritional, rejuvenating, and preventive properties [30,50,51]. Recent investigations have underscored its potential in modulating immune system activities [52,53,54,55,56]. The plant harbors various bioactive compounds, including flavonoids, polyphenols, and ascorbic acid, which contribute to its immunomodulatory effects [56,57].
Evidence suggests that Moringa oleifera extracts can increase the production of immune cells such as lymphocytes and macrophages [58,59,60,61]. This is important, as these cells play a critical role in fighting infections within the body. Notably, Moringa oleifera exhibits anti-inflammatory activity that reduces inflammation, thereby promoting improved immune function [62]. Overall, the research indicates that Moringa oleifera exerts beneficial effects on both innate and adaptive immune systems (Table 1 and Table 2). To better elucidate the mechanisms by which Moringa oleifera modulates immunity, we investigated the associated cellular responses and cytokine signaling in both innate and adaptive immunity.

4.2. Cellular Responses and Cytokine Pathways in Innate and Adaptive Immunity

Recent evidence identifies Moringa oleifera as a source of bioactive compounds with immunomodulatory effect [54,58,60,81]. In vivo, Moringa oleifera leaf extract enhanced natural killer cell activity and stimulated cytokine production [64], highlighting its potential to modulate innate immune surveillance with potential translational relevance for immune-based therapies.
The modulatory properties of Moringa oleifera on immune cell activation were confirmed by Zhuping Dong et al. [96]. The result shows through an in vitro experiment that the polysaccharide MOP-1 (glucose, galactose, and arabinose) enhances the proliferation and activation of immune cells through increasing the production of reactive oxygen species, nitric oxide, along with pro-inflammatory mediators IL-6 and TNF-α through upregulation of their mRNA expression in RAW 264.7 cells (Figure 4) [96].
The Moringa oleifera roots polysaccharide MRP-1 was also reported to significantly potentiate anti-inflammatory effects observed in both in vitro and in vivo models via attenuating the release of nitric oxide and iNOS mRNA expression induced by TNF-α, suggesting its potential use as a natural treatment for inflammatory diseases (Figure 4) [97]. Building on the characterization of cellular responses and cytokine pathways, we next explored how these mechanisms orchestrate multilevel modulation of innate and adaptive immunity.

4.3. Multilevel Modulation of Innate and Adaptive Immunity

4.3.1. Gut Microbiota Modulation

Moringa oleifera promotes a balanced gut microbiota, enhancing immune homeostasis, reducing inflammation, and improving immune function [56]. It has also been demonstrated in animal experimental models that Moringa oleifera polysaccharides participate in the homeostasis of the gut microbiota by enhancing the proliferation of beneficial bacterial populations in the human gut, including Lactobacillus species, and a decrease in the abundance of pathogenic bacteria in C57BL/6 mice. Moringa oleifera polysaccharides (MOPs) modulate gut microbiota through prebiotic activity, selectively enriching beneficial bacteria, including Bacteroides and Lactobacillus, while inhibiting pathogenic bacteria, thereby enhancing microbial diversity and homeostasis [98,99,100]. Fermentation of MOPs by gut microbiota produces short-chain fatty acids (SCFAs) such as butyrate, acetate and propionate, which act via G-protein-coupled receptors (GPR41/43) to strengthen intestinal barrier integrity, regulate mucosal immunity, and suppress pro-inflammatory NF-κB signaling pathways [101,102,103]. Furthermore, MOPs influence the gut-associated lymphoid tissue (GALT), enhancing a balanced Treg/Th17 cell ratio and promoting anti-inflammatory cytokines production (e.g., IL-10, TGF-β and IL-35) while decreasing pro-inflammatory cytokines (e.g., IL-6, TNF-α) [104,105,106,107].
Together, these mechanisms form an integrated bidirectional gut-immune network, linking microbial communities to systemic immune homeostasis and highlighting the therapeutic potential of MOPs in managing inflammatory and immune-related disorders [98,106,107,108]. The composition of gut microbiota plays a central role in shaping immune responses and preserving systemic homeostasis. Moringa oleifera supports this balance through promoting beneficial microbes, which in turn enhances anti-inflammatory signaling and immune homeostasis.

4.3.2. Immune Modulation and Anti-Inflammatory Balance

Moringa oleifera promotes immune homeostasis through enhancing anti-inflammatory cytokines while suppressing proinflammatory mediators. Its bioactive compounds act via key signaling pathways, including MAPK, NF-κB and PI3K/AKT/mTOR [35,109,110,111], thereby mitigating tissue damage and improving outcomes in inflammatory disorders. Additionally, Moringa oleifera polysaccharides were found to modulate the immune system, to increase the levels of anti-inflammatory cytokines, and reduce the inflammatory bowel disease [102]. Moreover, administration of Moringa leaf extract at a concentration of 100 mg/kg to mice enhances the production of eosinophil cells, immature neutrophils, and lymphocytes while reducing the amount of segmented neutrophils and monocytes [70]. Another study investigates the impact of Moringa oleifera leaf extract on proliferation capacity, physiological parameters, immune responses, and the expression of immunity-related genes in prawns exposed to both Vibrio anguillarum and ammonia stress [112]. The results showed that administration of 0.5% Moringa oleifera in prawns -after being exposed to ammonia stress- increases the levels of haemolymph SOD, glutathione peroxidase, NO, iNOS, and HSP70 upregulation in hepatopancreas while reducing the levels of haemolymph catalase, peroxiredoxin 5 in hepatopancreas, and nuclear factor-kappa-B-inhibitor alpha (IκB-α) expression compared to the control group. These findings indicate that Moringa oleifera leaf extract has a protective effect against ammonia-induced stress [112]. Similarly, Z Abidin et al. demonstrated that feeding whiteleg shrimp with 2.5 g of Moringa oleifera leaves’ extract significantly enhanced the innate immunity of the shrimp by increasing the production of total hemocyte count, phenoloxidase, phagocytic rate, phagocytic index, and superoxide anion. Also, the extract improved the shrimp’s resistance against Vibrio alginolyticus, a common pathogen in shrimp farming [113]. These findings suggest that consumption of moringa in the diet may increase the upregulation of certain genes associated with the immune system involved in catalytic activity, antimicrobial defense, antioxidant enzyme activity, and blood clotting protein production.
In agreement with the previous studies, Moringa oleifera leaves ethanolic extract exhibits immunomodulatory effects in broiler chicks at a dose of 200 mg/L in drinking water by increasing differential and total leukocyte counts, enhancing both the phagocytic index and phagocytic activity, elevating serum hemagglutination inhibition antibody levels against Newcastle disease virus, and reducing the mortality rate. In addition, it promotes the immune system function of the Bursa of Fabricius, spleen, and thymus indices, and stimulates lymphocytes proliferation induced by the Moringa oleifera leaves ethanolic extract [114]. These findings into multilevel immune regulation provide a foundation for assessing how Moringa oleifera engages these mechanisms to exert immunomodulatory and anti-inflammatory effects.

4.4. Immunomodulatory and Anti-Inflammatory Role of Moringa oleifera

Moringa oleifera has gained increasing attention as a bioactive plant for its promising immunomodulatory and anti-inflammatory properties [79,80,102,115,116], with accumulating evidence demonstrating its ability to regulate essential cytokine-mediated pathways [79,102,116,117,118], modulate inflammatory signaling cascades [74,102,116,119,120], restore immune homeostasis, and improve overall immune functions [50,56,82,102,107,116,121,122]. These effects have been observed across various experimental models and clinical settings [123,124,125,126,127], underscoring its promise as a natural therapeutic intervention for the prevention and management of chronic inflammatory and immune-mediated conditions.
The protective effects of Moringa oleifera are mediated through multiple molecular pathways. Recent studies have elucidated that its phytoconstituents exhibit multifaceted mechanisms, including the suppression of pro-inflammatory mediators such as TNF-α, IL-1β, iNOS, PGE-2, and COX-2 [76,79,111,128], alongside the activation of anti-inflammatory signaling pathways through PPAR-γ, NF-κB and MAPK [107,119,129,130,131]. Beyond innate immune regulation, Moringa oleifera influences adaptive immunity by enhancing antibody responses and modulation the PI3K/AKT/mTOR axis [34,81,119,132,133], an important regulator of cellular homeostasis and immune metabolism. Supported by these mechanistic observations, an important in vivo study examined the potential protective properties of Moringa oleifera ethanolic leaf extract (MOE) in Oreochromis niloticus to mitigate oxidative stress and immune dysfunction [72] caused by abamectin, an insecticide and acaricide known for its toxic activities on aquatic organisms, also shown to have immunosuppressive activity on Oreochromis niloticus by reducing the levels of lysozyme, nitric oxide, and IgM. The study demonstrated that the consumption of MOE dietary supplements ameliorated the adverse impact of Agaricus blazei Murrill (ABM), a mushroom known for its immunomodulatory activities, on lysozyme, nitric oxide, and IgM levels in the ABM + MOE-treated Oreochromis niloticus group [72].
The immunomodulatory effect of MOE could be attributed to cinnamic acid, naringenin, as well as additional phenolic and flavonoid compounds [72]. Importantly, administration of MOE in the treated group with ABM has been demonstrated to suppress the release of pro-inflammatory cytokines, including TNF-α and IL-1β, while upregulating the expression of genes involved in anti-inflammatory pathways, such as TGF-β and IL-10β. Additionally, Moringa oleifera pod extract inhibits the activation of mitogen-activated protein kinases (MAPKs) signaling cascades, thereby further contributing to the attenuation of inflammatory responses [72].
The protective effect of MOE on liver diseases [134,135,136,137] is probably due to the presence of quercetin [138], which plays a pivotal role in the prevention of liver inflammation through inhibiting NF-kB/TLR/NLRP3, inactivation of autophagy-mediated cell apoptosis (under ER stress) through the mTOR pathway [139,140,141,142,143] (Figure 5). Alterations in the mTOR signaling pathway have been associated with various human pathologies, notably liver diseases, and have also been correlated with the inhibition of cancer cell growth through suppression of the AKT/PI3K/mTOR axis [110,144] (Figure 5). Another recent study conducted by Adewale et al. demonstrates that the ethanolic extract of Moringa oleifera leaves exhibits an immunomodulatory effect by triggering programmed cell death pathway (apoptosis) in Jurkat cancer cells through up-regulation of CCR7, especially when those cancer cells were stimulated with CD3 and CD54 or CD28 [145]. Oral administration of aqueous extract of red Moringa oleifera leaves at 42 mg/kg body weight (BW) increased the number of CD4+CD62L+ and CD8+CD62L+ T cells in Salmonella typhimurium-infected BABL/c mice [145]. In a related study, the anti-inflammatory and immunostimulant effects of Moringa oleifera ethanolic extract were determined. The extract leads to a decrease in TNF-α on benzene-induced leukemia in Wistar rats, demonstrating its promising immunotherapy for leukemia. It has been reported that the reduction in serum levels of tumor necrosis factor alpha (TNF-α) following systemic chemotherapy in lymphoproliferative malignancies (leukemia and lymphoma) in patients is linked with response rates [146,147].
Similarly, MOEs have been shown to exert immunostimulatory effects in cyclophosphamide–induced immunosuppressed Swiss albino mice [148], supporting their potential to modulate immune responses under immunosuppressive conditions. This study found that gavage with seed and root extracts at a dose of 2 g/kg BW for seven consecutive days increased the levels of white blood cells, relative spleen and thymus weights, and enhanced immunity by supporting the proliferation and activation of lymphocytes [148]. The study indicated that the extracts decrease the delayed type of hypersensitivity response, enhance the concentration of serum IgM, and increase relative spleen weight, which was associated with a higher increase in white blood cells. Also, it was demonstrated that TNF-α and IL-2 concentrations were up-regulated (Figure 5) in the treated group with Moringa oleifera extracts. The study concluded that Moringa oleifera extracts could improve patients with common variable prognosis with immunodeficiency disorders through the stimulation of the activity of hematopoietic cells, bone marrow, lymphocytes, cellular and humoral immunity [148]. These immunomodulatory and anti-inflammatory properties form the basis for investigating the molecular mechanisms by which Moringa oleifera impacts cancer progression.

5. Moringa oleifera and Cancer Progression: Molecular Mechanisms and Therapeutic Potential

Moringa oleifera exhibits broad cytotoxic and antitumor activities through multiple mechanisms, including induction of apoptosis, cell-cycle arrest, modulation of oxidative stress via Nrf2 activation, and suppression of pro-oncogenic signaling pathways NF-κB/MAPK and inhibition of oncogenic kinases like CDK2 and Src [34,81,149,150]. Furthermore, its bioactive compounds, especially moringa isothiocyanate (MIC-1; also called moringin) and niazimicin, modulate the tumor microenvironment (TME) by increasing cytotoxic T lymphocyte and NK cell activity while decreasing Treg-mediated immunosuppression [34,110,120,151,152].

5.1. Apoptosis, Proliferation, and Cell-Cycle Control in Moringa oleifera-Mediated Cancer Progression

Moringa oleifera extract from leaves, seeds, and oils along with the purified MIC-1 have been shown to reduce tumor cell viability and clonogenicity through triggering caspase-dependent apoptosis and arresting cells at the G1/S checkpoint. In renal cancer cells, MIC-1 suppresses growth and migration by inhibiting PTP1B-dependent Src/Ras/Raf/ERK signaling, enhances the pro-apoptotic Bax/Bcl-2 ratio in xenograft models while inhibiting ERK phosphorylation (Figure 5), establishing a mechanistic bridge between upstream phosphatase targeting and downstream caspase-driven apoptosis [34]. Furthermore, phenolic fractions from Moringa oleifera leaves induce activation of caspases 3/7/9, alongside mitochondrial ROS production and AIF translocation in melanoma cancer cells, suggesting simultaneous caspase-dependent and independent apoptosis signaling pathways [153].
In liver cancer, more particularly hepatocellular carcinoma (HepG2), phenolic compounds from Moringa oleifera fruit and leaves initiate ROS-mediated cellular signaling cascade, trigger caspase-3 activity and drive mitochondrial pathway-dependent apoptosis [152]. Concurrently, another study combining in-silico CDK2 docking with in vitro validation in ER-positive breast cancer reported that Moringa oleifera phytochemicals, including chlorogenic acid, niazirin, kaempferol, quercetin and ellagic acid act as putative CDK2 inhibitors, supporting the inhibition of cell growth through promoting cell cycle arrest at G1/S in hormone-receptor-positive disease [154]. Beyond its role in epithelial cancers, Moringa oleifera leaf protein decreases proliferation, induces apoptosis and promotes cell-cycle arrest in Jurkat T-ALL cells via MAPK/AKT pathway modulation (Figure 5), underscoring its relevance in hematologic malignancies [81].
The antiproliferative and pro-apoptotic signature is consistently observed across additional cancer cell lines, with A549 (NSCLC) cells displaying caspase 9/3 activation and Bax/Bcl-2 shifts in response to Moringa oleifera-based extracts [155]. Similarly, colon cancer cells respond to glucosinolate-rich lead fractions with decreased proliferation [35,156], and Dalton’s lymphoma cells demonstrate both in vitro and in vivo growth inhibition with upregulated Bax, cytochrome-c release and cleaved caspase 3 along with decreased Bcl-2 expression, accompanied by cell cycle arrest at G2/M, apoptosis induction, and importantly, significant improvement in hematological parameters in mice bearing Dalton’s Lymphoma tumors [157]. Collectively, these studies converge on (1) caspase cascade engagement, (2) suppression of pro-survival signals such as ERK, and (3) CDK-axis inhibition at G1/S, providing a mechanistic basis for Moringa oleifera constituents as adjuncts to cytotoxic antitumor or targeted therapies, while emphasizing the need for standardized preparations to ensure reproducibility across experimental models.

5.2. Moringa oleifera in Cancer: Dual Regulation of Oncogenic Signaling and Oxidative Stress

Recent investigations have highlighted the dual modulatory effects of Moringa oleifera constituents on oncologic signaling and redox homeostasis, elucidating their potential as adjuncts in cancer therapy. Notably, the MIC-1 has been shown to inhibit migration and proliferation in renal cancer cells through modulation of PTP1B-dependent Src/Ras/Raf/ERK signaling [34]. This inhibition leads to reduced phosphorylation of Src (Tyr416), K-Rad, Raf and ERK1/2 [34], thus impairing tumor cell growth and migration. Concurrently, MIC-1 triggers the Nrf2/ARE pathway, upregulating the expression of cytoprotective genes such as NQO1 and enhancing cellular defense against oxidative stress [34]. This dual activity may potentially modulate tumor cell adaptation under stress conditions. In addition, in vivo studies further support these findings, showing that MIC-1 treatment results in increased Bax/Bcl-2 ratios in tumor tissues, a pivotal indicator of the induction of apoptosis [34]. Also, it has been reported that Moringa oleifera extracts modulate key oncogenic and stress-related signaling pathways implicated in multiple cancer types, such as breast, hepatocellular, melanoma, colon, NSCLC (Non-Small Cell Lung Cancer) and hematologic malignancies. These effects are mediated through the coordinated regulation of Nrf2-Keap1, TLR4/NF-κB and Wnt/β-catenin, thereby orchestrating oxidative stress and inflammatory responses, processes that are crucial to tumor growth, survival and therapy resistance [152].

5.3. Inhibition of Angiogenesis and Metastasis Progression

Recent studies have elucidated the potential role of Moringa oleifera extracts to modulate angiogenesis and metastasis processes (Figure 5), which are essential for tumor progression [158,159,160]. Although direct evidence of anti-angiogenic effect remains limited, several in vitro and in vivo investigations have demonstrated inhibition of cell migration and downregulation of metastasis-associated pathways. In a recent study, Raju and Kei (2022) reported that Moringa oleifera leaf extract exhibits significant anti-angiogenic activity, with the 100% aqueous extract showing an 81.33% reduction in vascular formation in the chick chorioallantoic membrane assay [161], suggesting the presence of bioactive compounds capable of inhibiting angiogenesis, a key process in tumor growth and metastasis. Furthermore, studies on Moringa oleifera-silver nanoparticles (MO-AgNPs) have been shown to inhibit metastasis through modulating pathways involving Snail and TGF-β. These findings suggest that MO-AgNPs may serve as a promising and effective targeted strategy to inhibit tumor metastasis [162].
Emerging evidence suggests that Moringa oleifera possesses significant capacity to modulate immune-regulatory networks within the TME. Notably, a recent study demonstrated that Moringa oleifera leaf protein inhibited the proliferation of T-lymphoblastic leukemia cells by modulating immune-related signaling pathways, specifically the MAPK (p38 and ERK) and AKT axis (Figure 5), while simultaneously inducing apoptosis, cell-cycle arrest, and autophagy [81]. Additionally, recent investigations have further revealed the ability of Moringa oleifera-derived polysaccharides to reprogram the TME through targeting innate immune cells. In a Lewis lung carcinoma model, Moringa oleifera leaf polysaccharides were shown to remodel tumor-associated macrophages from a pro-tumoral M2 phenotype toward an antitumoral M1 phenotype. This repolarization was mediated via TLR4 signaling, leading to upregulated chemokine expression CXCL9 and CXCL10 and increased intra-tumoral T-cell infiltration, thereby exerting powerful immune-activating effects [58]. In parallel, Moringa oleifera seed oil extract has demonstrated antioxidant, anti-inflammatory, and antitumor activities in Ehrlich ascites carcinoma-bearing Swiss albino mice treated at a dose of 500 mg/Kg [80], where treatment improved systemic oxidative stress markers through reducing malondialdehyde levels while elevating the activities of essential antioxidant enzymes, superoxide dismutase and catalase, indicating significant oxidative homeostasis amelioration. Furthermore, Moringa oleifera seed oil extract attenuated proinflammatory responses in tumor-bearing mice; collectively, these findings underscore its therapeutic potential to restore redox balance and mitigate the inflammatory reactions within the TME [80]. Nevertheless, caution must be taken regarding the use of Moringa oleifera preparations in combination with standard chemotherapy regimens, as it has been shown that in an obesity-associated triple-negative breast cancer model (MDA-MB-231 xenografts in C57BL/6J), the concomitant oral administration of Moringa oleifera seed extract concentrate (high-fat diet supplemented with 0.6% w/w Moringa concentrate) with conventional chemotherapy drug—comprising doxorubicin (2 mg/kg) and cyclophosphamide (100 mg/kg)—not only negated its angiogenesis-reducing benefit but also aggravated tumor progression, underscoring possible context-specific pro-tumor effects or adverse reactions [163]. These outcomes highlight the need for mechanistic and pharmacokinetic/pharmacodynamic clarity when exploring Moringa oleifera in combination with chemotherapy.

5.4. From Bench to Bedside: Addressing Translation Challenges in Moringa oleifera-Based Cancer Therapy

To advance Moringa oleifera as a reliable adjunct in cancer therapy, critical translational gaps must first be addressed. (i) Standardization and characterization of Moringa oleifera preparations. Defining Good Manufacturing Practice (GMP)-grade materials with rigorously quantified active constituents such as MIC-1, niazimicin, and principal flavonoids, alongside comprehensive impurity profiling and batch-level certificates to ensure validity and reproducibility in both preclinical and clinical investigations. Recent studies have highlighted variability in bioactive compounds across different Moringa oleifera sources, emphasizing the urgent need for standardized preparations [152]. (ii) A thorough understanding of pharmacodynamic, pharmacokinetic and target engagement is required. Determining human-relevant exposure levels of MIC-1 (both free and conjugated forms), evaluating tissue distribution, and confirming interaction with key molecular targets, including PTP1B and Nrf2, are crucial for predicting therapeutic outcomes and clinical efficacy [164,165,166]. Computational and experimental studies indicate that Moringa oleifera phytochemicals may modulate oncogenic and pro-survival proteins, such as Bcl-2 [81,144,167,168,169,170], providing mechanistic avenues that support translational potential. (iii) Context specificity should be rigorously addressed. The effect of Moringa oleifera can vary depending on tumor subtype, metabolic status (e.g., obesity), and concomitant treatments. Preclinical evidence demonstrates that Moringa oleifera modulates important signaling pathways, including YAP/TAZ, Wnt/β-catenin, Nrf2-Keap1 and TLR4/NF-κB, which play divergent roles depending on the tumor cellular context, microenvironment, and cancer-related immune-metabolic axis [152]. Consequently, tailoring interventions to the specific disease context is crucial to minimize adverse and prevent counterproductive outcomes, thereby enhancing therapeutic efficacy and advancing the development of targeted cancer therapies. (iv) Assessment of immuno-oncology endpoints should extend beyond conventional chemotherapeutic regimens. Evaluating Moringa oleifera’s impact on the TME, including Treg/CD8+ balance, NK activity and M1/M2 macrophage polarization in immunocompetent models, with or without checkpoint blockade, is essential to define its immunomodulatory potential. Emerging evidence shows that Moringa oleifera extracts can enhance immune activation, suggesting promising opportunities for combination with immunotherapeutic strategies [81]. (v) Early-phase clinical studies are essential to translate Moringa oleifera findings to humans. Phase 0 or early pharmacokinetic trials, using biomarkers such as NQO1 activation, ERK signaling and cytokine profiles can guide dosing, pharmacodynamic response, and safety before disease-focused randomized controlled trials. Despite limited clinical data on cancer research, pharmacokinetics, antioxidant and anti-inflammatory activities provide a strong rationale for systemic investigation in oncology [20].
Together, addressing these translational priorities, including standardized Moringa oleifera preparations, rigorous pharmacological characterization, context-specific evaluation, and early clinical investigation, will enable the rational incorporation of Moringa oleifera into cancer therapy while ensuring mechanistic understanding, reproducibility, and safety.

6. Clinical Trials on the Immunomodulatory Properties of Moringa oleifera in Disease Prevention and Cancer Therapy

Moringa oleifera has emerged as a promising candidate due to its rich phytochemical composition and demonstrated ability to modulate both innate and adaptive immune responses through interconnected mechanisms. Recent clinical studies have begun to provide evidence supporting its immunostimulatory effects in humans, reflected by improved immune biomarkers, hematological parameters, and disease-specific outcomes [123,124,125,126,127]. These trials provide a translational perspective, bridging preclinical research findings with potential clinical applications for immune enhancement, disease prevention and management. Extending on its systemic immunomodulatory properties, clinical studies have demonstrated that Moringa oleifera improved immune responses in patients with gum disease [171] (CTRI/2022/02/040594), increases CD4 levels and nutritional status in HIV-positive patients [127,172], and improves immunological and hematological parameters in patients undergoing highly active antiretroviral therapy [173]. Moreover, clinical investigations have highlighted the potential of Moringa oleifera as an adjunct in cancer immunotherapy. In particular, an aqueous leaf extract exhibited significant anticancer activities by inducing apoptosis and inhibiting tumor progression without affecting normal physiological functions [174].
Another important study showed that Moringa oleifera leaf polysaccharides possess the ability to modulate the TME through inducing a phenotypic switch from immunosuppressive M2 macrophages to pro-inflammatory M1 phenotypes via TLR4 targeting [58], suggesting their promising role in cancer immunomodulation. In addition, the combination of Moringa oleifera with other phytochemicals, including saffron or green tea, has been demonstrated to enhance both anticancer efficacy and immunomodulatory activity, highlighting the therapeutic potential of synergistic strategies in cancer prevention and therapy [54].
Overall, these clinical findings underscore the translational potential of Moringa oleifera for disease prevention, immune modulation, and as an adjunct in cancer immunotherapy. While clinical trials demonstrate the therapeutic benefits of Moringa oleifera, investigating its long-term safety and toxicity remains critical to support its sustained therapeutic application.

7. Long-Term Safety and Toxicity of Moringa oleifera

The long-term safety and toxicity profile of Moringa oleifera has been extensively investigated in both preclinical and clinical studies, with most evidence supporting its well-established tolerability. A consolidated overview of these findings is presented in Table 3, which summarizes the available evidence across plant parts, experimental models, dosing regimens, and key safety outcomes.
Preclinical studies on animal models have provided insights into the safety profile of Moringa oleifera. For instance, a 13-week repeated-dose study of an optimized aqueous leaf extract revealed no serious toxicity but recommended caution with long-term doses above 500 mg/kg in rodents, establishing a no-observed-adverse-effect level (NOAEL) close to 500 mg/kg [180]. In parallel, a 28-day repeated-dose toxicity study in rats administered orally with aqueous leaf extract exhibited no mortality or organ toxicity at 100–500 mg/kg/day, although a minor reduction in body weight was observed at the highest doses, findings that align with an acute safety margin up to 2000 mg/kg [181,182]. Moreover, disease-model studies, notably in gout, also produced favorable outcomes, further strengthening the evidence for systemic safety [183]. Reproductive and developmental toxicity assessments likewise have shown an absence of adverse effects at nutritionally relevant doses, thereby supporting its long-term safety [184,185,186]. Evidence on Moringa oleifera seeds and roots suggests a more restricted safety compared to leaves. A 14-day oral toxicity evaluation of a hydro-alcoholic seed extract did not report any significant toxicological effects, supporting its short-term tolerability [62,175]. In contrast, seed oil administered at 200–800 mg/kg in murine malaria models not only reduced parasitemia but also induced renal and hepatic lesions at higher doses, indicating a narrower therapeutic window for seed oils compared to Moringa oleifera leaf preparations [187]. Likewise, a 21-day oral administration of ethanol extract (150–600 mg/kg/day) in rats showed no nephrotoxic or hepatotoxicity on standard biochemical parameters, although the authors recommended longer-term evaluations due to the generally higher caution warranted for roots compared with leaves [188]. These findings emphasize the critical role of plant-part specificity, with leaves demonstrating broadly safer, whereas seed and root derivatives may require more cautious dosing and further long-term evaluation. Additional preclinical studies on related species, including Moringa stenopetala herbal tea blends, also support low toxicity in rats [189], though direct extrapolation to Moringa oleifera should be approached cautiously.
Clinical trials further corroborate these findings, indicating that oral supplementation with whole leaf powders or extracts is generally safe, well-tolerated and without consistent adverse effects in humans, while highlighting the need for standardized products and longer follow-up [62,190]. Both randomized controlled and observational studies have consistently reported no significant adverse effects, even at relatively high doses over extended periods. These observations are further supported by systematic reviews indicating that Moringa oleifera is well-tolerated, does not produce significant adverse effects, and serious toxicity [190,191,192,193]. Importantly, Moringa oleifera supplementation did not compromise hematological, renal, or hepatic functions, highlighting its favorable systemic safety profile [194,195,196]. Nevertheless, caution is warranted with Moringa oleifera parts consumption, especially the bark and roots. Overuse of these parts may carry toxicity risk due to the presence of potentially toxic bioactive compounds such as alkaloids [197,198,199,200]. Moreover, pharmacokinetic evidence indicates the possible interaction of Moringa oleifera with drugs metabolized by cytochrome P450 enzymes, including antihyperglycemic drugs, necessitating the need for careful use in patients receiving such therapies [192]. Rare clinical case reports have identified cutaneous reactions and potential renal/hepatic perturbations, as well as possible drug interactions, emphasizing the necessity for cautious dosing and pharmacovigilance, particularly in vulnerable populations [201]. Taken together, although Moringa oleifera demonstrates a generally favorable safety profile in both preclinical and clinical studies, its long-term use should be approached with consideration of potential drug–nutrient interactions, doses, and the specific parts of the plant employed.

8. Conclusions and Future Directions

Moringa oleifera has long been known to exhibit medicinal properties, and recent scientific investigations have shown its efficacy in modulating the immune system. These research findings suggest that Moringa oleifera plays an essential role in maintaining homeostasis between pro-inflammatory and anti-inflammatory cytokine responses and regulating the functions of CD4+ T-cells subsets by targeting transcription factors and their signal transducers. These results highlight its promise in managing T cell-mediated autoimmune disorders and coordinating innate and adaptive immune responses. Its extracts also enhance hematopoietic activity and strengthen both cellular and humoral immunity, suggesting benefits for immunodeficiency disorders, while supporting gut microbial balance to reinforce immune homeostasis and anti-inflammatory signaling. Beyond immune regulation, Moringa oleifera exhibits potent anticancer activity through inducing apoptosis, cell-cycle arrest, activation of antioxidant pathways such as Nrf2, and suppressing oncogenic signaling. Evidence also indicates its capacity to inhibit angiogenesis, metastasis, and modulate immune networks within the tumor microenvironment.
To meet accepted standards, it is important to conduct high-throughput screening, analyses of the bioactive compounds, and identify the chemical targets when studying the immunomodulatory and anti-cancer activities of Moringa oleifera extracts. Those extracts should be subject to qualitative and quantitative analysis through validated analytical procedures to define phytochemical levels in Moringa oleifera extracts.
Several bioactive compounds from Moringa oleifera, such as isothiocyanate, flavonoids and polysaccharides (e.g., MOP-2, MRP-1) have been shown to modulate immune signaling pathways and exhibit immunomodulatory and anti-cancer effects. Nevertheless, it is necessary to conduct further mechanistic investigations with a comprehensive update on the current state of research, through multi-scale approaches spanning cellular and molecular mechanisms, host–microbiome interactions, and systemic physiology, to better understand how these compounds regulate innate, adaptive immune system responses and tumor biology. A deeper understanding of how Moringa oleifera modulates λδ T cells, natural killer T cells, mucosal-associated invariant T cells, and their roles in immune-regulatory circuits within the TME will be critical to advance Moringa oleifera from traditional medicine toward evidence-based immunotherapy and cancer interventions.
Despite the promising therapeutic effects of Moringa oleifera, critical gaps remain regarding its long-term safety, potential drug–nutrient interactions, optimal dosing, and the specific plant parts employed. Accordingly, rigorous standardized preclinical testing, encompassing toxicological, pharmacokinetic, and bioavailability studies, will be essential to ensure both safety and effective translation into clinical use.

Author Contributions

Conceptualization, M.T.; formal analysis, M.T. and J.E.K.; writing—original draft preparation, M.T. and J.E.K.; writing—review and editing, M.T.; visualization and critically reviewed the study design, H.A.M. and A.Z. 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

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CCR7C-C Chemokine Receptor Type 7
EREndoplasmic Reticulum
HSP70Heat Shock Protein 70
iNOSInducible Nitric Oxide Synthase
MOPMoringa oleifera Polysaccharide
Mop3Motif 3 Protein
MRP-1Moringa Root Polysaccharide 1
NLRP3NOD (Nucleotide-binding Oligomerization) LRR (Leucine-Rich Repeat) Pyrin Domain-containing Protein 3
NQO1NAD(P)H: Quinone Oxidoreductase 1
PGE-2Prostaglandin E2
PTP1BProtein Tyrosine Phosphatase 1B
SODSuperoxide Dismutase
TLRToll-Like Receptor
TMETumor Microenvironment

References

  1. Pant, T.; Uche, N.; Juric, M.; Zielonka, J.; Bai, X. Regulation of Immunomodulatory Networks by Nrf2-Activation in Immune Cells: Redox Control and Therapeutic Potential in Inflammatory Diseases. Redox Biol. 2024, 70, 103077. [Google Scholar] [CrossRef]
  2. Strzelec, M.; Detka, J.; Mieszczak, P.; Sobocińska, M.K.; Majka, M. Immunomodulation—A General Review of the Current State-of-the-Art and New Therapeutic Strategies for Targeting the Immune System. Front. Immunol. 2023, 14, 1127704. [Google Scholar] [CrossRef]
  3. Ma, Y.; Kroemer, G. The Cancer-Immune Dialogue in the Context of Stress. Nat. Rev. Immunol. 2024, 24, 264–281. [Google Scholar] [CrossRef]
  4. Carnet Le Provost, K.; Kepp, O.; Kroemer, G.; Bezu, L. Trial Watch: Dexmedetomidine in Cancer Therapy. OncoImmunology 2024, 13, 2327143. [Google Scholar] [CrossRef]
  5. Walczak, J.; Iwaszkiewicz-Grześ, D.; Cholewiński, G. Approaches Towards Better Immunosuppressive Agents. Curr. Top. Med. Chem. 2024, 24, 1230–1263. [Google Scholar] [CrossRef] [PubMed]
  6. La-Beck, N.M.; Owoso, J. Updates and Emerging Trends in the Management of Immune-Related Adverse Events Associated with Immune Checkpoint Inhibitor Therapy. Asia-Pac. J. Oncol. Nurs. 2024, 11, 100549. [Google Scholar] [CrossRef] [PubMed]
  7. Ganguli, S.; Debnath, T. Drugs Used for Immunomodulation. In Essentials of Pharmacodynamics and Drug Action; Chakraverty, R., Mathur, R., Chakraborty, P., Eds.; Springer Nature: Singapore, 2024; pp. 229–240. ISBN 978-981-97-2776-6. [Google Scholar]
  8. Sohail, R.; Mathew, M.; Patel, K.K.; Reddy, S.A.; Haider, Z.; Naria, M.; Habib, A.; Abdin, Z.U.; Razzaq Chaudhry, W.; Akbar, A. Effects of Non-Steroidal Anti-Inflammatory Drugs (NSAIDs) and Gastroprotective NSAIDs on the Gastrointestinal Tract: A Narrative Review. Cureus 2023, 15, e37080. [Google Scholar] [CrossRef] [PubMed]
  9. Kawai, T.; Suzuki, C.; Honda, Y.; Fernandez, J.L. Long-Term Safety and Effectiveness of Vonoprazan for Prevention of Gastric and Duodenal Ulcer Recurrence in Patients on Nonsteroidal Anti-Inflammatory Drugs in Japan: A 12-Month Post-Marketing Surveillance Study. Expert Opin. Drug Saf. 2023, 22, 425–431. [Google Scholar] [CrossRef]
  10. Chakik, R.M.; Alqahtani, N.I.; Al-Hagawi, Y.; Nasser Alsharif, S.; Alqahtani, A.S.; Hadi Asiri, D.; Al-Mani, S.Y. Diagnosis of Small Intestinal Diaphragms and Strictures Induced by Non-Steroidal Anti-Inflammatory Drugs Through Intraoperative Enteroscopy: A Case Study from Saudi Arabia. Cureus 2024, 16, e59752. [Google Scholar] [CrossRef]
  11. Choe, Y.; Park, J.M.; Kim, J.S.; Cho, Y.K.; Kim, B.-W.; Choi, M.-G.; Kim, N.J. Drugs Effective for Nonsteroidal Anti-Inflammatory Drugs or Aspirin-Induced Small Bowel Injuries: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Clin. Gastroenterol. 2024, 58, 1003–1010. [Google Scholar] [CrossRef]
  12. Berge, L.A.M.; Andreassen, B.K.; Stenehjem, J.S.; Heir, T.; Karlstad, Ø.; Juzeniene, A.; Ghiasvand, R.; Larsen, I.K.; Green, A.C.; Veierød, M.B.; et al. Use of Immunomodulating Drugs and Risk of Cutaneous Melanoma: A Nationwide Nested Case-Control Study. Clin. Epidemiol. 2020, 12, 1389–1401. [Google Scholar] [CrossRef] [PubMed]
  13. Rahman, A.; Haider, M.F. A Comprehensive Review on Glucocorticoids Induced Osteoporosis: A Medication Caused Disease. Steroids 2024, 207, 109440. [Google Scholar] [CrossRef]
  14. Tsubosaka, M.; Maruyama, M.; Lui, E.; Kushioka, J.; Toya, M.; Gao, Q.; Shen, H.; Li, X.; Chow, S.K.-H.; Zhang, N.; et al. Preclinical Models for Studying Corticosteroid-Induced Osteonecrosis of the Femoral Head. J. Biomed. Mater. Res. Part B Appl. Biomater. 2024, 112, e35360. [Google Scholar] [CrossRef]
  15. Hsu, C.H.; Hsu, C.L.; Langley, A.; Wojcik, C.; Iraganje, E.; Grygiel-Górniak, B. Glucocorticoid-Induced Osteoporosis—From Molecular Mechanism to Clinical Practice. Drugs Ther. Perspect. 2024, 40, 315–329. [Google Scholar] [CrossRef]
  16. Elgert, K.D. Immunology: Understanding the Immune System, 2nd ed.; Wiley-Blackwell: Hoboken, NJ, USA, 2009; ISBN 978-0-470-08157-0. [Google Scholar]
  17. Kamran, M.; Hussain, S.; Abid, M.A.; Syed, S.K.; Suleman, M.; Riaz, M.; Iqbal, M.; Mahmood, S.; Saba, I.; Qadir, R. Phytochemical Composition of Moringa oleifera Its Nutritional and Pharmacological Importance. Postep. Biol. Komorki 2020, 47, 321–334. [Google Scholar]
  18. Singh, A.K.; Rana, H.K.; Tshabalala, T.; Kumar, R.; Gupta, A.; Ndhlala, A.R.; Pandey, A.K. Phytochemical, Nutraceutical and Pharmacological Attributes of a Functional Crop Moringa oleifera Lam: An Overview. S. Afr. J. Bot. 2020, 129, 209–220. [Google Scholar] [CrossRef]
  19. Camilleri, E.; Blundell, R. A Comprehensive Review of the Phytochemicals, Health Benefits, Pharmacological Safety and Medicinal Prospects of Moringa oleifera. Heliyon 2024, 10, e27807. [Google Scholar] [CrossRef]
  20. Pareek, A.; Pant, M.; Gupta, M.M.; Kashania, P.; Ratan, Y.; Jain, V.; Pareek, A.; Chuturgoon, A.A. Moringa oleifera: An Updated Comprehensive Review of Its Pharmacological Activities, Ethnomedicinal, Phytopharmaceutical Formulation, Clinical, Phytochemical, and Toxicological Aspects. Int. J. Mol. Sci. 2023, 24, 2098. [Google Scholar] [CrossRef]
  21. Owon, M.; Osman, M.; Ibrahim, A.; Salama, M.A.; Matthäus, B. Characterisation of Different Parts from Moringa oleifera Regarding Protein, Lipid Composition and Extractable Phenolic Compounds. OCL 2021, 28, 45. [Google Scholar] [CrossRef]
  22. Leone, A.; Spada, A.; Battezzati, A.; Schiraldi, A.; Aristil, J.; Bertoli, S. Moringa oleifera Seeds and Oil: Characteristics and Uses for Human Health. Int. J. Mol. Sci. 2016, 17, 2141. [Google Scholar] [CrossRef] [PubMed]
  23. Takase, M.; Essandoh, P.K.; Asare, R.K.; Nazir, K.-H. The Value Chain of Moringa oleifera Plant and the Process of Producing Its Biodiesel in Ghana. Sci. World J. 2022, 2022, 1827514. [Google Scholar] [CrossRef]
  24. Anwar, F.; Ashraf, M.; Bhanger, M.I. Interprovenance Variation in the Composition of Moringa oleifera Oilseeds from Pakistan. J. Am. Oil Chem. Soc. 2005, 82, 45–51. [Google Scholar] [CrossRef]
  25. Corbett, P. It’s Time for an Oil Change! Opportunities for High-Oleic Vegetable Oils. Inf.-Int. News Fats Oils Relat. Mater. 2003, 14, 480–481. [Google Scholar]
  26. Gharsallah, K.; Rezig, L.; Msaada, K.; Chalh, A.; Soltani, T. Chemical Composition and Profile Characterization of Moringa oleifera Seed Oil. S. Afr. J. Bot. 2021, 137, 475–482. [Google Scholar] [CrossRef]
  27. Kaur, G.; Invally, M.; Sanzagiri, R.; Buttar, H.S. Evaluation of the Antidepressant Activity of Moringa oleifera Alone and in Combination with Fluoxetine. J. Ayurveda Integr. Med. 2015, 6, 273. [Google Scholar] [CrossRef] [PubMed]
  28. Masitlha, E.P.; Seifu, E.; Teketay, D. Nutritional Composition and Mineral Profile of Leaves of Moringa oleifera Provenances Grown in Gaborone, Botswana. Food Prod. Process Nutr. 2024, 6, 3. [Google Scholar] [CrossRef]
  29. Marrufo, T.; Nazzaro, F.; Mancini, E.; Fratianni, F.; Coppola, R.; De Martino, L.; Agostinho, A.B.; De Feo, V. Chemical Composition and Biological Activity of the Essential Oil from Leaves of Moringa oleifera Lam. Cultivated in Mozambique. Molecules 2013, 18, 10989–11000. [Google Scholar] [CrossRef]
  30. Divya, S.; Pandey, V.K.; Dixit, R.; Rustagi, S.; Suthar, T.; Atuahene, D.; Nagy, V.; Ungai, D.; Ahmed, A.E.M.; Kovács, B.; et al. Exploring the Phytochemical, Pharmacological and Nutritional Properties of Moringa oleifera: A Comprehensive Review. Nutrients 2024, 16, 3423. [Google Scholar] [CrossRef]
  31. Bhattacharya, S.B.; Das, A.K.; Banerji, N. Chemical Investigations on the Gum Exudate from Sajna (Moringa oleifera). Carbohydr. Res. 1982, 102, 253–262. [Google Scholar] [CrossRef]
  32. Faizi, S.; Siddiqui, B.S.; Saleem, R.; Siddiqui, S.; Aftab, K.; Gilani, A.H. Isolation and Structure Elucidation of New Nitrile and Mustard Oil Glycosides from Moringa oleifera and Their Effect on Blood Pressure. J. Nat. Prod. 1994, 57, 1256–1261. [Google Scholar] [CrossRef]
  33. Waterman, C.; Rojas-Silva, P.; Tumer, T.B.; Kuhn, P.; Richard, A.J.; Wicks, S.; Stephens, J.M.; Wang, Z.; Mynatt, R.; Cefalu, W.; et al. Isothiocyanate-rich Moringa oleifera Extract Reduces Weight Gain, Insulin Resistance, and Hepatic Gluconeogenesis in Mice. Mol. Nutr. Food Res. 2015, 59, 1013–1024. [Google Scholar] [CrossRef]
  34. Xie, J.; Qian, Y.-Y.; Yang, Y.; Peng, L.-J.; Mao, J.-Y.; Yang, M.-R.; Tian, Y.; Sheng, J. Isothiocyanate from Moringa oleifera Seeds Inhibits the Growth and Migration of Renal Cancer Cells by Regulating the PTP1B-Dependent Src/Ras/Raf/ERK Signaling Pathway. Front. Cell Dev. Biol. 2022, 9, 790618. [Google Scholar] [CrossRef]
  35. Cuellar-Núñez, M.L.; Loarca-Piña, G.; Berhow, M.; Gonzalez de Mejia, E. Glucosinolate-Rich Hydrolyzed Extract from Moringa oleifera Leaves Decreased the Production of TNF-α and IL-1β Cytokines and Induced ROS and Apoptosis in Human Colon Cancer Cells. J. Funct. Foods 2020, 75, 104270. [Google Scholar] [CrossRef]
  36. Chodur, G.M.; Olson, M.E.; Wade, K.L.; Stephenson, K.K.; Nouman, W.; Garima; Fahey, J.W. Wild and Domesticated Moringa oleifera Differ in Taste, Glucosinolate Composition, and Antioxidant Potential, but Not Myrosinase Activity or Protein Content. Sci. Rep. 2018, 8, 7995. [Google Scholar] [CrossRef] [PubMed]
  37. Zarina; Wani, A.W.; Rawat, M.; Kaur, H.; Das, S.; Kaur, T.; Akram, N.; Faisal, Z.; Jan, S.S.; Oyshe, N.N.; et al. Medicinal Utilization and Nutritional Properties of Drumstick (Moringa oleifera)—A Comprehensive Review. Food Sci. Nutr. 2024, 12, 4546–4568. [Google Scholar] [CrossRef]
  38. Engsuwan, J.; Waranuch, N.; Limpeanchob, N.; Ingkaninan, K. HPLC Methods for Quality Control of Moringa oleifera Extract Using Isothiocyanates and Astragalin as Bioactive Markers. ScienceAsia 2017, 43, 169–174. [Google Scholar] [CrossRef]
  39. Riaz, A.; Rasul, A.; Hussain, G.; Zahoor, M.K.; Jabeen, F.; Subhani, Z.; Younis, T.; Ali, M.; Sarfraz, I.; Selamoglu, Z. Astragalin: A Bioactive Phytochemical with Potential Therapeutic Activities. Adv. Pharmacol. Sci. 2018, 2018, 9794625. [Google Scholar] [CrossRef]
  40. Leone, A.; Fiorillo, G.; Criscuoli, F.; Ravasenghi, S.; Santagostini, L.; Fico, G.; Spadafranca, A.; Battezzati, A.; Schiraldi, A.; Pozzi, F. Nutritional Characterization and Phenolic Profiling of Moringa oleifera Leaves Grown in Chad, Sahrawi Refugee Camps, and Haiti. Int. J. Mol. Sci. 2015, 16, 18923–18937. [Google Scholar] [CrossRef] [PubMed]
  41. Zduńska, K.; Dana, A.; Kolodziejczak, A.; Rotsztejn, H. Antioxidant Properties of Ferulic Acid and Its Possible Application. Ski. Pharmacol. Physiol. 2018, 31, 332–336. [Google Scholar] [CrossRef]
  42. Klimek-Szczykutowicz, M.; Gaweł-Bęben, K.; Rutka, A.; Blicharska, E.; Tatarczak-Michalewska, M.; Kulik-Siarek, K.; Kukula-Koch, W.; Malinowska, M.A.; Szopa, A. Moringa oleifera (Drumstick Tree)—Nutraceutical, Cosmetological and Medicinal Importance: A Review. Front. Pharmacol. 2024, 15, 1288382. [Google Scholar] [CrossRef]
  43. Nikkon, F.; Hasan, S.; Salam, K.A.; Mosaddik, M.A.; Khondkar, P.; Haque, M.E.; Rahman, M. Benzylcarbamothioethionate from Root Bark of Moringa oleifera Lam. and Its Toxicological Evaluation. Boletín Latinoam. Caribe Plantas Med. Aromát. 2009, 8, 130–138. [Google Scholar]
  44. Alhakmani, F.; Kumar, S.; Khan, S.A. Estimation of Total Phenolic Content, in–Vitro Antioxidant and Anti–Inflammatory Activity of Flowers of Moringa oleifera. Asian Pac. J. Trop. Biomed. 2013, 3, 623–627. [Google Scholar] [CrossRef] [PubMed]
  45. Atawodi, S.E.; Atawodi, J.C.; Idakwo, G.A.; Pfundstein, B.; Haubner, R.; Wurtele, G.; Bartsch, H.; Owen, R.W. Evaluation of the Polyphenol Content and Antioxidant Properties of Methanol Extracts of the Leaves, Stem, and Root Barks of Moringa oleifera Lam. J. Med. Food 2010, 13, 710–716. [Google Scholar] [CrossRef] [PubMed]
  46. Calva, J.; Cuenca, M.B.; León, A.; Benítez, Á. Chemical Composition, Acetylcholinesterase-Inhibitory Potential and Antioxidant Activity of Essential Oils from Three Populations of Parthenium Hysterophorus L. in Ecuador. Molecules 2025, 30, 2712. [Google Scholar] [CrossRef]
  47. Malaspina, P.; Polito, F.; Mainetti, A.; Khedhri, S.; De Feo, V.; Cornara, L. Exploring Chemical Variability in the Essential Oil of Artemisia Absinthium L. in Relation to Different Phenological Stages and Geographical Location. Chem. Biodivers. 2025, 20, e00743. [Google Scholar] [CrossRef]
  48. Hridoy, A.A.M.; Munny, F.J.; Shahriar, F.; Rahman, M.; Islam, F.; Kazmi, A.; Kawsar, A.; Bailey, C. Exploring the Potentials of Sajana (Moringa oleifera Lam.) as a Plant-Based Feed Ingredient to Sustainable and Good Aquaculture Practices: An Analysis of Growth Performance and Health Benefits. Aquac. Res. 2025, 2025, 3580123. [Google Scholar] [CrossRef]
  49. Tilaoui, M.; Mouse, H.A.; Jaafari, A.; Zyad, A. Comparative Phytochemical Analysis of Essential Oils from Different Biological Parts of Artemisia Herba Alba and Their Cytotoxic Effect on Cancer Cells. PLoS ONE 2015, 10, e0131799. [Google Scholar] [CrossRef]
  50. Balasubramaniam, M.; Sapuan, S.; Hashim, I.F.; Ismail, N.I.; Yaakop, A.S.; Kamaruzaman, N.A.; Mokhtar, A.M.A. The Properties and Mechanism of Action of Plant Immunomodulators in Regulation of Immune Response—A Narrative Review Focusing on Curcuma longa L., Panax Ginseng C. A. Meyer and Moringa oleifera Lam. Heliyon 2024, 10, e28261. [Google Scholar] [CrossRef] [PubMed]
  51. Goel, F. Exploring the Therapeutic Role of Moringa oleifera in Neurodegeneration: Antioxidant, Anti-Inflammatory, and Neuroprotective Mechanisms. Inflammopharmacology 2025, 33, 3653–3669. [Google Scholar] [CrossRef]
  52. Gaber, H.G.; Younis, N.A.; Saleh, S.Y. Comparative Physiological and Immunological Impacts of Moringa oleifera Leaf and Seed Water Supplements on African Catfish (Clarias gariepinus): Effects on Disease Resistance and Health Parameters. BMC Vet. Res. 2025, 21, 320. [Google Scholar] [CrossRef]
  53. Kamble, M.T.; Gallardo, W.; Salin, K.R.; Pumpuang, S.; Chavan, B.R.; Bhujel, R.C.; Medhe, S.V.; Kettawan, A.; Thompson, K.D.; Pirarat, N. Effect of Moringa oleifera Leaf Extract on the Growth Performance, Hematology, Innate Immunity, and Disease Resistance of Nile Tilapia (Oreochromis niloticus) against Streptococcus Agalactiae Biotype 2. Animals 2024, 14, 953. [Google Scholar] [CrossRef]
  54. Talib, W.H.; Al Junaidi, H.S.; Alshaeri, H.K.; Alasmari, M.M.; Hadi, R.W.; Alsayed, A.R.; Law, D. Immunomodulatory and Anticancer Effects of Moringa Polyherbal Infusions: Potentials for Preventive and Therapeutic Use. Front. Immunol. 2025, 16, 1597602. [Google Scholar] [CrossRef] [PubMed]
  55. Rao, Q.; Hua, H.; Zhao, J. Advancements in Plant-Derived sRNAs Therapeutics: Classification, Delivery Strategies, and Therapeutic Applications. Int. J. Mol. Sci. 2025, 26, 4277. [Google Scholar] [CrossRef]
  56. Mohai Ud Din, R.; Eman, S.; Zafar, M.H.; Chong, Z.; Saleh, A.A.; Husien, H.M.; Wang, M. Moringa oleifera as a Multifunctional Feed Additive: Synergistic Nutritional and Immunomodulatory Mechanisms in Livestock Production. Front. Nutr. 2025, 12, 1615349. [Google Scholar] [CrossRef]
  57. Abdelazim, A.M.; Afifi, M.; Abu-Alghayth, M.H.; Alkadri, D.H. Moringa oleifera: Recent Insights for Its Biochemical and Medicinal Applications. J. Food Biochem. 2024, 2024, 1270903. [Google Scholar] [CrossRef]
  58. Wang, S.; Hu, Q.; Chang, Z.; Liu, Y.; Gao, Y.; Luo, X.; Zhou, L.; Chen, Y.; Cui, Y.; Wang, Z. Moringa oleifera Leaf Polysaccharides Exert Anti-Lung Cancer Effects upon Targeting TLR4 to Reverse the Tumor-Associated Macrophage Phenotype and Promote T-Cell Infiltration. Food Funct. 2023, 14, 4607–4620. [Google Scholar] [CrossRef]
  59. Valentina, A.S.; Nugroho, T.; Susilaningsih, N. The Effect of Ethanolic Extract of Moringa oleifera Leaves on the Macrophage Count and VEGF Expression on Wistar Rats with Burn Wound. Biosci. Med. J. Biomed. Transl. Res. 2022, 6, 1959–1964. [Google Scholar] [CrossRef]
  60. Xi, C.; Li, W.; Liu, X.; Xie, J.; Li, S.; Tian, Y.; Song, S. The Potential Role of Moringa oleifera Lam. Leaf Proteins in Moringa Allergy by Functionally Activating Murine Bone Marrow-Derived Dendritic Cells and Inducing Their Differentiation toward a Th2-Polarizing Phenotype. Nutrients 2023, 16, 7. [Google Scholar] [CrossRef] [PubMed]
  61. Pilotos, J.; Ibrahim, K.A.; Mowa, C.N.; Opata, M.M. Moringa oleifera Treatment Increases Tbet Expression in CD4+ T Cells and Remediates Immune Defects of Malnutrition in Plasmodium Chabaudi-Infected Mice. Malar. J. 2020, 19, 62. [Google Scholar] [CrossRef]
  62. Azlan, U.K.; Mediani, A.; Rohani, E.R.; Tong, X.; Han, R.; Misnan, N.M.; Jam, F.A.; Bunawan, H.; Sarian, M.N.; Hamezah, H.S. A Comprehensive Review with Updated Future Perspectives on the Ethnomedicinal and Pharmacological Aspects of Moringa oleifera. Molecules 2022, 27, 5765. [Google Scholar] [CrossRef]
  63. Tran, T.H.M.; Lee, S.; Huh, J.-E.; Perumalsamy, H.; Balusamy, S.R.; Kim, Y.-J. Fermentation of Moringa oleifera Lam. Using Bifidobacterium Animalis Subsp. Lactis Enhances the Anti-Inflammatory Effect in RAW 264.7 Macrophages. J. Funct. Foods 2023, 109, 105752. [Google Scholar] [CrossRef]
  64. Abdel-Latif, M.; Sakran, T.; Badawi, Y.K.; Abdel-Hady, D.S. Influence of Moringa oleifera Extract, Vitamin C, and Sodium Bicarbonate on Heat Stress-Induced HSP70 Expression and Cellular Immune Response in Rabbits. Cell Stress Chaperones 2018, 23, 975–984. [Google Scholar] [CrossRef]
  65. Potestà, M.; Minutolo, A.; Gismondi, A.; Canuti, L.; Kenzo, M.; Roglia, V.; Macchi, F.; Grelli, S.; Canini, A.; Colizzi, V.; et al. Cytotoxic and Apoptotic Effects of Different Extracts of Moringa oleifera Lam on Lymphoid and Monocytoid Cells. Exp. Ther. Med. 2019, 18, 5–17. [Google Scholar] [CrossRef]
  66. Budhy, T.I.; Adam, D.; Azis, Z.M.R.; Syahputri, V.; Yuliani, M.G.A.; Suwarto, M.F.S.; Setiawan, F. The Potential of Moringa Leaf Nanoparticles (Moringa oleifera) on the Expression of TNFα, IL10, and HSP 27 in Oral Cavity Cancer. J. Multidiscip. Appl. Nat. Sci. 2024, 4, 120–129. [Google Scholar] [CrossRef]
  67. Choi, E.-J.; Debnath, T.; Tang, Y.; Ryu, Y.-B.; Moon, S.-H.; Kim, E.-K. Topical Application of Moringa oleifera Leaf Extract Ameliorates Experimentally Induced Atopic Dermatitis by the Regulation of Th1/Th2/Th17 Balance. Biomed. Pharmacother. 2016, 84, 870–877. [Google Scholar] [CrossRef] [PubMed]
  68. Omodanisi, E.I.; Aboua, Y.G.; Chegou, N.N.; Oguntibeju, O.O. Hepatoprotective, Antihyperlipidemic, and Anti-Inflammatory Activity of Moringa oleifera in Diabetic-Induced Damage in Male Wistar Rats. Pharmacogn. Res. 2017, 9, 182. [Google Scholar]
  69. Drue, G.; Minor, R.C. Moringa oleifera Tea Alters Neutrophil but Not Lymphocyte Levels in Blood of Acutely Stressed Mice. Madridge J. Immunol. 2018, 2, 43–48. [Google Scholar] [CrossRef]
  70. Husni, E.; Badriyya, E.; Putri, L.; Aldi, Y. The Effect of Ethanol Extract of Moringa Leaf (Moringa oleifera Lam) Against the Activity and Capacity of Phagocytosis of Macrofag Cells and the Percentage of Leukosit Cells of White Mice. Pharmacogn. J. 2021, 13, 706–712. [Google Scholar] [CrossRef]
  71. Li, C.; Dong, Z.; Zhang, B.; Huang, Q.; Liu, G.; Fu, X. Structural Characterization and Immune Enhancement Activity of a Novel Polysaccharide from Moringa oleifera Leaves. Carbohydr. Polym. 2020, 234, 115897. [Google Scholar] [CrossRef]
  72. Muangnoi, C.; Chingsuwanrote, P.; Praengamthanachoti, P.; Svasti, S.; Tuntipopipat, S. Moringa oleifera Pod Inhibits Inflammatory Mediator Production by Lipopolysaccharide-Stimulated RAW 264.7 Murine Macrophage Cell Lines. Inflammation 2012, 35, 445–455. [Google Scholar] [CrossRef]
  73. Rachmawati, I.; Rifa’i, M. In Vitro Immunomodulatory Activity of Aqueous Extract of Moringa oleifera Lam. Leaf to the CD4+, CD8+ and B220+ Cells in Mus Musculus. J. Exp. Life Sci. 2014, 4, 15–20. [Google Scholar] [CrossRef]
  74. Hong, Z.-S.; Xie, J.; Wang, X.-F.; Dai, J.-J.; Mao, J.-Y.; Bai, Y.-Y.; Sheng, J.; Tian, Y. Moringa oleifera Lam. Peptide Remodels Intestinal Mucosal Barrier by Inhibiting JAK-STAT Activation and Modulating Gut Microbiota in Colitis. Front. Immunol. 2022, 13, 924178. [Google Scholar] [CrossRef]
  75. Cui, C.; Chen, S.; Wang, X.; Yuan, G.; Jiang, F.; Chen, X.; Wang, L. Characterization of Moringa oleifera Roots Polysaccharide MRP-1 with Anti-Inflammatory Effect. Int. J. Biol. Macromol. 2019, 132, 844–851. [Google Scholar] [CrossRef]
  76. Hunthayung, K.; Bhawamai, S. Polyphenol Compounds of Freeze-Dried Moringa oleifera Lam Pods and Their Anti-Inflammatory Effects on RAW 264.7 Macrophages Stimulated with Lipopolysaccharide. Bioact. Compd. Health Dis. 2024, 7, 185–198. [Google Scholar]
  77. Arulselvan, P.; Tan, W.S.; Gothai, S.; Muniandy, K.; Fakurazi, S.; Esa, N.M.; Alarfaj, A.A.; Kumar, S.S. Anti-Inflammatory Potential of Ethyl Acetate Fraction of Moringa oleifera in Downregulating the NF-κB Signaling Pathway in Lipopolysaccharide-Stimulated Macrophages. Molecules 2016, 21, 1452. [Google Scholar] [CrossRef]
  78. Praengam, K.; Muangnoi, C.; Dawilai, S.; Awatchanawong, M.; Tuntipopipat, S. Digested Moringa oleifera Boiled Pod Exhibits Anti-Inflammatory Activity in Caco-2 Cells. J. Herbs Spices Med. Plants 2015, 21, 148–160. [Google Scholar] [CrossRef]
  79. Luetragoon, T.; Pankla Sranujit, R.; Noysang, C.; Thongsri, Y.; Potup, P.; Suphrom, N.; Nuengchamnong, N.; Usuwanthim, K. Bioactive Compounds in Moringa oleifera Lam. Leaves Inhibit the Pro-Inflammatory Mediators in Lipopolysaccharide-Induced Human Monocyte-Derived Macrophages. Molecules 2020, 25, 191. [Google Scholar] [CrossRef]
  80. Aldayel, T.S.; Gad El Hak, H.N.; Nafie, M.S.; Saad, R.; Abdelrazek, H.M.A.; Kilany, O.E. Evaluation of Antioxidant, Anti-Inflammatory, Anticancer Activities and Molecular Docking of Moringa oleifera Seed Oil Extract against Experimental Model of Ehrlich Ascites Carcinoma in Swiss Female Albino Mice. BMC Complement. Med. Ther. 2023, 23, 457. [Google Scholar] [CrossRef] [PubMed]
  81. Liu, X.; Yang, H.; Xie, S.; Wang, X.; Tian, Y.; Song, S. Moringa oleifera Leaves Protein Suppresses T-Lymphoblastic Leukemogenesis via MAPK/AKT Signaling Modulation of Apoptotic Activation and Autophagic Flux Regulation. Front. Immunol. 2025, 16, 1546189. [Google Scholar] [CrossRef]
  82. Ababneh, S.K.; Khwaldeh, A.; Alsarhan, A.A.; Yousef, I.; Al-Shdefat, R.; Shoiab, A.; Ababneh, S. The Modulatory Effects of Statins, Vitamin E, and Moringa oleifera Extract on CD3 Expression in Ribociclib-Induced Hepatotoxicity in Rats. Acta Inf. Med. 2025, 33, 90–95. [Google Scholar] [CrossRef]
  83. Poluan, J.C.; Zubair, M.S.; Ramadani, A.P.; Hayati, F. Narrative Review: Potential of Flavonoids from Moringa (Moringa oleifera Lam.) Leaves as Immunomodulators. J. Farm. Galen. Galen. J. Pharm. 2023, 9, 270–283. [Google Scholar] [CrossRef]
  84. Swaroop, A.K.; Lalitha, C.M.V.N.; Shanmugam, M.; Subramanian, G.; Natarajan, J.; Selvaraj, J. Plant Derived Immunomodulators; A Critical Review. Adv. Pharm. Bull. 2022, 12, 712. [Google Scholar] [CrossRef]
  85. Nfambi, J.; Bbosa, G.S.; Sembajwe, L.F.; Gakunga, J.; Kasolo, J.N. Immunomodulatory Activity of Methanolic Leaf Extract of Moringa oleifera in Wistar Albino Rats. J. Basic Clin. Physiol. Pharmacol. 2015, 26, 603–611. [Google Scholar] [CrossRef]
  86. Al-Asmari, A.K.; Albalawi, S.M.; Athar, M.T.; Khan, A.Q.; Al-Shahrani, H.; Islam, M. Moringa oleifera as an Anti-Cancer Agent against Breast and Colorectal Cancer Cell Lines. PLoS ONE 2015, 10, e0135814. [Google Scholar] [CrossRef] [PubMed]
  87. Ma, N.; Tang, Q.; Wu, W.-T.; Huang, X.-A.; Xu, Q.; Rong, G.-L.; Chen, S.; Song, J.-P. Three Constituents of Moringa oleifera Seeds Regulate Expression of Th17-Relevant Cytokines and Ameliorate TPA-Induced Psoriasis-Like Skin Lesions in Mice. Molecules 2018, 23, 3256. [Google Scholar] [CrossRef]
  88. Zhang, J.; Liu, X.; Wang, Z.; Zhang, H.; Gao, J.; Wu, Y.; Meng, X.; Zhong, Y.; Chen, H. Potential Allergenicity Response to Moringa oleifera Leaf Proteins in BALB/c Mice. Nutrients 2022, 14, 4700. [Google Scholar] [CrossRef] [PubMed]
  89. Wuryandari, M.R.E.; Atho’illah, M.F.; Laili, R.D.; Fatmawati, S.; Widodo, N.; Widjajanto, E.; Rifa’i, M. Lactobacillus Plantarum FNCC 0137 Fermented Red Moringa oleifera Exhibits Protective Effects in Mice Challenged with Salmonella Typhi via TLR3/TLR4 Inhibition and down-Regulation of Proinflammatory Cytokines. J. Ayurveda Integr. Med. 2022, 13, 100531. [Google Scholar] [CrossRef]
  90. Eladia, R.E.; Ampode, K.M.B. Moringa (Moringa oleifera Lam.) Pod Meal: Nutrient Analysis and Its Effect on the Growth Performance and Cell-Mediated Immunity of Broiler Chickens. J. Anim. Health Prod. 2021, 9, 170–177. [Google Scholar]
  91. Yasoob, T.B.; Khalid, A.R.; Zhang, Z.; Zhu, X.; Hang, S. Liver Transcriptome of Rabbits Supplemented with Oral Moringa oleifera Leaf Powder under Heat Stress Is Associated with Modulation of Lipid Metabolism and Up-Regulation of Genes for Thermo-Tolerance, Antioxidation, and Immunity. Nutr. Res. 2022, 99, 25–39. [Google Scholar] [CrossRef] [PubMed]
  92. Minutolo, A.; Potestà, M.; Roglia, V.; Cirilli, M.; Iacovelli, F.; Cerva, C.; Fokam, J.; Desideri, A.; Andreoni, M.; Grelli, S. Plant microRNAs from Moringa oleifera Regulate Immune Response and HIV Infection. Front. Pharmacol. 2021, 11, 620038. [Google Scholar] [CrossRef]
  93. Aldi, Y.; Ramadhan, Z.L.; Dillasamola, D.; Alianta, A.A. The Activities of Moringa oleifera Lam Leaf Extract on Specific Cellular Immune of Albino Male Mice Induced by the COVID-19 Antigen. Trop. J. Nat. Prod. Res. 2025, 9, 2421–2425. [Google Scholar] [CrossRef]
  94. Hazarika, S.; Lal, M.; Rahaman, S.B.; Debnath, U.; Rawal, R.K. Computational Screening of Moringa oleifera Compounds for Immunomodulatory Targets: Identification and Validation of Core Genes. S. Afr. J. Bot. 2025, 185, 331–347. [Google Scholar] [CrossRef]
  95. Lu, J.; Liu, X.; Li, W.; Xi, C.; Feng, D.; Song, S. Analysis of the Sensitization Activity of Moringa oleifera Leaves Protein. Front. Nutr. 2025, 11, 1509343. [Google Scholar] [CrossRef]
  96. Dong, Z.; Li, C.; Huang, Q.; Zhang, B.; Fu, X.; Liu, R.H. Characterization of a Novel Polysaccharide from the Leaves of Moringa oleifera and Its Immunostimulatory Activity. J. Funct. Foods 2018, 49, 391–400. [Google Scholar] [CrossRef]
  97. Zheng, L.; Lu, X.; Yang, S.; Zou, Y.; Zeng, F.; Xiong, S.; Cao, Y.; Zhou, W. The Anti-Inflammatory Activity of GABA-Enriched Moringa oleifera Leaves Produced by Fermentation with Lactobacillus Plantarum LK-1. Front. Nutr. 2023, 10, 1093036. [Google Scholar] [CrossRef] [PubMed]
  98. Li, L.; Ma, L.; Wen, Y.; Xie, J.; Yan, L.; Ji, A.; Zeng, Y.; Tian, Y.; Sheng, J. Crude Polysaccharide Extracted from Moringa oleifera Leaves Prevents Obesity in Association with Modulating Gut Microbiota in High-Fat Diet-Fed Mice. Front. Nutr. 2022, 9, 861588. [Google Scholar] [CrossRef]
  99. Husien, H.M.; Rehman, S.U.; Duan, Z.; Wang, M. Effect of Moringa oleifera Leaf Polysaccharide on the Composition of Intestinal Microbiota in Mice with Dextran Sulfate Sodium-Induced Ulcerative Colitis. Front. Nutr. 2024, 11, 1409026. [Google Scholar] [CrossRef] [PubMed]
  100. Jia, L.; Peng, X.; Deng, Z.; Zhang, B.; Li, H. The Structural Characterization of Polysaccharides from Three Cultivars of Moringa oleifera Lam. Root and Their Effects on Human Intestinal Microflora. Food Biosci. 2023, 52, 102482. [Google Scholar] [CrossRef]
  101. Wen, Z.; Tian, H.; Liang, Y.; Guo, Y.; Deng, M.; Liu, G.; Li, Y.; Liu, D.; Sun, B. Moringa oleifera Polysaccharide Regulates Colonic Microbiota and Immune Repertoire in C57BL/6 Mice. Int. J. Biol. Macromol. 2022, 198, 135–146. [Google Scholar] [CrossRef]
  102. Mohamed Husien, H.; Peng, W.; Su, H.; Zhou, R.; Tao, Y.; Huang, J.; Liu, M.; Bo, R.; Li, J. Moringa oleifera Leaf Polysaccharide Alleviates Experimental Colitis by Inhibiting Inflammation and Maintaining Intestinal Barrier. Front. Nutr. 2022, 9, 1055791. [Google Scholar] [CrossRef]
  103. Khalid, A.R.; Yasoob, T.B.; Zhang, Z.; Zhu, X.; Hang, S. Dietary Moringa oleifera Leaf Powder Improves Jejunal Permeability and Digestive Function by Modulating the Microbiota Composition and Mucosal Immunity in Heat Stressed Rabbits. Env. Sci. Pollut. Res. 2022, 29, 80952–80967. [Google Scholar] [CrossRef] [PubMed]
  104. Tian, H.; Liang, Y.; Liu, G.; Li, Y.; Deng, M.; Liu, D.; Guo, Y.; Sun, B. Moringa oleifera Polysaccharides Regulates Caecal Microbiota and Small Intestinal Metabolic Profile in C57BL/6 Mice. Int. J. Biol. Macromol. 2021, 182, 595–611. [Google Scholar] [CrossRef]
  105. Li, C.; Zhou, S.; Fu, X.; Huang, Q.; Chen, Q. In Vitro Digestibility and Prebiotic Activities of a Bioactive Polysaccharide from Moringa oleifera Leaves. J. Food Biochem. 2021, 45, e13944. [Google Scholar] [CrossRef]
  106. Zhao, C.; Li, H.; Gao, C.; Tian, H.; Guo, Y.; Liu, G.; Li, Y.; Liu, D.; Sun, B. Moringa oleifera Leaf Polysaccharide Regulates Fecal Microbiota and Colonic Transcriptome in Calves. Int. J. Biol. Macromol. 2023, 253, 127108. [Google Scholar] [CrossRef]
  107. Husien, H.M.; Peng, W.; Essa, M.O.A.; Adam, S.Y.; Ur Rehman, S.; Ali, R.; Saleh, A.A.; Wang, M.; Li, J. The Anti-Inflammatory Properties of Polysaccharides Extracted from Moringa oleifera Leaves on IEC6 Cells Stimulated with Lipopolysaccharide In Vitro. Animals 2024, 14, 3508. [Google Scholar] [CrossRef]
  108. Tian, H.; Wen, Z.; Liu, Z.; Guo, Y.; Liu, G.; Sun, B. Comprehensive Analysis of Microbiome, Metabolome and Transcriptome Revealed the Mechanisms of Moringa oleifera Polysaccharide on Preventing Ulcerative Colitis. Int. J. Biol. Macromol. 2022, 222, 573–586. [Google Scholar] [CrossRef]
  109. Chiricosta, L.; Silvestro, S.; Pizzicannella, J.; Diomede, F.; Bramanti, P.; Trubiani, O.; Mazzon, E. Transcriptomic Analysis of Stem Cells Treated with Moringin or Cannabidiol: Analogies and Differences in Inflammation Pathways. Int. J. Mol. Sci. 2019, 20, 6039. [Google Scholar] [CrossRef]
  110. Wu, Y.-Y.; Xu, Y.-M.; Lau, A.T.Y. Anti-Cancer and Medicinal Potentials of Moringa Isothiocyanate. Molecules 2021, 26, 7512. [Google Scholar] [CrossRef] [PubMed]
  111. Cuellar-Núñez, M.L.; De Mejia, E.G.; Loarca-Piña, G. Moringa oleifera Leaves Alleviated Inflammation through Downregulation of IL-2, IL-6, and TNF-α in a Colitis-Associated Colorectal Cancer Model. Food Res. Int. 2021, 144, 110318. [Google Scholar] [CrossRef] [PubMed]
  112. Kaleo, I.V.; Gao, Q.; Liu, B.; Sun, C.; Zhou, Q.; Zhang, H.; Shan, F.; Xiong, Z.; Bo, L.; Song, C. Effects of Moringa oleifera Leaf Extract on Growth Performance, Physiological and Immune Response, and Related Immune Gene Expression of Macrobrachium Rosenbergii with Vibrio Anguillarum and Ammonia Stress. Fish Shellfish Immunol. 2019, 89, 603–613. [Google Scholar] [CrossRef]
  113. Abidin, Z.; Huang, H.-T.; Liao, Z.-H.; Chen, B.-Y.; Wu, Y.-S.; Lin, Y.-J.; Nan, F.-H. Moringa oleifera Leaves’ Extract Enhances Nonspecific Immune Responses, Resistance against Vibrio Alginolyticus, and Growth in Whiteleg Shrimp (Penaeus vannamei). Animals 2021, 12, 42. [Google Scholar] [CrossRef] [PubMed]
  114. Mohamed, M.A.; El-Mleeh, A.A.; Hamad, R.T.; Abu-Alya, I.S.; El-Hewaity, M.H.; Elbestawy, A.R.; Elbagory, A.M.; Sayed-Ahmed, A.S.; Abd Eldaim, M.A.; Elshabrawy, O.I. Immunostimulant Potential of Moringa oleifera Leaves Alcoholic Extract versus Oregano Essential Oil (OEO) against Cyclophosphamide-Induced Immunosuppression in Broilers Chicks. Trop. Anim. Health Prod. 2023, 55, 209. [Google Scholar] [CrossRef]
  115. Zhang, S.; Cao, Y.; Huang, Y.; Zhang, S.; Wang, G.; Fang, X.; Bao, W.; Aqueous, M. Oleifera Leaf Extract Alleviates DSS-Induced Colitis in Mice through Suppression of Inflammation. J. Ethnopharmacol. 2024, 318, 116929. [Google Scholar] [CrossRef]
  116. Sailaja, B.S.; Aita, R.; Maledatu, S.; Ribnicky, D.; Verzi, M.P.; Raskin, I. Moringa Isothiocyanate-1 Regulates Nrf2 and NF-κB Pathway in Response to LPS-Driven Sepsis and Inflammation. PLoS ONE 2021, 16, e0248691. [Google Scholar] [CrossRef]
  117. Herman-Lara, E.; Rodríguez-Miranda, J.; Ávila-Manrique, S.; Dorado-López, C.; Villalva, M.; Jaime, L.; Santoyo, S.; Martínez-Sánchez, C.E. In Vitro Antioxidant, Anti-Inflammatory Activity and Bioaccessibility of Ethanolic Extracts from Mexican Moringa oleifera Leaf. Foods 2024, 13, 2709. [Google Scholar] [CrossRef]
  118. Luetragoon, T.; Daowtak, K.; Thongsri, Y.; Potup, P.; Calder, P.C.; Usuwanthim, K. Anti-Inflammatory Potential of 3-Hydroxy-β-Ionone from Moringa oleifera: Decreased Transendothelial Migration of Monocytes Through an Inflamed Human Endothelial Cell Monolayer by Inhibiting the IκB-α/NF-κB Signaling Pathway. Molecules 2024, 29, 5873. [Google Scholar] [CrossRef]
  119. Shahbaz, M.; Naeem, H.; Batool, M.; Imran, M.; Hussain, M.; Mujtaba, A.; Alsagaby, S.A.; Al Abdulmonem, W.; El-Ghorab, A.H.; Ghoneim, M.M.; et al. Antioxidant, Anticancer, and Anti-inflammatory Potential of Moringa Seed and Moringa Seed Oil: A Comprehensive Approach. Food Sci. Nutr. 2024, 12, 6157–6173. [Google Scholar] [CrossRef]
  120. Farias-Pereira, R.; Camayoc, P.; Raskin, I. Isothiocyanate-Rich Moringa Seed Extract Activates SKN-1/Nrf2 Pathway in Caenorhabditis Elegans. Int. J. Mol. Sci. 2024, 25, 10917. [Google Scholar] [CrossRef] [PubMed]
  121. Nassar, A.A.M.A.; Gharib, A.A.E.-A.; Abdelgalil, S.Y.; AbdAllah, H.M.; Elmowalid, G.A. Immunomodulatory, Antioxidant, and Growth-Promoting Activities of Dietary Fermented Moringa oleifera in Nile Tilapia (Oreochromus niloticus) with in-Vivo Protection against Aeromonas Hydrophila. BMC Vet. Res. 2024, 20, 231. [Google Scholar] [CrossRef] [PubMed]
  122. Feng, Y.; Huang, J.; Gong, J.; Peng, W.; Yu, C.; Huai, Y.; Chen, C.; Bo, R.; Liu, M.; Li, J. Composition and Anti-Colitis Efficacy Analysis of Ethanol and Aqueous Extraction of Moringa oleifera Leaves. Fitoterapia 2025, 185, 106720. [Google Scholar] [CrossRef]
  123. Luetragoon, T.; Sranujit, R.P.; Noysang, C.; Thongsri, Y.; Potup, P.; Somboonjun, J.; Maichandi, N.; Suphrom, N.; Sangouam, S.; Usuwanthim, K. Evaluation of Anti-Inflammatory Effect of Moringa oleifera Lam. and Cyanthillium Cinereum (Less) H. Rob. Lozenges in Volunteer Smokers. Plants 2021, 10, 1336. [Google Scholar] [CrossRef]
  124. Díaz-Prieto, L.E.; Gómez-Martínez, S.; Vicente-Castro, I.; Heredia, C.; González-Romero, E.A.; Martín-Ridaura, M.D.C.; Ceinos, M.; Picón, M.J.; Marcos, A.; Nova, E. Effects of Moringa oleifera Lam. Supplementation on Inflammatory and Cardiometabolic Markers in Subjects with Prediabetes. Nutrients 2022, 14, 1937. [Google Scholar] [CrossRef]
  125. Prabowo, N.A. Effect of Moringa Leaf Extract on Disease Activity in Rheumatoid Arthritis Patients; Clinicaltrials.gov: Bethesda, MD, USA, 2022.
  126. Asare, B.; Selorm Segbefia, P.; Awuku-Larbi, R.; Asema Asandem, D.; Brenko, T.; Bentum-Ennin, L.; Osei, F.; Teye-Adjei, D.; Agyekum, G.; Akuffo, L. Immunomodulatory Effect of Moringa oleifera and Phyllanthus niruri extracts on Anti-HBV Cytokine Production by Human Peripheral Blood Mononuclear Cells. Open Res. Eur. 2025, 5, 149. [Google Scholar] [CrossRef]
  127. Gambo, A.; Moodley, I.; Babashani, M.; Babalola, T.K.; Gqaleni, N. A Double-Blind, Randomized Controlled Trial to Examine the Effect of Moringa oleifera Leaf Powder Supplementation on the Immune Status and Anthropometric Parameters of Adult HIV Patients on Antiretroviral Therapy in a Resource-Limited Setting. PLoS ONE 2021, 16, e0261935. [Google Scholar] [CrossRef]
  128. Gandhi, G.R.; Mohana, T.; Athesh, K.; Hillary, V.E.; Vasconcelos, A.B.S.; Farias de Franca, M.N.; Montalvão, M.M.; Ceasar, S.A.; Jothi, G.; Sridharan, G.; et al. Anti-Inflammatory Natural Products Modulate Interleukins and Their Related Signaling Markers in Inflammatory Bowel Disease: A Systematic Review. J. Pharm. Anal. 2023, 13, 1408–1428. [Google Scholar] [CrossRef] [PubMed]
  129. Zhang, Y.; Zhang, X.-Y.; Shi, S.-R.; Ma, C.-N.; Lin, Y.-P.; Song, W.-G.; Guo, S.-D. Natural Products in Atherosclerosis Therapy by Targeting PPARs: A Review Focusing on Lipid Metabolism and Inflammation. Front. Cardiovasc. Med. 2024, 11, 1372055. [Google Scholar] [CrossRef]
  130. Mohd Sahardi, N.F.N.; Makpol, S. Suppression of Inflamm-Aging by Moringa oleifera and Zingiber Officinale Roscoe in the Prevention of Degenerative Diseases: A Review of Current Evidence. Molecules 2023, 28, 5867. [Google Scholar] [CrossRef] [PubMed]
  131. Chiș, A.; Noubissi, P.A.; Pop, O.-L.; Mureșan, C.I.; Fokam Tagne, M.A.; Kamgang, R.; Fodor, A.; Sitar-Tăut, A.-V.; Cozma, A.; Orășan, O.H.; et al. Bioactive Compounds in Moringa oleifera: Mechanisms of Action, Focus on Their Anti-Inflammatory Properties. Plants 2023, 13, 20. [Google Scholar] [CrossRef]
  132. Ajzashokouhi, A.H.; Rezaee, R.; Omidkhoda, N.; Karimi, G. Natural Compounds Regulate the PI3K/Akt/GSK3β Pathway in Myocardial Ischemia-Reperfusion Injury. Cell Cycle 2023, 22, 741–757. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, T.; Zhao, L.; Xu, M.; Jiang, P.; Zhang, K. Moringin Alleviates DSS-Induced Ulcerative Colitis in Mice by Regulating Nrf2/NF-κB Pathway and PI3K/AKT/mTOR Pathway. Int. Immunopharmacol. 2024, 134, 112241. [Google Scholar] [CrossRef]
  134. Kim, C.G.; Chang, S.N.; Park, S.M.; Hwang, B.S.; Kang, S.-A.; Kim, K.S.; Park, J.G. Moringa oleifera Mitigates Ethanol-Induced Oxidative Stress, Fatty Degeneration and Hepatic Steatosis by Promoting Nrf2 in Mice. Phytomedicine 2022, 100, 154037. [Google Scholar] [CrossRef]
  135. Afolabi, O.A.; Akhigbe, T.M.; Akhigbe, R.E.; Alabi, B.A.; Gbolagun, O.T.; Taiwo, M.E.; Fakeye, O.O.; Yusuf, E.O. Methanolic Moringa oleifera Leaf Extract Protects against Epithelial Barrier Damage and Enteric Bacterial Translocation in Intestinal I/R: Possible Role of Caspase 3. Front. Pharmacol. 2022, 13, 989023. [Google Scholar] [CrossRef]
  136. Laftah, A.H.; Alhelfi, N.; Al Salait, S.K.; Altemimi, A.B.; Tabandeh, M.R.; Tsakali, E.; Van Impe, J.F.; Abd El-Maksoud, A.A.; Abedelmaksoud, T.G. Mitigation of Doxorubicin-Induced Liver Toxicity in Mice Breast Cancer Model by Green Tea and Moringa oleifera Combination: Targeting Apoptosis, Inflammation, and Oxidative Stress. J. Funct. Foods 2025, 124, 106626. [Google Scholar] [CrossRef]
  137. Reda, R.M.; Helmy, R.M.A.; Osman, A.; Ahmed, F.A.G.; Kotb, G.A.M.; El-Fattah, A.H.A. The Potential Effect of Moringa oleifera Ethanolic Leaf Extract against Oxidative Stress, Immune Response Disruption Induced by Abamectin Exposure in Oreochromis niloticus. Env. Sci. Pollut. Res. 2023, 30, 58569–58587. [Google Scholar] [CrossRef]
  138. Liga, S.; Magyari-Pavel, I.Z.; Avram, Ș.; Minda, D.I.; Vlase, A.-M.; Muntean, D.; Vlase, L.; Moacă, E.-A.; Danciu, C. Comparative Analysis of Moringa oleifera Lam. Leaves Ethanolic Extracts: Effects of Extraction Methods on Phytochemicals, Antioxidant, Antimicrobial, and In Ovo Profile. Plants 2025, 14, 1653. [Google Scholar] [CrossRef] [PubMed]
  139. Lin, J.; Li, F.; Jiao, J.; Qian, Y.; Xu, M.; Wang, F.; Sun, X.; Zhou, T.; Wu, H.; Kong, X. Quercetin, a Natural Flavonoid, Protects against Hepatic Ischemia–Reperfusion Injury via Inhibiting Caspase-8/ASC Dependent Macrophage Pyroptosis. J. Adv. Res. 2024, 70, 555–569. [Google Scholar] [CrossRef]
  140. Yu, J.; Fu, R.; Buhe, A.; Xu, B. Quercetin Attenuates Lipopolysaccharide-Induced Hepatic Inflammation by Modulating Autophagy and Necroptosis. Poult. Sci. 2024, 103, 103719. [Google Scholar] [CrossRef] [PubMed]
  141. Katsaros, I.; Sotiropoulou, M.; Vailas, M.; Kapetanakis, E.I.; Valsami, G.; Tsaroucha, A.; Schizas, D. Quercetin’s Potential in MASLD: Investigating the Role of Autophagy and Key Molecular Pathways in Liver Steatosis and Inflammation. Nutrients 2024, 16, 3789. [Google Scholar] [CrossRef]
  142. Xiong, F.; Zhang, Y.; Li, T.; Tang, Y.; Song, S.-Y.; Zhou, Q.; Wang, Y. A Detailed Overview of Quercetin: Implications for Cell Death and Liver Fibrosis Mechanisms. Front. Pharmacol. 2024, 15, 1389179. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, Y.; Qu, X.; Gao, H.; Zhai, J.; Tao, L.; Sun, J.; Song, Y.; Zhang, J. Quercetin Attenuates NLRP3 Inflammasome Activation and Apoptosis to Protect INH-Induced Liver Injury via Regulating SIRT1 Pathway. Int. Immunopharmacol. 2020, 85, 106634. [Google Scholar] [CrossRef]
  144. Guon, T.E.; Chung, H.S. Moringa oleifera Fruit Induce Apoptosis via Reactive Oxygen Species-Dependent Activation of Mitogen-Activated Protein Kinases in Human Melanoma A2058 Cells. Oncol. Lett. 2017, 14, 1703–1710. [Google Scholar] [CrossRef]
  145. Adewale, G.B.; Olowoparija, S.F.; Olayemi, J.O.; Olayinka, E. Effect of Ethanol Extract of Moringa oleifera Leaves on T-Cell Proliferation. West. J. Med. Biomed. Sci. 2020, 1, 176–184. [Google Scholar]
  146. Akanni, E.O.; Adedeji, A.L.; Akanni, R.A.; Oloke, J.K. Up-Regulation of TNF-α by Ethanol Extract of Moringa oleifera in Benzene-Induced Leukemic Wister Rat: A Possible Mechanism of Anticancer Property. Res. Rev. J. Oncol. Haematol. 2014, 3, 16–21. [Google Scholar]
  147. Abutu, P.; Amuda, O.; Osinaya, O.; Babatunde, B. Regulatory Activity of Ethanol Leaves Extract of Moringa oleifera on Benzene Induced Leukemia in Wister Rat Using TNF-α Analysis. Am. J. Med. Biol. Res. 2019, 7, 6–11. [Google Scholar]
  148. Van Pham, A.T.; Luong, M.H.; Dinh, H.T.T.; Mai, T.P.; Trinh, Q.V.; Luong, L.H. Immunostimulatory Effect of Moringa oleifera Extracts on Cyclophosphamide-Induced Immunosuppressed Mice. J. Herbs Spices Med. Plants 2021, 27, 377–385. [Google Scholar] [CrossRef]
  149. Asaduzzaman, A.K.M.; Hasan, I.; Chakrabortty, A.; Zaman, S.; Islam, S.S.; Ahmed, F.R.S.; Kabir, K.A.; Nurujjaman, M.; Uddin, M.B.; Alam, M.T. Moringa oleifera Seed Lectin Inhibits Ehrlich Ascites Carcinoma Cell Growth by Inducing Apoptosis through the Regulation of Bak and NF-κB Gene Expression. Int. J. Biol. Macromol. 2018, 107, 1936–1944. [Google Scholar] [CrossRef]
  150. Ndlovu, S.S.; Chuturgoon, A.A.; Ghazi, T. Moringa oleifera Lam Leaf Extract Stimulates NRF2 and Attenuates ARV-Induced Toxicity in Human Liver Cells (HepG2). Plants 2023, 12, 1541. [Google Scholar] [CrossRef]
  151. Sailaja, B.S.; Hassan, S.; Cohen, E.; Tmenova, I.; Farias-Pereira, R.; Verzi, M.P.; Raskin, I. Moringa Isothiocyanate-1 Inhibits LPS-Induced Inflammation in Mouse Myoblasts and Skeletal Muscle. PLoS ONE 2022, 17, e0279370. [Google Scholar] [CrossRef]
  152. Villegas-Vazquez, E.Y.; Gómez-Cansino, R.; Marcelino-Pérez, G.; Jiménez-López, D.; Quintas-Granados, L.I. Unveiling the Miracle Tree: Therapeutic Potential of Moringa oleifera in Chronic Disease Management and Beyond. Biomedicines 2025, 13, 634. [Google Scholar] [CrossRef] [PubMed]
  153. Do, B.H.; Hoang, N.S.; Nguyen, T.P.T.; Ho, N.Q.C.; Le, T.L.; Doan, C.C. Phenolic Extraction of Moringa oleifera Leaves Induces Caspase-Dependent and Caspase-Independent Apoptosis through the Generation of Reactive Oxygen Species and the Activation of Intrinsic Mitochondrial Pathway in Human Melanoma Cells. Nutr. Cancer 2021, 73, 869–888. [Google Scholar] [CrossRef]
  154. Sultan, R.; Ahmed, A.; Wei, L.; Saeed, H.; Islam, M.; Ishaq, M. The Anticancer Potential of Chemical Constituents of Moringa oleifera Targeting CDK-2 Inhibition in Estrogen Receptor Positive Breast Cancer Using in-Silico and in Vitro Approches. BMC Complement. Med. Ther. 2023, 23, 396. [Google Scholar] [CrossRef]
  155. Qian, D.; Zha, D.; Sang, Y.; Tao, J.; Cheng, Y. Moringa oleifera Mediated Green Synthesis of Gold Nanoparticles and Their Anti-Cancer Activity against A549 Cell Line of Lung Cancer through ROS/Mitochondrial Damage. Front. Chem. 2025, 13, 1521089. [Google Scholar] [CrossRef]
  156. Kaavyaunni; Antony, S.; Chanthini, K.M.-P. Biogenically Synthesized Moringa oleifera Leaf Extract-Derived Silver Nanoparticles Exhibit Potent Antimicrobial and Anticancer Effects via Oxidative Stress and Apoptotic Pathways in AGS Gastric Cancer Cells. Biocatal. Agric. Biotechnol. 2025, 67, 103674. [Google Scholar] [CrossRef]
  157. Kumar, S.; Verma, P.K.; Shukla, A.; Singh, R.K.; Patel, A.K.; Yadav, L.; Kumar, S.; Kumar, N.; Kaushalendra; Acharya, A. Moringa oleifera L. Leaf Extract Induces Cell Cycle Arrest and Mitochondrial Apoptosis in Dalton’s Lymphoma: An in Vitro and in Vivo Study. J. Ethnopharmacol. 2023, 302, 115849. [Google Scholar] [CrossRef]
  158. Al-Shalabi, R.; abdul Samad, N.; Al-Deeb, I.; Joseph, J.; Abualsoud, B.M. Assessment of Antiangiogenic and Cytotoxic Effects of Moringa oleifera Silver Nanoparticles Using Cell Lines. Curr. Res. Nutr. Food Sci. J. 2024, 12, 196–211. [Google Scholar] [CrossRef]
  159. Choirunnisa, N.L.; Kezia, D.C.; Permana, A.Z.; Malek, N.A.N.N.; Nurkolis, F.; Permatasari, H.K. Modulation of pAKT and NF-κB Pathways by Moringa oleifera Silver Nanoparticles: Inducing Apoptosis in Cervical Cancer Cells. CyTA J. Food 2025, 23, 2543878. [Google Scholar] [CrossRef]
  160. Abd Allah, R.S.; Ahmmed, N.H.; Walaa, A.; Heibashy, D.M. The Curative Role of Moringa Leaf Extract Nanomaterial and Low Doses of Gamma Irradiation on Ehrlich Carcinoma Bearing Mice. Lett. Appl. NanoBioSci. 2025, 14, 74. [Google Scholar]
  161. Raju, N.S.C.; Kei, W.W. Anti-Angiogenic Screening of Moringa oleifera Leaves Extract Using Chorioallantonic Membrane Assay. Iraqi J. Pharm. Sci. 2022, 31, 225–232. [Google Scholar] [CrossRef]
  162. Susanto, H.; Firdaus, S.R.A.; Sholeh, M.; Endharti, A.T.; Taufiq, A.; Malek, N.A.N.N.; Permatasari, H.K. Moringa oleifera Leaf Powder—Silver Nanoparticles (MOLP-AgNPs) Efficiently Inhibit Metastasis and Proliferative Signaling in HT-29 Human Colorectal Cancer Cells. J. Agric. Food Res. 2024, 16, 101149. [Google Scholar] [CrossRef]
  163. Zunica, E.R.; Yang, S.; Coulter, A.; White, C.; Kirwan, J.P.; Gilmore, L.A. Moringa oleifera Seed Extract Concomitantly Supplemented with Chemotherapy Worsens Tumor Progression in Mice with Triple Negative Breast Cancer and Obesity. Nutrients 2021, 13, 2923. [Google Scholar] [CrossRef] [PubMed]
  164. Armagan, G.; Sevgili, E.; Gürkan, F.T.; Köse, F.A.; Bilgiç, T.; Dagcı, T.; Saso, L. Regulation of the Nrf2 Pathway by Glycogen Synthase Kinase-3β in MPP+-Induced Cell Damage. Molecules 2019, 24, 1377. [Google Scholar] [CrossRef]
  165. Bourebaba, L.; Komakula, S.S.B.; Weiss, C.; Adrar, N.; Marycz, K. The PTP1B Selective Inhibitor MSI-1436 Mitigates Tunicamycin-Induced ER Stress in Human Hepatocarcinoma Cell Line through XBP1 Splicing Modulation. PLoS ONE 2023, 18, e0278566. [Google Scholar] [CrossRef]
  166. Xu, Q.; Wu, N.; Li, X.; Guo, C.; Li, C.; Jiang, B.; Wang, H.; Shi, D. Inhibition of PTP1B Blocks Pancreatic Cancer Progression by Targeting the PKM2/AMPK/mTOC1 Pathway. Cell Death Dis. 2019, 10, 874. [Google Scholar] [CrossRef] [PubMed]
  167. Hagoel, L.; Vexler, A.; Kalich-Philosoph, L.; Earon, G.; Ron, I.; Shtabsky, A.; Marmor, S.; Lev-Ari, S. Combined Effect of Moringa oleifera and Ionizing Radiation on Survival and Metastatic Activity of Pancreatic Cancer Cells. Integr. Cancer Ther. 2019, 18, 1534735419828829. [Google Scholar] [CrossRef]
  168. Aja, P.M.; Agu, P.C.; Ezeh, E.M.; Awoke, J.N.; Ogwoni, H.A.; Deusdedit, T.; Ekpono, E.U.; Igwenyi, I.O.; Alum, E.U.; Ugwuja, E.I.; et al. Prospect into Therapeutic Potentials of Moringa oleifera Phytocompounds against Cancer Upsurge: De Novo Synthesis of Test Compounds, Molecular Docking, and ADMET Studies. Bull. Natl. Res. Cent. 2021, 45, 99. [Google Scholar] [CrossRef]
  169. Saha, A.; Desai, B.M.A.; Biswas, P. Multi-Ligand Simultaneous Docking Analysis of Moringa oleifera Phytochemicals Reveals Enhanced BCL-2 Inhibition via Synergistic Action. In Proceedings of the 2024 5th International Conference on Biomedical Engineering (IBIOMED), Bali, Indonesia, 23–25 October 2024; pp. 51–56. [Google Scholar]
  170. Bhadresha, K.; Thakore, V.; Brahmbhatt, J.; Upadhyay, V.; Jain, N.; Rawal, R. Anticancer Effect of Moringa oleifera Leaves Extract against Lung Cancer Cell Line via Induction of Apoptosis. Adv. Cancer Biol. Metastasis 2022, 6, 100072. [Google Scholar] [CrossRef]
  171. ICTRP Search Portal. Available online: https://trialsearch.who.int/Trial2.aspx?TrialID=CTRI/2022/02/040594 (accessed on 26 August 2025).
  172. Tshingani, K.; Donnen, P.; Mukumbi, H.; Duez, P.; Dramaix-Wilmet, M. Impact of Moringa oleifera Lam. Leaf Powder Supplementation versus Nutritional Counseling on the Body Mass Index and Immune Response of HIV Patients on Antiretroviral Therapy: A Single-Blind Randomized Control Trial. BMC Complement. Altern. Med. 2017, 17, 420. [Google Scholar] [CrossRef] [PubMed]
  173. Aprioku, J.S.; Robinson, O.; Obianime, A.W.; Tamuno, I. Moringa Supplementation Improves Immunological Indices and Hematological Abnormalities in Seropositive Patients Receiving HAARTs. Afr. Health Sci. 2022, 22, 1–11. [Google Scholar] [CrossRef]
  174. Barhoi, D.; Upadhaya, P.; Barbhuiya, S.N.; Giri, A.; Giri, S. Aqueous Extract of Moringa oleifera Exhibit Potential Anticancer Activity and Can Be Used as a Possible Cancer Therapeutic Agent: A Study Involving In Vitro and In Vivo Approach. J. Am. Coll. Nutr. 2021, 40, 70–85. [Google Scholar] [CrossRef]
  175. Kim, Y.; Jaja-Chimedza, A.; Merrill, D.; Mendes, O.; Raskin, I. A 14-Day Repeated-Dose Oral Toxicological Evaluation of an Isothiocyanate-Enriched Hydro-Alcoholic Extract from Moringa oleifera Lam. Seeds in Rats. Toxicol. Rep. 2018, 5, 418–426. [Google Scholar] [CrossRef]
  176. Taweerutchana, R.; Lumlerdkij, N.; Vannasaeng, S.; Akarasereenont, P.; Sriwijitkamol, A. Effect of Moringa oleifera Leaf Capsules on Glycemic Control in Therapy-Naïve Type 2 Diabetes Patients: A Randomized Placebo Controlled Study. Evid. Based Complement. Altern. Med. 2017, 2017, 6581390. [Google Scholar] [CrossRef]
  177. Aliyu, A.; Shaari, M.R.; Ahmad Sayuti, N.S.; Reduan, F.H.; Sithambaram, S.; Mohamed Mustapha, N.; Shaari, K.; Hamzah, H.B. Moringa oleifera Hydorethanolic Leaf Extract Induced Acute and Sub-Acute Hepato-Nephrotoxicity in Female ICR-Mice. Sci. Prog. 2021, 104, 00368504211004272. [Google Scholar] [CrossRef]
  178. Manzo, M.L.; Hallarou, M.E.; Halidou, M.D.; Maiga, D.A.; Bahwere, P.; Salimata, W.; Castetbon, K.; Wilmet-Dramaix, M.; Donnen, P. Effect of Moringa Supplementation in the Management of Moderate Malnutrition in Children under 5 Receiving Ready-to-Use Supplementary Foods in Niger: A Randomized Clinical Trial. GSC Adv. Res. Rev. 2021, 8, 071–086. [Google Scholar] [CrossRef]
  179. Kajuju Malla, J.; Ochola, S.; Ogada, I.; Munyaka, A. Effect of Moringa oleifera Fortified Porridge Consumption on Protein and Vitamin A Status of Children with Cerebral Palsy in Nairobi, Kenya: A Randomized Controlled Trial. PLoS Glob. Public Health 2022, 2, e0001206. [Google Scholar] [CrossRef] [PubMed]
  180. Silva, N.R.G.; Costa, W.K.; Ferreira, M.R.A.; Coelho, L.C.B.B.; Soares, L.A.L.; Napoleão, T.H.; Paiva, P.M.G.; de Oliveira, A.M. 13-Week Repeated-Dose Toxicity Study of Optimized Aqueous Extract of Moringa oleifera Leaves in Mice. J. Ethnopharmacol. 2024, 335, 118637. [Google Scholar] [CrossRef] [PubMed]
  181. Adedapo, A.A.; Mogbojuri, O.M.; Emikpe, B.O. Safety Evaluations of the Aqueous Extract of the Leaves of Moringa oleifera in Rats. J. Med. Plants Res. 2009, 3, 586–591. [Google Scholar]
  182. Iliyasu, D.; Rwuaan, J.S.; Sani, D.; Nwannenna, A.I.; Njoku, C.O.; Mustapha, A.R.; Peter, I.D. Evaluation of Safety, Proximate and Efficacy of Graded Dose of Moringa oleifera Aqueous Seed Extract as Supplement That Improve Live-Body Weight and Scrotal Circumference in Yankasa Ram. Int. J. Livest. Res. 2020, 10, 33–46. [Google Scholar] [CrossRef]
  183. Palomino-Pacheco, M.; Rojas-Armas, J.P.; Ortiz-Sánchez, J.M.; Arroyo-Acevedo, J.L.; Justil-Guerrero, H.J.; Martínez-Heredia, J.T. Assessment of Oral Toxicity of Moringa oleifera Lam Aqueous Extract and Its Effect on Gout Induced in a Murine Model. Vet. World 2024, 17, 1449. [Google Scholar] [CrossRef]
  184. Musa, A.H.; Gebru, G.; Debella, A.; Makonnen, E.; Asefa, M.; Woldekidan, S.; Abebe, A.; Lengiso, B.; Bashea, C. Prenatal Developmental Toxicity Study of Herbal Tea of Moringa stenopetala and Mentha spicata Leaves Formulation in Wistar Rats. Toxicol. Rep. 2022, 9, 1853–1862. [Google Scholar] [CrossRef] [PubMed]
  185. Hajar, A.; Abdelmounaim, B.; Hamid, K.; Jaouad, L.; Abdelfattah, A.B.; Majda, B.; Loubna, E.Y.; Mohammed, L.; Rachida, A.; Abderrahman, C. Developmental Toxicity of Moringa oleifera and Its Effect on Postpartum Depression, Maternal Behavior and Lactation. S. Afr. J. Bot. 2024, 171, 257–266. [Google Scholar] [CrossRef]
  186. Soliman, S.S.; Suliman, A.A.; Fathy, K.; Sedik, A.A. Ovario-Protective Effect of Moringa oleifera Leaf Extract against Cyclophosphamide-Induced Oxidative Ovarian Damage and Reproductive Dysfunction in Female Rats. Sci. Rep. 2025, 15, 1054. [Google Scholar]
  187. Abdulahi, S.K.; Dada, E.O.; Adebayo, R.O. Histopathological Effects of Seed Oil of Moringa oleifera Lam. on Albino Mice Infected with Plasmodium Berghei (NK65). Adv. J. Grad. Res. 2022, 11, 71–79. [Google Scholar] [CrossRef]
  188. Oghama, O.E.; Timothy, O.; Gabriel, B.O.; Abaku, S.N. 21 Days Effects of Ethanol Root Extract of Moringa oleifera Lam. on Kidney and Liver Functions in Wistar Rats. Trop. J. Chem. 2025, 1, 14–18. [Google Scholar] [CrossRef]
  189. Musa, A.H.; Hagos, A.D.; Dimsu, G.G.; Eshetu, E.M.; Tola, M.A.; Admas, A.; Gelagle, A.A.; Tullu, B.L. Subchronic Toxicity Study of Herbal Tea of Moringa stenopetala (Baker f.) Cudof. and Mentha spicata L. Leaves Formulation in Wistar Albino Rats. Toxicol. Rep. 2022, 9, 797–805. [Google Scholar] [CrossRef]
  190. Stohs, S.J.; Hartman, M.J. Review of the Safety and Efficacy of Moringa oleifera. Phytother. Res. 2015, 29, 796–804. [Google Scholar] [CrossRef] [PubMed]
  191. Thanikachalam, P.V.; Ramesh, K.; Hydar, M.I.; Dhalapathy, V.V.; Devaraji, M. Therapeutic Potential of Moringa oleifera Lam. in Metabolic Disorders: A Molecular Overview. Asian Pac. J. Trop. Biomed. 2025, 15, 263. [Google Scholar] [CrossRef]
  192. Nakata, H.; Nakayama, S.M.M.; Kataba, A.; Toyomaki, H.; Doya, R.; Beyene Yohannes, Y.; Ikenaka, Y.; Ishizuka, M. Does Moringa oleifera Affect Element Accumulation Patterns and Lead Toxicity in Sprague–Dawley Rats? J. Funct. Foods 2022, 97, 105242. [Google Scholar] [CrossRef]
  193. Popoola, J.O.; Aworunse, O.S.; Oyesola, O.L.; Akinnola, O.O.; Obembe, O.O. A Systematic Review of Pharmacological Activities and Safety of Moringa oleifera. J. Herbmed Pharmacol. 2020, 9, 174–190. [Google Scholar] [CrossRef]
  194. Abdelwanis, F.M.; Abdelaty, H.S.; Saleh, S.A. Exploring the Multifaceted Uses of Moringa oleifera: Nutritional, Industrial and Agricultural Innovations in Egypt. Discov. Food 2024, 4, 146. [Google Scholar] [CrossRef]
  195. Elabd, H.; Soror, E.; El-Asely, A.; El-Gawad, E.A.; Abbass, A. Dietary Supplementation of Moringa Leaf Meal for Nile Tilapia Oreochromis niloticus: Effect on Growth and Stress Indices. Egypt. J. Aquat. Res. 2019, 45, 265–271. [Google Scholar] [CrossRef]
  196. Sallam, G.R.; Abdel-Rahim, M.M.; Lotfy, A.M.; Fayed, W.M.; Shehata, A.I.; El Basuini, M.F.; Elwan, R.I.; Al-absawey, M.A.; Elhetawy, A.I.G. Long Term Dietary Moringa oleifera Leaf Extract to Florida Red Tilapia Oreochromis Sp Improves Performance Immunity Maturation and Reproduction in Saltwater. Sci. Rep. 2025, 15, 20261. [Google Scholar] [CrossRef] [PubMed]
  197. Ofulue, O.O.; Ebomoyi, M.I. Acute Toxicity Test of Alkaloid Fraction of Moringa oleifera Leaf and Its Effect on Reproductive Hormones of Pregnant Wistar Rats. J. Appl. Sci. Environ. Manag. 2023, 27, 2235–2241. [Google Scholar] [CrossRef]
  198. Tilaoui, M.; Ait Mouse, H.; Zyad, A. Update and New Insights on Future Cancer Drug Candidates from Plant-Based Alkaloids. Front. Pharmacol. 2021, 12, 719694. [Google Scholar] [CrossRef]
  199. Ma, J.; Li, M.; Li, N.; Chan, W.Y.; Lin, G. Pyrrolizidine Alkaloid-Induced Hepatotoxicity Associated with the Formation of Reactive Metabolite-Derived Pyrrole–Protein Adducts. Toxins 2021, 13, 723. [Google Scholar] [CrossRef]
  200. Yang, N.; Guo, J.; Zhang, J.; Gao, S.; Xiang, Q.; Wen, J.; Huang, Y.; Rao, C.; Chen, Y. A Toxicological Review of Alkaloids. Drug Chem. Toxicol. 2024, 47, 1267–1281. [Google Scholar] [CrossRef]
  201. Sagrera, A.; Montenegro, T.; Borrego, L. Toxicodermia por Moringa oleifera. Actas Dermosifiliogr. 2021, 112, 953–954. [Google Scholar] [CrossRef]
Figure 1. Major fatty acids identified in Moringa oleifera seeds.
Figure 1. Major fatty acids identified in Moringa oleifera seeds.
Ijms 27 00263 g001
Figure 2. Chemical structures of representative flavonoids from Moringa oleifera leaves.
Figure 2. Chemical structures of representative flavonoids from Moringa oleifera leaves.
Ijms 27 00263 g002
Figure 3. Structures of key phenolic acids present in Moringa oleifera leaves.
Figure 3. Structures of key phenolic acids present in Moringa oleifera leaves.
Ijms 27 00263 g003
Figure 4. Moringa oleifera regulates diverse factors to modulate immune responses. Moringa oleifera modulates immunity through interconnected cellular and molecular pathways. Innate immunity is enhanced via neutrophile chemotaxis, macrophage phagocytic and secretory functions, NK cell-mediated cytotoxicity and the activation of bone marrow-derived mesenchymal stem cells that contribute to immune repair and cytokine balance. Upregulation of GM-CSF and NO further amplifies myeloid and effector cell activity. Adaptive immunity is regulated through CD4+ T-cell differentiation, balancing Treg/Th17 ratios, enhancing Th2 functions via RORγT, leading to increased IL-10 and TGF-β and reduced IL-6, IL-1beta and TNF-α. Key signaling pathways, including NF-κB, MAPK, PI3K/Akt, PDK1, STAT, TSLP, and PD-1 are modulated to maintain immune homeostasis, suppression of chronic inflammation and regulation of programmed cell death. AKT, Protein Kinase B; DC, Dendritic cell; GM-CSF, Granulocyte-Macrophage Colony-Stimulating Factor; IFN, interferon; MAPK, Mitogen-activated protein Kinase; NFκB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NK, Natural killer; NO, Nitric oxide; PD-1, Programmed cell death protein 1; PDK1, 3-Phosphoinositide-dependent protein kinase-1; PI3K, Phosphoinositide 3-Kinase; RORγT, Retinoic acid receptor-related orphan receptor gamma t; STAT, Signal transducer and activator of transcription; TNF, Tumor necrosis factor; TSLP, Thymic stromal lymphopoietin. ⟶ Promote/lead to; ↑ upregulate/increase expression; ↓ downregulate/decrease expression. ⊥ inhibition.
Figure 4. Moringa oleifera regulates diverse factors to modulate immune responses. Moringa oleifera modulates immunity through interconnected cellular and molecular pathways. Innate immunity is enhanced via neutrophile chemotaxis, macrophage phagocytic and secretory functions, NK cell-mediated cytotoxicity and the activation of bone marrow-derived mesenchymal stem cells that contribute to immune repair and cytokine balance. Upregulation of GM-CSF and NO further amplifies myeloid and effector cell activity. Adaptive immunity is regulated through CD4+ T-cell differentiation, balancing Treg/Th17 ratios, enhancing Th2 functions via RORγT, leading to increased IL-10 and TGF-β and reduced IL-6, IL-1beta and TNF-α. Key signaling pathways, including NF-κB, MAPK, PI3K/Akt, PDK1, STAT, TSLP, and PD-1 are modulated to maintain immune homeostasis, suppression of chronic inflammation and regulation of programmed cell death. AKT, Protein Kinase B; DC, Dendritic cell; GM-CSF, Granulocyte-Macrophage Colony-Stimulating Factor; IFN, interferon; MAPK, Mitogen-activated protein Kinase; NFκB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NK, Natural killer; NO, Nitric oxide; PD-1, Programmed cell death protein 1; PDK1, 3-Phosphoinositide-dependent protein kinase-1; PI3K, Phosphoinositide 3-Kinase; RORγT, Retinoic acid receptor-related orphan receptor gamma t; STAT, Signal transducer and activator of transcription; TNF, Tumor necrosis factor; TSLP, Thymic stromal lymphopoietin. ⟶ Promote/lead to; ↑ upregulate/increase expression; ↓ downregulate/decrease expression. ⊥ inhibition.
Ijms 27 00263 g004
Figure 5. Moringa oleifera and its role in the modulation of cancer immunity. Moringa oleifera enhances innate immunity through stimulating neutrophil activation and macrophage functions, driving TNF-γ, H2O2 and NO2 release, and enhancing phagocytosis with IL-2, IFN-γ and GM-CSF release, thereby leading to tumor elimination. In parallel, Moringa oleifera modulates mast cell activity, upregulating TNF-α, IL-1 and IL-2, which contribute to antitumor responses. Furthermore, Moringa oleifera promotes DC maturation, T and B cell activation, contributing to antibody production, IL-12 release, and NK cell-mediated cytotoxicity via perforin and granzyme secretion. In addition, H2O2 suppresses pro-tumorigenic cyclooxygenase activity and modulate PI3K/AKT/mTOR signaling cascades through p-AKT, p-AMPK, and TSC2, and engages p53 and Beclin-1 to trigger, apoptosis, autophagolysosome through inhibition of autophagosome formation, and inhibition of carcinogenesis, with concomitant downregulation of Bcl-2 and Bcl-xL. Antioxidant and redox regulation through H2O2 and NO2 balance reduces oxidative stress, while modulation of NF-κB, STAT, and MAPK pathways limits chronic inflammation and supports cytotoxic T-cell and antibody-mediated responses against cancer cells. Overall, H2O2 orchestrates innate and adaptive immunity, modulates signaling pathways and balances oxidative stress to promote effective tumor elimination. COX, Cyclooxygenase; DC, Dendritic cell; GM-CSF, Granulocyte-Macrophage Colony-Stimulating Factor; IFN, interferon; mTOR, Mechanistic Target of Rapamycin; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NK, Natural killer; P-AMPK, Phosphorylated AMP-Activated protein Kinase; PAKT, Phosphorylated protein kinase B (Akt); PI3K, Phosphoinositide 3-Kinase; STAT, Signal transducer and activator of transcription; TNF, Tumor necrosis factor; TSC2, Tuberous sclerosis complex 2. ⟶ Promote/lead to; ↑ upregulate/increase expression; ↓ downregulate/decrease expression. ⊥ inhibition.
Figure 5. Moringa oleifera and its role in the modulation of cancer immunity. Moringa oleifera enhances innate immunity through stimulating neutrophil activation and macrophage functions, driving TNF-γ, H2O2 and NO2 release, and enhancing phagocytosis with IL-2, IFN-γ and GM-CSF release, thereby leading to tumor elimination. In parallel, Moringa oleifera modulates mast cell activity, upregulating TNF-α, IL-1 and IL-2, which contribute to antitumor responses. Furthermore, Moringa oleifera promotes DC maturation, T and B cell activation, contributing to antibody production, IL-12 release, and NK cell-mediated cytotoxicity via perforin and granzyme secretion. In addition, H2O2 suppresses pro-tumorigenic cyclooxygenase activity and modulate PI3K/AKT/mTOR signaling cascades through p-AKT, p-AMPK, and TSC2, and engages p53 and Beclin-1 to trigger, apoptosis, autophagolysosome through inhibition of autophagosome formation, and inhibition of carcinogenesis, with concomitant downregulation of Bcl-2 and Bcl-xL. Antioxidant and redox regulation through H2O2 and NO2 balance reduces oxidative stress, while modulation of NF-κB, STAT, and MAPK pathways limits chronic inflammation and supports cytotoxic T-cell and antibody-mediated responses against cancer cells. Overall, H2O2 orchestrates innate and adaptive immunity, modulates signaling pathways and balances oxidative stress to promote effective tumor elimination. COX, Cyclooxygenase; DC, Dendritic cell; GM-CSF, Granulocyte-Macrophage Colony-Stimulating Factor; IFN, interferon; mTOR, Mechanistic Target of Rapamycin; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; NK, Natural killer; P-AMPK, Phosphorylated AMP-Activated protein Kinase; PAKT, Phosphorylated protein kinase B (Akt); PI3K, Phosphoinositide 3-Kinase; STAT, Signal transducer and activator of transcription; TNF, Tumor necrosis factor; TSC2, Tuberous sclerosis complex 2. ⟶ Promote/lead to; ↑ upregulate/increase expression; ↓ downregulate/decrease expression. ⊥ inhibition.
Ijms 27 00263 g005
Table 1. Effect of Moringa oleifera on innate immunity.
Table 1. Effect of Moringa oleifera on innate immunity.
Plant Part UsedForm of ApplicationModel/SystemKey Mechanistic InsightsRef
LeavesLeaf polysaccharide fractionLewis lung carcinoma (C57BL/6 mice, oral). In vitro bone marrow-derived macrophagesActivation of macrophage through toll-like receptor reprograms TAMs from M2 to M1 through TLR4, ↑ CXCL9/10, ↑ T-cell infiltration[58,63]
LeavesOral administrationRabbits exposed to heat stressEnhancement of NK cell activity through up-regulation of perforin/granzyme secretion[64,65,66]
LeavesPurified protein fractionMurine BMSCs (In vitro), followed by allogenic MLR and in vivo IgE measurement in miceActivation and maturation of DCs via upregulation of co-stimulatory molecules and the secretion of cytokines and chemokines: ↑ CD80/CD86/MHCII; ↑ IL12 and TNF-α secretion; OX40L-TIM-4-CCL17/22 axis drives Th2 polarization; enhanced T-cell proliferation[60,67,68]
LeavesOral administrationDiabetic-induced damage in male Wistar ratsReduction in pro-inflammatory cytokines (TNF-α, IL6) and oxidative stress[68]
LeavesTopical ethanolic extract (10%)
Moringa tea (oral drinking water)
Wistar rats with experimental burn wounds
Restraint-stressed mice
Modulation of neutrophil migration/chemotaxis and enhancing phagocytosis[59,69]
LeavesOral administration of ethanolic leaf extract at 10, 30 and 100 mg/Kg for 7 daysStaphylococcus aureus-challenged mice (mouse peritoneal macrophage)↑ Macrophage phagocytic activity and capacity; ↑ Neutrophile percentage[70]
LeavesPurified polysaccharide fractionRAW 264.7 macrophages (in vitro)Enhances the gene expression of antimicrobial peptides such as Mop3 through ↑ pinocytosis; ↑ ROS/NO; ↑ IL6; TNF-α and iNOS expression[71]
PodsBoiled extractRAW 264.7 murine macrophage cell lineInhibits pro-inflammatory cytokines (IL6, TNF-α); suppresses iNOS and COX-2 expression; decreases NO production[72]
LeavesEthyl acetate fractionLPS-stimulated RAW 264.7 macrophagesRegulation of genes encoding pro-inflammatory cytokines through the regulation of MAPK and NF-κB signaling cascades[73]
SeedsPurified peptide (sequence KETTTIVR) administered orallyDextran Sulfate Sodium-induced colitis in miceModulates gut microbiota and metabolomic profiles, inhibits JAK-STAT signaling, and protects the intestinal barrier[74]
LeavesEthanolic extract; oral and in vitro exposureSwiss albino mice peritoneal macrophages; RAW 264.7Increase phagocytosis activity through activation of phagocytic receptors and engulfment efficiency[70]
RootsHot water extract,
ethanolic extract
LPS-induced RAW 264.7 macrophages murine macrophage cell lineInhibits NO and TNF-α production; decreases iNOS mRNA production[75]
PodsFreeze-dried pod polyphenol extractRAW 264.7 macrophages (LPS-stimulation)↓ NO, ↓ TNF-α, strong anti-inflammatory effect[76]
LeavesEthyl acetate extract (oral and in vitro)RAW 264.7 macrophagesInhibits NF-κB, suppresses pro-inflammatory cytokines (TNF-α, IL6); ↓ NO production[77]
PodsDigested boiled pod extractCaco-2 cells (intestinal epithelial)Suppressed inflammatory mediators; promoted epithelial anti-inflammatory defense[78]
Leaves (aqueous and ethanolic extracts)Oral administration and in vitro exposureVarious animal models (Wistar rats and BALB/c mice) and the RAW 264.7 macrophage cell lineAnti-inflammatory and antioxidant activities via free radical scavenging and inhibition of pro-inflammatory cytokines such as IL-1β, IL-6 and TNF-α[68,77,79,80]
BMSCs, Bone marrow-derived mesenchymal stem cells; CCL, C-C motif chemokine ligand; CXCL, C-X-C motif chemokine ligand; DCs, Dendritic cells; iNOS, Inducible nitric oxide synthase; JAK, Janus kinase; LPS, Lipopolysaccharide; MAPK, Mitogen-activated protein Kinase; NF-κB, Nuclear factor kappa-light-chain-enhancer of activated B cells; OX40, OX40 ligand; STAT, Signal transducer and activator of transcription; TIM, T cell immunoglobulin and mucin domain containing protein 4. ↑ upregulate/increase expression; ↓ downregulate/decrease expression.
Table 2. Effect of Moringa oleifera on adaptive immunity.
Table 2. Effect of Moringa oleifera on adaptive immunity.
Plant Part UsedForm of ApplicationModel/SystemKey Mechanistic InsightsRef
LeavesPurified leaf proteinIn vitro, Human T-lymphoblastic leukemia (Jurkat cells)Modulation of T cell activation and proliferation through the regulation of T cell receptor signaling cascades[60,81,82]
LeavesAqueous extractIn vitro, murine splenocyte cultureB-cell activation[73]
LeavesMethanolic extractSRBC immunized Wistar ratsAntibody production via B cell receptor stimulation[83,84,85]
LeavesEthanolic extractBreast cancer cells (MD-MA-231)↓ of NF-κB and p65; Transcription factor modulation; ↓ cancer cell viability[86]
LeavesTopical applicationAtopic dermatitis mouse modelPromote T-helper cell differentiation through modulation of Th1, Th2, Th17 and Treg cells responses[61,67,87,88,89]
Leaves
Leaf proteins
Oral administration
Protein extract
Rabbits exposed to heat stress
Murine BMDCs
Induction of Foxp3 expression and promotion of Treg cells[60,64]
PodsPod meal (dietary inclusion)Broiler chickensEnhanced growth performance; improved cell-mediated immunity[90]
LeavesLeaf protein fraction or extractBALB/c mice (intraperitoneal sensitization + oral exposure, allergy model); BMDCs (in vitro) and IgE induction after DCs transfer (in vivo)Regulation of antigen presentation and processing[60,88,91]
LeavesMethanolic extractWistar albino ratsStimulates neutrophil and lymphocyte function; Enhances humoral immunity; increases antigen-specific antibody secretion and WBC levels[85]
SeedsMicroRNA-enriched extractHuman PBMCs (HIV+)Modulates T-cell differentiation and memory T-cell subsets; reduces HIV replication[92]
LeavesProtein extract (oral and in vitro)BALB/c mice; Murine BMDCsInduces IgE production and Th2 polarization; activates DC; modulates humoral immunity by enhancing the production of antigen-specific antibodies[60,93,94,95]
BMDCs, Bone marrow-derived dendritic cells; DCs, Dendritic cells; Foxp3, Forkhead box P3 gene; PBMCs, Peripheral blood mononuclear cells; SRBC, Sheep red blood cells; WBC, White blood cells. ↓ downregulate/decrease expression.
Table 3. Long-term safety and toxicity profile of Moringa oleifera: insights from preclinical and clinical studies.
Table 3. Long-term safety and toxicity profile of Moringa oleifera: insights from preclinical and clinical studies.
Plant PartStudy ModelDose/DurationKey Findings/Safety OutcomesRef
Seeds (hydroalcoholic extract)Rats (14-day toxicity study)100–2000 mg/kg/dayNo significant toxicological effects; normal hematological and biochemical parameters[175]
Leaf capsules in T2DM patientsHumans: adults with T2DM (therapy naïve)Nutritional dose, 4 weeksWell tolerated; no hypoglycemia; renal (BUN, creatinine) and hepatic (AST, ALT) markers remained normal[176]
Leaves (hydroethanolic extract)Female ICR miceAcute: 2000 mg/kg, single dose.
Sub-acute: 125–1000 mg/kg daily for 28 days.
Acute: LD50 > 2000 mg/kg; signs of liver and kidney damage (increased AST, CK and creatinine; hepatic degeneration; renal necrosis).
Sub-acute: moderate hepato-nephrotoxicity (hepatic and renal necrosis, sinusoidal dilatation; glomerulonephritis).
Lower doses (125–500 mg/kg): relatively safe with minimal adverse effects.
[177]
Leaf powderHumans: adults400 mg capsules, 6x daily (2.4 g/day); 12 weeksWell tolerated; no adverse effects; kidney parameters (creatinine, urea), liver enzymes (AST, ALT), and hematological markers remained normal; decreased inflammatory parameters (CRP) and improved lipid profile (LDL-C, total cholesterol)[124]
Leaf powder (added to RUSF)Humans: children < 5 years with moderate malnutritionDaily supplementation: 5 weeks for the Moringa group and 4 weeks for the placebo group.No renal/hepatic toxicity reported; well tolerated[178]
Leaf powder (10% fortified porridge)Humans (observational study): children with cerebral palsyDaily intake (3 months)No serious adverse reactions or increased morbidity; improved nutritional (vitamin A and protein status) and immune markers[179]
ALT, Alanine aminotransferase; AST, Aspartate aminotransferase; BUN, Blood urea nitrogen; CK, Creatine Kinase; CRP, C-reactive protein; ICR mice, Institute of Cancer Research (mouse strain); LDL-C, Low-density lipoprotein cholesterol; RUSF, Ready-to-use supplementary food; T2DM, Type 2 Diabetes Mellitus.
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

Tilaoui, M.; El Karroumi, J.; Ait Mouse, H.; Zyad, A. Harnessing Moringa oleifera for Immune Modulation in Cancer: Molecular Mechanisms and Therapeutic Potential. Int. J. Mol. Sci. 2026, 27, 263. https://doi.org/10.3390/ijms27010263

AMA Style

Tilaoui M, El Karroumi J, Ait Mouse H, Zyad A. Harnessing Moringa oleifera for Immune Modulation in Cancer: Molecular Mechanisms and Therapeutic Potential. International Journal of Molecular Sciences. 2026; 27(1):263. https://doi.org/10.3390/ijms27010263

Chicago/Turabian Style

Tilaoui, Mounir, Jamal El Karroumi, Hassan Ait Mouse, and Abdelmajid Zyad. 2026. "Harnessing Moringa oleifera for Immune Modulation in Cancer: Molecular Mechanisms and Therapeutic Potential" International Journal of Molecular Sciences 27, no. 1: 263. https://doi.org/10.3390/ijms27010263

APA Style

Tilaoui, M., El Karroumi, J., Ait Mouse, H., & Zyad, A. (2026). Harnessing Moringa oleifera for Immune Modulation in Cancer: Molecular Mechanisms and Therapeutic Potential. International Journal of Molecular Sciences, 27(1), 263. https://doi.org/10.3390/ijms27010263

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop